WO2019023553A1 - Compounds and methods for the targeted degradation of androgen receptor - Google Patents

Compounds and methods for the targeted degradation of androgen receptor Download PDF

Info

Publication number
WO2019023553A1
WO2019023553A1 PCT/US2018/044051 US2018044051W WO2019023553A1 WO 2019023553 A1 WO2019023553 A1 WO 2019023553A1 US 2018044051 W US2018044051 W US 2018044051W WO 2019023553 A1 WO2019023553 A1 WO 2019023553A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
alkyl
halo
independently
branched
Prior art date
Application number
PCT/US2018/044051
Other languages
French (fr)
Inventor
Andrew P. Crew
Michael Berlin
Xin Chen
Craig M. Crews
Hanqing Dong
Yimin Qian
Lawrence Snyder
Jing Wang
Kurt Zimmermann
Original Assignee
Arvinas, 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
Priority claimed from US15/663,273 external-priority patent/US20170327469A1/en
Application filed by Arvinas, Inc. filed Critical Arvinas, Inc.
Priority to CA3069544A priority Critical patent/CA3069544A1/en
Priority to BR112020001825-2A priority patent/BR112020001825A2/en
Priority to JP2020504388A priority patent/JP2020528918A/en
Priority to CN201880062879.9A priority patent/CN111212835A/en
Priority to RU2020108515A priority patent/RU2020108515A/en
Priority to KR1020207005895A priority patent/KR20200035435A/en
Priority to EP18756023.0A priority patent/EP3658548A1/en
Priority to AU2018306606A priority patent/AU2018306606B2/en
Publication of WO2019023553A1 publication Critical patent/WO2019023553A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • A61K31/422Oxazoles not condensed and containing further heterocyclic rings
    • 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
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/555Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound pre-targeting systems involving an organic compound, other than a peptide, protein or antibody, for targeting specific cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C255/00Carboxylic acid nitriles
    • C07C255/49Carboxylic acid nitriles having cyano groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton
    • C07C255/54Carboxylic acid nitriles having cyano groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton containing cyano groups and etherified hydroxy groups bound to the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/08Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms
    • C07C271/24Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atom of at least one of the carbamate groups bound to a carbon atom of a ring other than a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/10Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no 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
    • C07D207/16Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • C07D213/82Amides; Imides in position 3
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/02Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings
    • C07D237/06Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D237/10Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-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
    • C07D237/24Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • 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
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D411/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen and sulfur atoms as the only ring hetero atoms
    • C07D411/02Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen and sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D411/12Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen and sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D411/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen and sulfur atoms as the only ring hetero atoms
    • C07D411/14Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen and sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • 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
    • 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
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06034Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/04Systems containing only non-condensed rings with a four-membered ring

Definitions

  • the present description relates to bifunctional compounds, which are useful for the modifying the ubiquitination and subsequent degradation of target polypeptides and proteins, in particular, androgen receptor.
  • the compounds comprise a Von Hippel-Lindau (VHL) binding moiety, which binds to the VHL E3 ubiquitin ligase, a target protein binding moiety, which binds to the target protein (e.g., androgen receptor), and optionally a linker moiety which links the VHL binding moiety and target protein binding moiety.
  • VHL Von Hippel-Lindau
  • Androgen Receptor belongs to a nuclear hormone receptor family that is activated by androgens, such as testosterone and
  • Hsp90 Heat Shock Protein 90
  • NLS Nuclear Localization Signal
  • AR deficiency leads to Androgen Insensitivity Syndrome, formerly termed testicular feminization.
  • a commonly measured target gene of AR activity is the secreted Prostate Specific Antigen (PSA) protein.
  • PSA Prostate Specific Antigen
  • the current treatment regimen for prostate cancer involves inhibiting the androgen-AR axis by two methods. The first approach relies on reduction of androgens, while the second strategy aims to inhibit AR function (Nat. Rev. Drug Discovery, 2013, 12,823–824).
  • An alternative approach for the treatment of prostate cancer involves eliminating the AR protein. Because AR is a critical driver of tumorigenesis in many forms of prostate cancers, its elimination should lead to therapeutically remedial response.
  • the present disclosure describes compounds, including compositions comprising the same, which function to recruit endogenous proteins to an E3 ubiquitin ligase, e.g., Von Hippel-Lindau (VHL) E3 ubiquitin ligase, for ubiquitination and subsequent degradation, and methods of using the same.
  • E3 ubiquitin ligase e.g., Von Hippel-Lindau (VHL) E3 ubiquitin ligase
  • VHL Von Hippel-Lindau
  • AR androgen receptor
  • the description provides methods of using an effective amount of the compounds as described herein for the treatment or amelioration of a disease condition including cancer, e.g., prostate cancer, and Kennedy’s Disease.
  • the disclosure provides compounds which function to recruit endogenous proteins, e.g., AR proteins, to E3 Ubiquitin Ligase for ubiquintination and degradation.
  • the compounds have the following general structure:
  • ABM is an AR binding moiety
  • ULM is an E3 ligase binding moiety, e.g., a VHL E3 ligase binding moiety (VLM)
  • L is a bond or a linker moiety which links the ABM and ULM.
  • VLM is a VHL E3 ligase binding moiety and L is a bond or a linker moiety which links the ABM and VLM.
  • the VLM comprises a hydroxyl prolyl moiety.
  • the ULM is a moiety specific for an E3 ubiquitin ligase such as, e.g., cereblon, mouse double minute 2 homolog (Mdm2), or inhibitor of apoptosis (IAP), wherein the ULM moiety is coupled to an ABM as described herein.
  • E3 ubiquitin ligase such as, e.g., cereblon, mouse double minute 2 homolog (Mdm2), or inhibitor of apoptosis (IAP), wherein the ULM moiety is coupled to an ABM as described herein.
  • the description provides AR binding moieties (ABM).
  • ABSM AR binding moieties
  • the description provides compounds having the following general structure: ABM-L, wherein ABM is an AR binding moiety as described herein, and L is a chemical linker moiety, or optionally a bond.
  • ABM and/or L are coupled to a ULM as described herein.
  • the ABM is selected from following structures:
  • W 1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CN, C ⁇ CH, CF 3 , C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl), C 1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C 2-6 alkenyl, C 2-6 alkynyl;
  • Y 1 , Y 2 are each independently NR Y1 , O, S;
  • Q is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, optionally substituted with 0-6 R Q , each R Q is independently H, OH, C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl), or 2 R Q groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
  • R 1 , R 2 , R a , R b , R Y1 , R Y2 are each independently H, OH, C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl), or R 1 , R 2 together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms;
  • W 2 is a bond, C 1-6 alkyl, alicyclic (e.g., C1-6 alicyclic), heterocyclic, aryl, heteroaryl, bicyclic, biaryl, biheteroaryl, biheterocyclic, each optionally substituted by 1, 2 or 3 R W2 ; and each R W2 is independently H, halo, C 1-6 alkyl (optionally substituted by 1 or more F), OC 1-3 alkyl (optionally substituted by 1 or more F), OH, NH 2 , NR Y1 R Y2 , CN.
  • the ABM can comprise or consist of a structure as set forth herein, in particular in any of the ABMs as provided in
  • the ULM (derivatized or configured to be linked or coupled to an ABM via a linker (as indicated by the dashed line)) has the structure,
  • W 3 is optionally substituted aryl, optionally substituted heteroaryl, or
  • each R 9 and R 10 is independently hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted hydroxyalkyl, optionally substituted heteroaryl, or haloalkyl; or R 9 , R 10 , and the carbon atom to which they are attached form an optionally substituted cycloalkyl;
  • R 11 is optionally substituted heterocyclic, optionally substituted alkoxy, optionally
  • R 12 is H or optionally substituted alkyl
  • R 13 is H, optionally substituted alkyl, optionally substituted alkylcarbonyl, optionally substituted (cycloalkyl)alkylcarbonyl, optionally substituted aralkylcarbonyl, optionally substituted arylcarbonyl, optionally substituted (heterocyclyl)carbonyl, or optionally substituted aralkyl;
  • R 14a, R 14b is each independently H, haloalkyl, or optionally substituted alkyl;
  • W 5 is a phenyl or a 5-10 membered heteroaryl
  • R 15 is H, halogen, CN, OH, NO 2 , N R 14a R 14b , OR 14a , CONR 14a R 14b , NR 14a COR 14b , SO 2 NR 14a R 14b , NR 14a SO 2 R 14b , optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted haloalkoxy, optionally substituted aryl, optionally substituted heteroaryl,
  • R 17 is H, halo, optionally substituted C 3-6 cycloalkyl, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 alkenyl, or C 1-6 haloalkyl;
  • Xa is S or O;
  • each R 16 is independently halo, optionally substituted alkyl, optionally substituted haloalkyl, hydroxy, or optionally substituted haloalkoxy;
  • o 0, 1, 2, 3, or 4;
  • each R 18 is independently halo, optionally substituted alkoxy, cyano, optionally substituted alkyl, haloalkyl, haloalkoxy or a linker;
  • p 0, 1, 2, 3, or 4.
  • the ULM has the structure
  • R 9 is H
  • R 10 is isopropyl, tert-butyl, sec-butyl, cyclopentyl, or cyclohexyl;
  • R 12 is H
  • R 13 is H, optionally substituted alkyl, optionally substituted alkylcarbonyl, optionally
  • substituted (cycloalkyl)alkylcarbonyl optionally substituted aralkylcarbonyl, optionally substituted arylcarbonyl, optionally substituted (heterocyclyl)carbonyl, or optionally substituted arylalkyl;
  • R 14a is H, haloalkyl, methyl, ethyl, isopropyl, cyclopropyl, or C 1 -C 6 alkyl (linear, branched, optionally substituted), each optionally substituted with 1 or more halo, hydroxyl, nitro, CN, C 1 -C 6 alkyl (linear, branched, optionally substituted), or C 1 -C 6 alkoxyl (linear, branched, optionally substituted); and R 15 is wherein R 17 is H, halo, optionally substituted C 3-6 cycloalkyl, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 alkenyl, and C 1-6 haloalkyl; and Xa is S or O.
  • an androgen receptor binding moiety has a structure of
  • W 1 is aryl or heteroaryl, independently substituted by 1 or more halo, CF 3 , hydroxyl, nitro, CN, C ⁇ CH, C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl), C 1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C 2-6 alkenyl, C 2-6 alkynyl;
  • Y 1 , Y 2 are each independently NR Y1 , O, S;
  • R Q is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, optionally substituted with 0-6 R Q , each R Q is independently H, OH, C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl), or 2 R Q groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); R Y1 , R Y2 are each independently H, OH, C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl);
  • W 2 is a bond, C 1-6 alkyl, alicyclic, heterocyclic, aryl, heteroaryl, bicyclic, biaryl,
  • each R W2 is independently H, halo, C 1-6 alkyl (optionally substituted by 1 or more F), OC 1- 3 alkyl (optionally substituted by 1 or more F), OH, NH 2 , NR Y1 R Y2 , CN.
  • the compounds comprise a plurality of E3 ligase binding moieties and/or a plurality of ABMs.
  • the linker group (L) comprises a chemical structural unit represented by the formula:
  • q is an integer greater than 1;
  • R L1 or R L2 each, independently, can be linked to another A group to form cycloalkyl and/or heterocyclyl moeity that can be further substituted with 0-4 R L5 groups.
  • the description provides a bifunctional compound having a structure selected from the group consisting of Examples 1-870, a salt, a polymorph, and a prodrug thereof.
  • compositions comprising compounds as described herein, and a pharmaceutically acceptable carrier.
  • the compositions are therapeutic or pharmaceutical compositions comprising an effective amount of a compound as described herein and a pharmaceutally acceptable carrier.
  • the therapeutic or pharmaceutical compositions comprise an additional biologically active agent, e.g., an agent effective for the treatment of cancer.
  • the therapeutic compositions comprising compounds described herein can be in any suitable dosage form, e.g., solid, or liquid, and configured to be delivered by any suitable route, e.g., oral, parenteral, intravenous, intraperitoneal, subcutaneous, intramuscular, etc., and in any desired unit dosage form.
  • the therapeutic composition as described herein is configured to be administered or consumed by a subject one or more times over a descired time period, e.g., day, week, month, etc.
  • the disclosure provides methods of modulating protein ubiquitination and degradation in a subject, e.g., a cell, a tissue, mammal, or human patient, the method comprising administering an effective amount of a compound as described herein or a composition comprising an effective amount of the same to a subject, wherein the compound or composition comprising the same is effective in modulating protein ubquitination and degration of the protiein in the subject.
  • the protein is androgen receptor (AR).
  • the disclosure provides methods of modulating AR protein ubiquitination and degradation in a subject, e.g., a cell, a tissue, mammal, or human patient, the method comprising administering an effective amount of a compound as described herein or a composition comprising an effective amount of the same to a subject, wherein the compound or composition comprising the same is effective in modulating AR protein ubquitination and degration of the protiein in the subject.
  • the disclosure provides methods of treating or ameliorating a symptom of a disease related to AR activity in a subject, e.g., a cell, a tissue, mammal, or human patient, the method comprising administering an effective amount of a compound as described herein or a composition comprising an effective amount of the same to a subject in need thereof, wherein the compound or composition comprising the same is effective in treating or ameliorating a symptom of a disease related to AR activity in the subject.
  • a subject e.g., a cell, a tissue, mammal, or human patient
  • the method comprising administering an effective amount of a compound as described herein or a composition comprising an effective amount of the same to a subject in need thereof, wherein the compound or composition comprising the same is effective in treating or ameliorating a symptom of a disease related to AR activity in the subject.
  • the disease to be treated is cancer, e.g., prostate cancer, or Kennedy’s Disease.
  • the subject is a human.
  • the disclosure provides methods for identifying the effects of the degradation of proteins of interest in a biological system using compounds according to the present invention.
  • kits comprising compounds or compositions as described herein.
  • the kit may be promoted, distributed, or sold as a unit for performing the methods of the present invention.
  • the kits of the present invention may preferably contain instructions which describe a suitable use.
  • Such kits can be conveniently used, e.g., in clinical settings, to treat patients exhibiting symptoms of, e.g., cancer or Kennedy’s Disease.
  • FIG. 1A Exemplary PROTACs comprise an androgen receptor targeting moiety (ABM; darkly shaded rectangle), a Von Hippel-Lindau (VHL) E3 ubiquitin ligase binding moiety (VLM; lightly shaded triangle), and a linker moiety (L; black line) coupling or tethering the ABM to the VLM (as described herein, L can be absent or a bond or a chemical linker moiety).
  • FIG 1B Illustrates the functional use of the PROTACs as described herein.
  • the VLM recognizes and binds to Von Hippel-Lindau (VHL) E3 ubiquitin ligase
  • VHL Von Hippel-Lindau
  • ABM binds and recruits androgen receptor and brings it into close proximity to the Von Hippel-Lindau (VHL) E3 ubiquitin ligase.
  • the E3 ubiquitin ligase is complexed with an E2 ubiquitin- conjugating protein, and either alone or via the E2 protein catalyzes attachment of ubiquitin (dark circles) to a lysine on the target protein via an isopeptide bond.
  • the poly-ubiquitinated protein far right is then targeted for degration by the proteosomal machinery of the cell.
  • FIG. 1 Apoptosis in VCaP cells.
  • VCaP cells were cultured in Charcoal Stripped Serum containing media supplemented with 0.1 nM R1881 for 48 hrs. The degree of apoptosis was ascertained with CaspaseGlo assay (Promega). These results demonstrated that PROTACs are much more potent in inducing apoptosis than an AR antagonist enzalutamide. Further, the degree of AR degradation correlates with their ability to induce apoptosis in VCaP cells.
  • FIG. 4 PSA suppression in LNCaP F876L.
  • LNCaP cells transduced with AR F876L construct were cultured in Charcoal Stripped Serum containing media supplemented with 0.1 nM R1881 for 7 days.
  • FIG. 5 Prostate involution in C57B6 mouse model. 12-week old male C57BL/6 mice were treated with AR PROTAC Example 163 and its inactive epimer analog Compound A which is unable to bind to VHL E3 ligase. Enzalutamide (PO, QD, 30 mpk), Example 163 (IP, QD, 1 and 3 mpk) and Compound A (IP, QD, 1 and 3 mpk) were administered for 10 days, upon which the prostates were isolated and weighed. These results demonstrated that the ability of PROTAC Example 163 to degrade AR leads to significant prostate involution in mice at very low doses.
  • FIG. 6 Tumor growth inhibition in VCaP xenograft model.
  • VCaP cells were implanted into CB17 scid mice subcutaneously. Once the tumors were palpable, the mice were castrated, leading to temporary tumor stasis. Upon regrowth of tumors, the mice were dosed with enzalutamide (PO, QD, 30 mpk) or AR PROTAC Example 163 (IP, QD, at 30, 10 and 3 mpk) as indicated. Tumor growth inhibition was observed in all treatment arms.
  • enzalutamide PO, QD, 30 mpk
  • AR PROTAC Example 163 IP, QD, at 30, 10 and 3 mpk
  • FIG. 7A and Figure 7B AR degradation of PROTAC is E3 ligase dependent.
  • Figure 7A AR PROTAC Example 1 was added to LNCaP cells at indicated concentrations for 24 hours in the presence or absence of 10 uM VHL E3 ligase ligand compound B.
  • Figure 7B LNCaP cells were treated with AR PROTAC Example 1 and its inactive epimer analog compound C which is unable to bind to VHL E3 ligase.” DETAILED DESCRIPTION
  • the present description relates to the surprising and unexpected discovery that an E3 ubiquitin ligase protein can ubiquitinate a target protein, in particular androgen receptor, once the E3 ubiquitin ligase protein and the target protein are brought into proximity by a chimeric construct (e.g., PROTAC) as described herein, in which a moiety that binds the E3 ubiquitin ligase protein is coupled, e.g., covalently, to a moiety that bind the androgen receptortarget protein.
  • a chimeric construct e.g., PROTAC
  • the present description provides compounds, compositions comprising the same, and associated methods of use for ubiquitination and degradation of a chosen target protein, e.g., androgen receptor (See Figure 1A and Figure 1B).
  • a reference to "A and/or B", when used in conjunction with open-ended language such as “comprising” can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.
  • the phrase "at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from anyone or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements.
  • This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase "at least one" refers, whether related or unrelated to those elements specifically identified.
  • At least one of A and B can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.
  • co-administration and “co-administering” or“combination therapy” can refer to both concurrent administration (administration of two or more therapeutic agents at the same time) and time varied administration (administration of one or more therapeutic agents at a time different from that of the administration of an additional therapeutic agent or agents), as long as the therapeutic agents are present in the patient to some extent, preferably at effective amounts, at the same time.
  • one or more of the present compounds described herein are coadministered in combination with at least one additional bioactive agent, especially including an anticancer agent.
  • the co-administration of compounds results in synergistic activity and/or therapy, including anticancer activity.
  • the term“effective” can mean, but is in no way limited to, that amount/dose of the active pharmaceutical ingredient, which, when used in the context of its intended use, effectuates or is sufficient to prevent, inhibit the occurrence, ameliorate, delay or treat (alleviate a symptom to some extent, preferably all) the symptoms of a condition, disorder or disease state in a subject in need of such treatment or receiving such treatment.
  • effective subsumes all other effective amount or effective concentration terms, e.g.,“effective amount/dose,” “pharmaceutically effective amount/dose” or“therapeutically effective amount/dose,” which are otherwise described or used in the present application.
  • the effective amount depends on the type and severity of disease, the
  • composition used used, the route of administration, the type of mammal being treated, the physical characteristics of the specific mammal under consideration, concurrent medication, and other factors which those skilled in the medical arts will recognize.
  • the exact amount can be ascertainable by one skilled in the art using known techniques (see, e.g., Lieberman,
  • therapeutic formulation or“pharmaceutically acceptable formulation” can mean, but is in no way limited to, a composition or formulation that allows for the effective distribution of an agent provided by the invention, which is in a form suitable for administration to the physical location most suitable for their desired activity, e.g., systemic administration.
  • pharmaceutically acceptable or“pharmacologically acceptable” can mean, but is in no way limited to, entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal, or a human, as appropriate.
  • compositions can mean, but is in no way limited to, any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Suitable carriers are described in the most recent edition of Remington's Pharmaceutical Sciences, a standard reference text in the field, which is incorporated herein by reference. Preferred examples of such carriers or diluents include, but are not limited to, water, saline, finger's solutions, dextrose solution, and 5% human serum albumin. Liposomes and non-aqueous vehicles such as fixed oils may also be used. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.
  • systemic administration refers to a route of administration that is, e.g., enteral or parenteral, and results in the systemic distribution of an agent leading to systemic absorption or accumulation of drugs in the blood stream followed by distribution throughout the entire body.
  • Suitable forms depend upon the use or the route of entry, for example oral, transdermal, or by injection. Such forms should not prevent the composition or formulation from reaching a target cell (i.e., a cell to which the negatively charged polymer is desired to be delivered to).
  • pharmacological compositions injected into the blood stream should be soluble.
  • Other factors are known in the art, and include considerations such as toxicity and forms which prevent the composition or formulation from exerting its effect.
  • Administration routes which lead to systemic absorption include, without limitations: intravenous, subcutaneous, intraperitoneal, inhalation, oral, intrapulmonary and intramuscular.
  • the rate of entry of a drug into the circulation has been shown to be a function of molecular weight or size.
  • the use of a liposome or other drug carrier comprising the compounds of the instant invention can potentially localize the drug, for example, in certain tissue types, such as the tissues of the reticular endothelial system (RES).
  • RES reticular endothelial system
  • a liposome formulation which can facilitate the association of drug with the surface of cells, such as, lymphocytes and macrophages is also useful.
  • the term“local administration” refers to a route of administration in which the agent is delivered to a site that is apposite or proximal, e.g., within about 10 cm, to the site of the lesion or disease.
  • the term compound generally refers to a single compound, but also may include other compounds such as stereoisomers, regioisomers and/or optical isomers (including racemic mixtures) as well as specific enantiomers or enantiomerically enriched mixtures of disclosed compounds.
  • the term also refers, in context to prodrug forms of compounds which have been modified to facilitate the administration and delivery of compounds to a site of activity. It is noted that in describing the present compounds, numerous substituents and variables associated with same, among others, are described.
  • both a double bond and single bond are represented within the context of the compound shown.
  • derivatives can mean compositions formed from the native compounds either directly, by modification, or by partial substitution.
  • analogs can mean compositions that have a structure similar to, but not identical to, the native compound.
  • Ubiquitin Ligase refers to a family of proteins that facilitate the transfer of ubiquitin to a specific substrate protein, targeting the substrate protein for
  • Von Hippel-Lindau E3 Ubiquitin Ligase or VCB E3 Ubiquitin Ligase is protein that alone or in combination with an E2 ubiquitin-conjugating enzyme causes the attachment of ubiquitin to a lysine on a target protein, and subsequently targets the specific protein substrates for degradation by the proteasome.
  • E3 ubiquitin ligase alone or in complex with an E2 ubiquitin conjugating enzyme is responsible for the transfer of ubiquitin to targeted proteins.
  • the ubiquitin ligase is involved in polyubiquitination such that a second ubiquitin is attached to the first; a third is attached to the second, and so forth.
  • Mono-ubiquitinated proteins are not targeted to the proteasome for degradation, but may instead be altered in their cellular location or function, for example, via binding other proteins that have domains capable of binding ubiquitin.
  • different lysines on ubiquitin can be targeted by an E3 to make chains. The most common lysine is Lys48 on the ubiquitin chain. This is the lysine used to make polyubiquitin, which is recognized by the proteasome.
  • the term“subject” is used throughout the specification to describe a cell, tissue, or animal, preferably a mammal, e.g., a human or a domesticated animal, to whom treatment, including prophylactic treatment, with the compositions according to the present invention is provided.
  • a mammal e.g., a human or a domesticated animal
  • the term patient refers to that specific animal, including a domesticated animal such as a dog or cat or a farm animal such as a horse, cow, sheep, etc.
  • the term patient refers to a human patient unless otherwise stated or implied from the context of the use of the term.
  • the present invention provides compounds useful for regulating protein activity.
  • the composition comprises a ubiquitin pathway protein binding moiety
  • ubiquitin pathway protein binding moiety recognizes an ubiquitin pathway protein and the targeting moiety recognizes a target protein (e.g., androgen receptor).
  • target protein e.g., androgen receptor
  • the description provides AR binding moieties (ABM).
  • ABSM AR binding moieties
  • the compounds having the following general structure: ABM-L, wherein ABM is an AR binding moiety as described herein, and L is a chemical linker moiety, e.g., a linker as described herein, or optionally a bond.
  • the ABM and/or L are coupled to a ULM as described herein below.
  • the disclosure provides compounds which function to recruit androgen receptor (AR) proteins to E3 Ubiquitin Ligase for ubiquintination and degradation.
  • AR androgen receptor
  • the compounds have the following general structure:
  • ULM is an E3 ligase binding moiety
  • ABM is an AR binding moiety, which binds to an AR protein
  • L is a bond or a chemical linker moiety which links the ABM and ULM.
  • the ULM is a moiety specific for an E3 ubiquitin ligase such as, e.g., Von Hippel-Lindau E3 ubiquitin ligase (VHL), cereblon, mouse double minute 2 homolog (Mdm2), or inhibitor of apoptosis (IAP), wherein the ULM moiety is coupled to an ABM as described herein.
  • VHL Von Hippel-Lindau E3 ubiquitin ligase
  • Mdm2 mouse double minute 2 homolog
  • IAP inhibitor of apoptosis
  • the AR is ubiquitinated by the ubiquitin ligase and degraded.
  • the ABM is chemically linked or coupled directly to the ULM group.
  • the ABM is chemically linked or coupled to the ULM via a chemical linker moiety.
  • the description provides compounds having the following general structure:
  • ABM is an AR binding moiety and VLM is a Von Hippel-Lindau E3 Ubiquitin Ligase (VHL) binding moiety
  • L is a bond or a chemical linker moiety which links the ABM and VLM.
  • the ULM or VLM group and ABM group may be covalently linked to the linker group through any covalent bond which is appropriate and stable to the chemistry of the linker.
  • the ULM or VLM comprises a hydroxyprolyl moiety. The hydroxyl prolyl moiety has been shown to be importantn for binding and recruiting of the VHL protein.
  • the general structures are exemplary and the respective moieties can be arranged in any desired order or configuration, e.g., ULM-L-ABM, and VLM-L- ABM respectively.
  • the compounds comprise a plurality of E3 ligase binding moieties and/or a plurality of ABMs.
  • ABM alone, without forming ABM-L-ULM, provides desired properties in regulating protein activity.
  • the ULM comprises a chemical structure selected from the group ULM-a:
  • a dashed line indicates the attachment of at least one ABM, another ULM or VLM (i.e., ULM’ or VLM’), or a chemical linker moiety coupling at least one ABM, a ULM’ or VLM’ to the other end of the linker;
  • R Y3 , R Y4 are each independently H, linear or branched C 1-6 alkyl, optionally substituted by 1 or more halo, optionally substituted C P
  • W 3 is an optionally substituted–T-N(R 1a R 1b ),–T-Aryl, an optionally substituted–T- Heteroaryl, an optionally substituted–T-Heterocycle, an optionally substituted -NR 1 -T- Aryl, an optionally substituted -NR 1 -T-Heteroaryl or an optionally substituted -NR 1 -T- Heterocycle, where T is covalently bonded to X 1;
  • each R 1 , R 1a , R 1b is independently H, a C 1 -C 6 alkyl group (linear, branched, optionally
  • W 4 is an optionally substituted -NR 1 -T-Aryl, an optionally substituted -NR 1 -T-Heteroaryl group or an optionally substituted -NR 1 -T-Heterocycle, wherein -NR 1 is covalently bonded to X 2
  • R 1 is H or CH 3 , preferably H
  • T is an optionally substituted–(CH 2 ) n - group, wherein each one of the methylene groups may be optionally substituted with one or two substituents, preferably selected from halogen, a C 1 -C 6 alkyl group (linear, branched, optionally substituted by 1 or more halogen, -OH) or the sidechain of an amino acid as otherwise described herein, preferably methyl, which may be optionally substituted; and n is 0 to 6, often 0, 1, 2, or 3, preferably 0.
  • T may also be a–(CH 2 O) n - group, a–(OCH 2 ) n - group, a–
  • T is an optionally substituted–(CH 2 ) n - group, wherein each one of the methylene groups may be optionally substituted with one or two substituents, preferably selected from halogen, an amino acid sidechain as otherwise described herein or a C 1 -C 6 alkyl group (linear, branched, optionally substituted by 1 or more halo, -OH), preferably one or two methyl groups, which may be optionally substituted; and n is 0 to 6, often 0, 1, 2 or 3, preferably 0 or 1.
  • T may also be a–(CH 2 O) n - group, a–(OCH 2 ) n - group, a–
  • W 3 and/or W 4 can be attached to a linker moiety as described herein.
  • aryl groups for W 3 include optionally substituted phenyl or naphthyl groups, preferably phenyl groups, wherein the phenyl or naphthyl group is optionally substituted with a linker group to which is attached a ABM group (including a ULM’ group) and/or a halogen (preferably F or Cl), an amine, monoalkyl- or dialkyl amine (preferably, dimethylamine), an amido group (preferably a–(CH 2 ) m -NR 1 C(O)R 2 group wherein m, R 1 and R 2 are the same as for R 1 ), a halogen (often F or Cl), OH, CH 3 , CF 3 , OMe, OCF 3 , NO 2 , CN or a S(O) 2 R S group (R S is a a C 1 -C 6 alkyl group, an optionally substituted aryl, heteroaryl or heterocycle group or a
  • said substituent phenyl group is an optionally substituted phenyl group (i.e., the substituent phenyl group itself is preferably substituted with at least one of F, Cl, OH, SH, COOH, CH 3 , CF 3 , OMe, OCF 3 , NO 2 , CN or a linker group to which is attached a ABM group (including a ULM’ group), wherein the substitution occurs in ortho-, meta- and/or para- positions of the phenyl ring, preferably para-), a naphthyl group, which may be optionally substituted including as described above, an optionally substituted heteroaryl (preferably an optionally substituted isoxazole including a methylsubstituted isoxazole, an optionally substituted oxazole including a methylsubstituted oxazole, an optionally substituted thiazole including a methyl substituted thiazole, an optionally substituted pyrrole including a
  • methylsubstituted pyrrole an optionally substituted imidazole including a methylimidazole, a benzylimidazole or methoxybenzylimidazole, an oximidazole or methyloximidazole, an optionally substituted diazole group, including a methyldiazole group, an optionally substituted triazole group, including a methylsubstituted triazole group, a pyridine group, including a halo- (preferably, F) or methylsubstitutedpyridine group or an oxapyridine group (wherein the pyridine group is linked to the phenyl group by an oxygen) or an optionally substituted heterocycle (tetrahydrofuran, tetrahydrothiophene, pyrrolidine, piperidine, morpholine, piperazine, tetrahydroquinoline, oxane or thiane.
  • aryl, heteroaryl or heterocyclic groups may be
  • heteroaryl groups for W 3 include an optionally substituted quinoline (which may be attached to the pharmacophore or substituted on any carbon atom within the quinoline ring), an optionally substituted indole (including dihydroindole), an optionally substituted indolizine, an optionally substituted azaindolizine (2, 3 or 4-azaindolizine) an optionally substituted benzimidazole, benzodiazole, benzoxofuran, an optionally substituted imidazole, an optionally substituted isoxazole, an optionally substituted oxazole (preferably methyl substituted), an optionally substituted diazole, an optionally substituted triazole, a tetrazole, an optionally substituted benzofuran, an optionally substituted thiophene, an optionally substituted thiazole (preferably methyl and/or thiol substituted), an optionally substituted isothiazole, an optionally
  • S c is CHR SS , NR URE , or O;
  • R HET is H, CN, NO 2 , halo (preferably Cl or F), optionally substituted C 1 -C 6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups (e.g. CF 3 ), optionally substituted O(C 1 -C 6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted acetylenic group–C ⁇ C-R a wherein R a is H or a C 1 -C 6 alkyl group (preferably C 1 -C 3 alkyl);
  • R SS is H, CN, NO 2 , halo (preferably F or Cl), optionally substituted C 1 -C 6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups), optionally substituted O-(C 1 -C 6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted -C(O)(C 1 -C 6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups);
  • R URE is H, a C 1 -C 6 alkyl (preferably H or C 1 -C 3 alkyl) or a–C(O)(C 1 -C 6 alkyl), each of
  • groups is optionally substituted with one or two hydroxyl groups or up to three halogen, preferably fluorine groups, or an optionally substituted heterocycle, for example piperidine, morpholine, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, piperidine, piperazine, each of which is optionally substituted; and
  • Y C is N or C-R YC , wherein R YC is H, OH, CN, NO 2 , halo (preferably Cl or F), optionally substituted C 1 -C 6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups (e.g. CF 3 ), optionally substituted O(C 1 -C 6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted acetylenic group–C ⁇ C-R a wherein R a is H or a C 1 -C 6 alkyl group (preferably C 1 -C 3 alkyl).
  • Each of said heteroaryl groups may be optionally substituted with a linker group to which is attached a ABM group (including a ULM’ group).
  • heterocycle groups for W 3 include
  • R PRO is H, optionally substituted C 1 -C 6 alkyl or an optionally substituted aryl (phenyl or napthyl), heteroaryl or heterocyclic group selected from the group consisting of oxazole, isoxazole, thiazole, isothiazole, imidazole, diazole, oximidazole, pyrrole, pyrollidine, furan, dihydrofuran, tetrahydrofuran, thiene, dihydrothiene, tetrahydrothiene, pyridine, piperidine, piperazine, morpholine, quinoline, (each preferably substituted with a C 1 -C 3 alkyl group, preferably methyl or a halo group, preferably F or Cl), benzofuran, indole, indolizine, azaindolizine; R PRO1 and R PRO2 are each independently H, an optionally subsituted C 1 -C 3 alkyl group or together
  • each n is 0, 1, 2, 3, 4, 5, or 6 (preferably 0 or 1), wherein each of said Heteocycle groups may be optionally substituted with a linker group to which is attached a ABM group
  • W 3 substituents for use in the present invention also include specifically (and without limitation to the specific compound disclosed) the W 3 substituents which are found in the identified compounds disclosed herein (which includes the specific compounds which are disclosed in the present specification, and the figures which are attached hereto). Each of these W 3 substituents may be used in conjunction with any number of W 4 substituents, which are also disclosed herein.
  • Aryl groups for W 4 include optionally substituted phenyl or naphthyl groups, preferably phenyl groups, wherein the phenyl group is optionally substituted with a linker group to which is attached an ABM group (including a ULM’ group), a halogen (preferably F or Cl), an amine, monoalkyl- or dialkyl amine (preferably, dimethylamine), F, Cl, OH, COOH, C 1 -C 6 alkyl, preferably CH 3 , CF 3 , OMe, OCF 3 , NO 2 , or CN group (each of which may be substituted in ortho-, meta- and/or para- positions of the phenyl ring, preferably para-), an optionally substituted phenyl group (the phenyl group itself is preferably substituted with a linker group attached to a ABM group, including a ULM’ group), and/or at least one of F, Cl, OH, COOH, CH
  • S c is CHR SS , NR URE , or O;
  • R HET is H, CN, NO 2 , halo (preferably Cl or F), optionally substituted C 1 -C 6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups (e.g. CF 3 ), optionally substituted O(C 1 -C 6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted acetylenic group–C ⁇ C-R a wherein R a is H or a C 1 -C 6 alkyl group (preferably C 1 -C 3 alkyl);
  • R SS is H, CN, NO 2 , halo (preferably F or Cl), optionally substituted C 1 -C 6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups), optionally substituted O-(C 1 -C 6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted -C(O)(C 1 -C 6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups);
  • R URE is H, a C 1 -C 6 alkyl (preferably H or C 1 -C 3 alkyl) or a–C(O)(C 1 -C 6 alkyl) each of which groups is optionally substituted with one or two hydroxyl groups or up to three halogen, preferably fluorine groups, or an optionally substituted phenyl group, an optionally substituted heteroaryl, or an optionally substituted heterocycle, preferably for example piperidine, morpholine, pyrrolidine, tetrahydrofuran);
  • R PRO is H, optionally substituted C 1 -C 6 alkyl or an optionally substituted aryl (phenyl or napthyl), heteroaryl or heterocyclic group selected from the group consisting of oxazole, isoxazole, thiazole, isothiazole, imidazole, diazole, oximidazole, pyrrole, pyrollidine, furan, dihydrofuran, tetrahydrofuran, thiene, dihydrothiene, tetrahydrothiene, pyridine, piperidine, piperazine, morpholine, quinoline, (each preferably substituted with a C 1 -C 3 alkyl group, preferably methyl or a halo group, preferably F or Cl), benzofuran, indole, indolizine, azaindolizine;
  • R PRO1 and R PRO2 are each independently H, an optionally subsituted C 1 -C 3 alkyl group or together form a keto group;
  • each n is independently 0, 1, 2, 3, 4, 5, or 6 (preferably 0 or 1), or an optionally substituted heterocycle, preferably tetrahydrofuran, tetrahydrothiene, piperidine, piperazine or morpholine (each of which groups when substituted, are preferably substituted with a methyl or halo (F, Br, Cl), each of which groups may be optionally substituted with a linker group to which is attached a ABM group (including a ULM’ group).
  • heteroaryl groups for W 4 include an optionally substituted quinoline (which may be attached to the pharmacophore or substituted on any carbon atom within the quinoline ring), an optionally substituted indole, an optionally substituted indolizine, an optionally substituted azaindolizine, an optionally substituted benzofuran, including an optionally substituted benzofuran, an optionally substituted isoxazole, an optionally substituted thiazole, an optionally substituted isothiazole, an optionally substituted thiophene, an optionally substituted pyridine (2-, 3, or 4-pyridine), an optionally substituted imidazole, an optionally substituted pyrrole, an optionally substituted diazole, an optionally substituted triazole, a tetrazole, an optionally substituted oximidazole, or a group according to the chemical structure:
  • S c is CHR SS , NR URE , or O;
  • R HET is H, CN, NO 2 , halo (preferably Cl or F), optionally substituted C 1 -C 6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups (e.g. CF 3 ), optionally substituted O(C 1 -C 6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted acetylenic group–C ⁇ C-R a wherein R a is H or a C 1 -C 6 alkyl group (preferably C 1 -C 3 alkyl);
  • R SS is H, CN, NO 2 , halo (preferably F or Cl), optionally substituted C 1 -C 6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups), optionally substituted O-(C 1 -C 6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted -C(O)(C 1 -C 6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups);
  • R URE is H, a C 1 -C 6 alkyl (preferably H or C 1 -C 3 alkyl) or a–C(O)(C 1 -C 6 alkyl), each of which groups is optionally substituted with one or two hydroxyl groups or up to three halogen, preferably fluorine groups, or an optionally substituted heterocycle, for example piperidine, morpholine, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, piperidine, piperazine, each of which is optionally substituted, and
  • Y C is N or C-R YC , wherein R YC is H, OH, CN, NO 2 , halo (preferably Cl or F), optionally substituted C 1 -C 6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups (e.g.
  • R a is H or a C 1 -C 6 alkyl group (preferably C 1 -C 3 alkyl), each of which groups may be optionally substituted with a linker group to which is attached a ABM group (including a ULM’ group).
  • heterocycle groups for W 4 include tetrahydrofuran, tetrahydrothiene, tetrahydroquinoline, piperidine, piperazine, pyrrollidine, morpholine, oxane or thiane, each of which groups may be optionally substituted, or a group according to the chemical structure:
  • R PRO is H, optionally substituted C 1 -C 6 alkyl or an optionally substituted aryl, heteroaryl or heterocyclic group;
  • R PRO1 and R PRO2 are each independently H, an optionally subsituted C 1 -C 3 alkyl group or together form a keto group and
  • each n is independently 0, 1, 2, 3, 4, 5, or 6 (often 0 or 1), each of which groups may be
  • W 4 substituents for use in the present invention also include specifically (and without limitation to the specific compound disclosed) the W 4 substituents which are found in the identified compounds disclosed herein (which includes the specific compounds which are disclosed in the present specification, and the figures which are attached hereto).Each of these W 4 substituents may be used in conjunction with any number of W 3 substituents which are also disclosed herein.
  • ULM-a is optionally substituted by 1-3 R P groups in the pyrrolidine moiety.
  • R P is independently H, halo, -OH, C 1-3 alkyl.
  • the W 3 , W 4 can independently be covalently coupled to a linker which is attached one or more ABM groups.
  • ULM is a group (derivatized or configured to be linked or coupled to an ABM via a linker (as indicated by the dashed line) according to the chemical structure:
  • W 3 is optionally substituted aryl, optionally substituted heteroaryl, or
  • each R 9 and R 10 is independently hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted hydroxyalkyl, optionally substituted heteroaryl, or haloalkyl; or R 9 , R 10 , and the carbon atom to which they are attached form an optionally substituted cycloalkyl; [0100] R 11 is optionally substituted heterocyclic, optionally substituted alkoxy, optionally
  • R 12 is H or optionally substituted alkyl
  • R 13 is H, optionally substituted alkyl, optionally substituted alkylcarbonyl, optionally substituted (cycloalkyl)alkylcarbonyl, optionally substituted aralkylcarbonyl, optionally substituted arylcarbonyl, optionally substituted (heterocyclyl)carbonyl, or optionally substituted aralkyl;
  • R 14a, R 14b is each independently H, haloalkyl, or optionally substituted alkyl;
  • W 5 is a phenyl or a 5-10 membered heteroaryl
  • R 15 is H, halogen, CN, OH, NO 2 , NR 14a R 14b , OR 14a , CONR 14a R 14b , NR 14a COR 14b , SO 2 NR 14a R 14b , NR 14a SO 2 R 14b , optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted haloalkoxy; optionally substituted aryl; optionally substituted heteroaryl; optionally substituted cycloalkyl; optionally substituted cycloheteroalkyl;
  • each R 16 is independently halo, optionally substituted alkyl, haloalkyl, hydroxy, optionally substituted alkoxy, or haloalkoxy;
  • o 0, 1, 2, 3, or 4;
  • each R 18 is independently halo, optionally substituted alkoxy, cyano, optionally substituted alkyl, haloalkyl, haloalkoxy or a linker;
  • p is 0, 1, 2, 3, or 4.
  • R 15 wherein R 17 is H, halo, optionally substituted C 3-6 cycloalkyl, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 alkenyl, and C 1-6 haloalkyl;
  • Xa is S or O.
  • R 17 is selected from the group methyl, ethyl, isopropyl, and cyclopropyl.
  • R 15 is selected from the rou consistin of:
  • R 11 is selected from the group consisting of:
  • the ULM (derivatized or configured to be linked or coupled to an ABM via a linker (as indicated by the dashed line)) has the structure:
  • R 9 is H
  • R 10 is isopropyl, tert-butyl, sec-butyl, cyclopentyl, or cyclohexyl;
  • R 11 is ;
  • R 12 is H
  • R 13 is H, optionally substituted alkyl, optionally substituted alkylcarbonyl, optionally substituted (cycloalkyl)alkylcarbonyl, optionally substituted aralkylcarbonyl, optionally substituted arylcarbonyl, optionally substituted (heterocyclyl)carbonyl, or optionally substituted aralkyl;
  • R14a is H, haloalkyl, methyl, ethyl, isopropyl, cyclopropyl, or C 1 -C 6 alkyl (linear, branched, optionally substituted), each optionally substituted with 1 or more halo, hydroxyl, nitro, CN, C 1 -C 6 alkyl (linear, branched, optionally substituted), or C 1 -C 6 alkoxyl (linear, branched, optionally substituted);and
  • R15 is wherein R17 is H, halo, optionally substituted C3-6cycloalkyl, optionally substituted C1-6alkyl, optionally substituted C1-6alkenyl, or C1-6haloalkyl; and Xa is S or O.
  • the ULM or VLM is selected from the group consisting of:
  • the ULM is selected from the group consisting of:
  • the compounds as described herein include one or more ABM chemically linked or coupled to one or more ULMs or VLMs via a chemical linker (L).
  • the linker group L is a group comprises one or more covalently connected structural units of A (e.g. -A 1... A q - ), wherein A 1 is coupled to an ABM moiety, and q is an integer greater than or equal to 0. In certain embodiments, q is an integer greater than or equal to 1.
  • a q is a group which is connected to a ULM or VLM moiety, and A 1 and A q are connected via structural units of A (number of such structural units of A: q-2).
  • a q is a group which is connected to A 1 and to a ULM or VLM moiety.
  • the structure of the linker group L is –A 1 –, and A 1 is a group which is connected to a ULM or VLM moiety and an ABM moiety.
  • q is an integer from 1 to 100, 1 to 90, 1 to 80, 1 to 70, 1 to 60, 1 to 50, 1 to 40, 1 to 30, 1 to 20, or 1 to 10.
  • R L1 , R L2 , R L3 , R L4 and R L5 are, each independently, H, halo, C 1-8 alkyl, OC 1-8 alkyl, SC 1-8 alkyl, NHC 1-8 alkyl, N(C 1-8 alkyl) 2 , C 3-11 cycloalkyl, aryl, heteroaryl, C 3- 11 heterocyclyl, OC 1-8 cycloalkyl, SC 1-8 cycloalkyl, NHC 1-8 cycloalkyl, N(C 1-8 cycloalkyl) 2 , N(C 1- 8 cycloalkyl)(C 1-8 alkyl), OH, NH 2 , SH, SO 2 C 1-8 alkyl, P(O)(OC 1-8 alkyl)(C 1-8 alkyl)(C 1-8 alkyl)(C 1-8 alkyl), OH, NH 2 , SH, SO 2 C 1-8 alkyl, P(O)(OC 1-8 alkyl)
  • the linker (L) is selected from the group consisting of:
  • the linker (L) comprises a structure selected from, but not limited to the structure shown below, wherein a dashed line indicates the attachment point to the ABM or ULM moieties: wherein:
  • W L1 and W L2 are each independently a 4-8 membered ring with 0-4 heteroatoms, optionally substituted with R Q , each R Q is independently a H, halo, OH, CN, CF 3 , NH 2 , carboxyl, C 1 -C 6 alkyl (linear, branched, optionally substituted), C 1 -C 6 alkoxy (linear, branched, optionally substituted), or 2 R Q groups taken together with the atom they are attached to, form a 4-8 membered ring system containing 0-4 heteroatoms;
  • Y L1 is each independently a bond, C 1 -C 6 alkyl (linear, branched, optionally substituted) and optionally one or more C atoms are replaced with O; or C 1 -C 6 alkoxy (linear, branched, optionally substituted); and
  • n 0-10.
  • the linker (L) comprises a structure selected from, but not limited to the structure shown below, wherein a dashed line indicates the attachment point to the ABM or ULM moieties:
  • W L1 and W L2 are each independently aryl, heteroaryl, cyclic, heterocyclic, C 1-6 alkyl (linear, branched, optionally substituted), C 1 -C 6 alkoxy (linear, branched, optionally substituted), bicyclic, biaryl, biheteroaryl,or biheterocyclic, each optionally substituted with R Q , each R Q is independently a H, halo, OH, CN, CF 3 , NH 2 , carboxyl, hydroxyl, nitro, C ⁇ CH, C 2-6 alkenyl, C 2-6 alkynyl, C 1 -C 6 alkyl (linear, branched, optionally substituted), C 1 -C 6 alkoxy (linear, branched, optionally substituted), OC 1-3 alkyl (optionally substituted by 1 or more F), OH, NH 2 , NR Y1 R Y2 , CN, or 2 R Q groups taken together with the
  • Q L is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, biheterocyclic, or bicyclic, optionally bridged, optionally substituted with 0-6 R Q , each R Q is independently H, C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl), or 2 R Q groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
  • R Y1 , R Y2 are each independently H, OH, C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl), or R 1 , R 2 together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms; and
  • n 0-10.
  • the linker group is optionally substituted
  • (poly)ethyleneglycol having between 1 and about 100 ethylene glycol units, between about 1 and about 50 ethylene glycol units, between 1 and about 25 ethylene glycol units, between about 1 and 10 ethylene glycol units, between 1 and about 8 ethylene glycol units and 1 and 6 ethylene glycol units, between 2 and 4 ethylene glycol units,or optionally substituted alkyl groups interdispersed with optionally substituted, O, N, S, P or Si atoms.
  • the linker is substituted with an aryl, phenyl, benzyl, alkyl, alkylene, or heterocycle group.
  • the linker may be asymmetric or symmetrical.
  • the description provides a PROTAC compound in which the linker is cleavable in vivo into a functional E3 ligase binding moiety, and target protein binding moiety.
  • the linker is configured or“tuned” to have the desired kinetics of cleavage into functional component molecules or active metabolites.
  • the enzyme responsible for cleavage of the linker is a liver enzyme, such as, e.g., oxidases, peroxidase, reductases, transferases, dehydrogenases, peroxidases.
  • the enzyme is at least one of cytochrome P450 oxidase, e.g., CYP3A4, Flavin- containing monooxygenase, alcohol dehydrogenase, aldehyde dehydrogenase, monoamine oxidase, peroxidase, glutathione S-transferase, cytochrome P450 reductase, sulfotransferase, methyltransferase, N-acetyltransferase, glucuronosyltransferase, transpeptidase, or combination thereof.
  • ABSMs Androgen Binding Moieties
  • the description provides AR binding moieties (ABM), which in certain aspects and embodiments are coupled to a linker and/or a ULM as described herein.
  • ABSM AR binding moieties
  • the ABM comprises a chemical moiety that binds to the androgen receptor (AR).
  • AR androgen receptor
  • various androgen receptor binding compounds have been described in literature, including various androgen derivatives such as testosterone, dihydrotestosterone, and metribolone (also known as methyltrienolone or R1881), and non- steroidal compounds such as bicalutamide, enzalutamide.
  • Those of ordinary skill in the art would appreciate that these androgen receptor binding compounds could be potentially used as an ABM moiety in a PROTAC compound.
  • Such literature includes, but not limited to, G. F. Allan et. al, Nuclear Receptor Signaling, 2003, 1, e009; R. H.
  • the ABM comprises a structure selected from, but not limited to the structures shown below, wherein a dashed line indicates the attachment point of linker moiety:
  • W 1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CF 3 , CN, C ⁇ CH, C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl), C 1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C 2-6 alkenyl, C 2-6 alkynyl;
  • Y 1 , Y 2 are each independently NR Y1 , O, S;
  • R Q is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, optionally substituted with 0-6 R Q , each R Q is independently H, OH, C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl), or 2 R Q groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); R 1 , R 2 , R a , R b , R Y1 , R Y2 are each independently H, OH, C 1-6 alkyl (linear, branched,
  • R 1 , R 2 together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms;
  • W 2 is a bond, C 1-6 alkyl, alicyclic, heterocyclic, aryl, heteroaryl, bicyclic, biaryl, biheteroaryl, or biheterocyclic, each optionally substituted by 1, 2 or 3 R W2 ;
  • each R W2 is independently H, halo, OH, NH 2 , C 1-6 alkyl (optionally substituted by 1 or more F), OC 1-3 alkyl (optionally substituted by 1 or more F), NR Y1 R Y2 , or CN.
  • the ABM comprises a structure shown below, wherein a dashed line indicates the point of attachment to a linker moiety:
  • W 1 is aryl or heteroaryl, each optionally substituted by 1 or more halo, hydroxyl, nitro, CN, CF 3 , C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl), or C 1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo);
  • each R Q is independently H, OH, C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl), or 2 R Q groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
  • R Y1 , R Y2 are each independently H, OH, or C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl);
  • W 2 is a bond, C 1-6 alkyl, aryl, heteroaryl, alicyclic, heterocyclic, bicyclic, biaryl, biheteroaryl, biheterocyclic, each optionally substituted by 1, 2 or 3 R W2 ;
  • each R W2 is independently H, OH, NH 2 , CR Y1 R Y2 , halo, C 1-6 alkyl (optionally substituted by 1 or more F), OC 1-3 alkyl (optionally substituted by 1 or more F).
  • the W 2 is covalently coupled to one or more ULM or VLM groups, or a linker to which is attached one or more ULM or VLM groups as described herein.
  • W 1 is
  • each R 22 is independently halo, optionally substituted alkyl, haloalkyl, cyano, or nitro;
  • each R 23 is independently H, halo, optionally substituted alkyl, haloalkyl, cyano, or nitro.
  • W 1 is selected from the group consisting of:
  • ABM is selected from the group consisting of:
  • the ABM comprises the structure:
  • W 1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CF 3 , CN, C ⁇ CH, C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl), C 1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C 2-6 alkenyl, C 2-6 alkynyl;
  • Q is a 4 membered alicyclic ring with 0-2 heteroatoms, optionally substituted with 0-6 R Q , each R Q is independently H, C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl), or 2 R Q groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
  • R Y1 , R Y2 are each independently H, OH, C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl);
  • W 2 is a bond, C 1-6 alkyl, alicyclic (e.g., C 1-6 alicyclic), heterocyclic, aryl, heteroaryl, bicyclic, biheterocyclic, biaryl, or biheteroaryl each optionally substituted by 1, 2 or 3 R W2 ; and each R W2 is independently H, halo, C 1-6 alkyl (optionally substituted by 1 or more F), OC 1- 3 alkyl (optionally substituted by 1 or more F), OH, NH 2 , NR Y1 R Y2 , CN.
  • the description provides an androgen receptor
  • bindingcompound comprising a structure of:
  • W 1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CN, CF 3 , C ⁇ CH, C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl), C 1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C 2-6 alkenyl, C 2-6 alkynyl;
  • Y 1 , Y 2 are each independently NR Y1 , O, S;
  • R Q is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, optionally substituted with 0-6 R Q , each R Q is independently H, OH, C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl), or 2 R Q groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); R Y1 , R Y2 are each independently H, OH, C 1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C 1-6 alkoxyl);
  • W 2 is a bond, C 1-6 alkyl, alicyclic, heterocyclic, aryl, heteroaryl, bicyclic, biheterocyclic, biaryl, biheteroaryl, each optionally substituted by 1, 2 or 3 R W2 ; and each R W2 is independently H, halo, C 1-6 alkyl (optionally substituted by 1 or more F), OC 1- 3 alkyl (optionally substituted by 1 or more F), OH, NH 2 , NR Y1 R Y2 , CN.
  • the androgen receptor binding compound of ABM-e is selected from the group consisting of:
  • hydrocarbyl shall mean a compound which contains carbon and hydrogen and which may be fully saturated, partially unsaturated or aromatic and includes aryl groups, alkyl groups, alkenyl groups and alkynyl groups.
  • alkyl shall mean within its context a linear, branch-chained or cyclic fully saturated hydrocarbon radical or alkyl group, preferably a C 1 -C 10 , more preferably a C 1 -C 6 , alternatively a C 1 -C 3 alkyl group, which may be optionally substituted.
  • alkyl groups are methyl, ethyl, n-butyl, sec-butyl, n-hexyl, n-heptyl, n-octyl, n-nonyl, n-decyl, isopropyl, 2-methylpropyl, cyclopropyl, cyclopropylmethyl, cyclobutyl, cyclopentyl, cyclopen- tylethyl, cyclohexylethyl and cyclohexyl, among others.
  • compounds according to the present invention which may be used to covalently bind to dehalogenase enzymes.
  • These compounds generally contain a side chain (often linked through a polyethylene glycol group) which terminates in an alkyl group which has a halogen substituent (often chlorine or bromine) on its distil end which results in covalent binding of the compound containing such a moiety to the protein.
  • a side chain often linked through a polyethylene glycol group
  • a halogen substituent often chlorine or bromine
  • Alkynyl refers to linear, branch-chained or cyclic C 2 -C 10 (preferably C 2 -C 6 ) hydrocarbon radicals containing at least one C ⁇ C bond.
  • alkylene when used, refers to a–(CH 2 ) n - group (n is an integer generally from 0-6), which may be optionally substituted.
  • the alkylene group preferably is substituted on one or more of the methylene groups with a C 1 -C 6 alkyl group (including a cyclopropyl group or a t-butyl group), more preferably a methyl group, but may also be substituted with one or more halo groups, preferably from 1 to 3 halo groups or one or two hydroxyl groups, O-(C 1 -C 6 alkyl) groups or amino acid sidechains as otherwise disclosed herein.
  • an alkylene group may be substituted with a urethane or alkoxy group (or other group) which is further substituted with a polyethylene glycol chain (of from 1 to 10, preferably 1 to 6, often 1 to 4 ethylene glycol units) to which is substituted (preferably, but not exclusively on the distal end of the polyethylene glycol chain) an alkyl chain substituted with a single halogen group, preferably a chlorine group.
  • a polyethylene glycol chain of from 1 to 10, preferably 1 to 6, often 1 to 4 ethylene glycol units
  • the alkylene (often, a methylene) group may be substituted with an amino acid sidechain group such as a sidechain group of a natural or unnatural amino acid, for example, alanine, ⁇ -alanine, arginine, asparagine, aspartic acid, cysteine, cystine, glutamic acid, glutamine, glycine, phenylalanine, histidine, isoleucine, lysine, leucine, methionine, proline, serine, threonine, valine, tryptophan or tyrosine.
  • the term“unsubstituted” shall mean substituted only with hydrogen atoms.
  • a range of carbon atoms which includes C 0 means that carbon is absent and is replaced with H.
  • a range of carbon atoms which is C 0 -C 6 includes carbons atoms of 1, 2, 3, 4, 5 and 6 and for C 0 , H stands in place of carbon.
  • substituted or“optionally substituted” shall mean independently (i.e., where more than substituent occurs, each substituent is independent of another substituent) one or more substituents (independently up to five substitutents, preferably up to three substituents, often 1 or 2 substituents on a moiety in a compound according to the present invention and may include substituents which themselves may be further substituted) at a carbon (or nitrogen) position anywhere on a molecule within context, and includes as substituents hydroxyl, thiol, carboxyl, cyano (C ⁇ N), nitro (NO 2 ), halogen (preferably, 1, 2 or 3 halogens, especially on an alkyl, especially a methyl group such as a trifluoromethyl), an alkyl group (preferably, C 1 -C 10 , more preferably, C 1 -C 6 ), aryl (especially phenyl and substituted phenyl for example benzyl or benzoyl), alkoxy group (
  • Substituents according to the present invention may include, for example–SiR 1 R 2 R 3 groups wherein each of R 1 and R 2 is as otherwise described herein and R 3 is H or a C 1 -C 6 alkyl group, preferably R 1 , R 2 , R 3 in this context is a C 1 - C 3 alkyl group (including an isopropyl or t-butyl group).
  • Each of the above-described groups may be linked directly to the substituted moiety or alternatively, the substituent may be linked to the substituted moiety (preferably in the case of an aryl or heteraryl moiety) through an optionally substituted–(CH 2 ) m - or alternatively an optionally substituted -(OCH 2 ) m -, - (OCH 2 CH 2 ) m - or–(CH 2 CH 2 O) m - group, which may be substituted with any one or more of the above-described substituents.
  • Alkylene groups alkylene groups–(CH 2 ) m - or–(CH 2 ) n - groups or other chains such as ethylene glycol chains, as identified above, may be substituted anywhere on the chain.
  • Preferred substitutents on alkylene groups include halogen or C 1 -C 6 (preferably C 1 -C 3 ) alkyl groups, which may be optionally substituted with one or two hydroxyl groups, one or two ether groups (O-C 1 -C 6 groups), up to three halo groups (preferably F), or a sideshain of an amino acid as otherwise described herein and optionally substituted amide (preferably carboxamide substituted as described above) or urethane groups (often with one or two C 0 -C 6 alkyl substitutents, which group(s) may be further substituted).
  • halogen or C 1 -C 6 (preferably C 1 -C 3 ) alkyl groups which may be optionally substituted with one or two hydroxyl groups, one or two ether groups (O-C 1 -C 6 groups), up to three halo groups (preferably F), or a sideshain of an amino acid as otherwise described herein and optionally substituted amide (preferably carboxamide substituted as described
  • the alkylene group (often a single methylene group) is substituted with one or two optionally substituted C 1 - C 6 alkyl groups, preferably C 1 -C 4 alkyl group, most often methyl or O-methyl groups or a sidechain of an amino acid as otherwise described herein.
  • a moiety in a molecule may be optionally substituted with up to five substituents, preferably up to three substituents. Most often, in the present invention moieties which are substituted are substituted with one or two substituents.
  • substituted (each substituent being independent of any other substituent) shall also mean within its context of use C 1 -C 6 alkyl, C 1 -C 6 alkoxy, halogen, amido, carboxamido, sulfone, including sulfonamide, keto, carboxy, C 1 -C 6 ester (oxyester or
  • R 1 and R 2 are each, within context, H or a C 1 -C 6 alkyl group (which may be optionally substituted with one or two hydroxyl groups or up to three halogen groups, preferably fluorine).
  • the term“substituted” shall also mean, within the chemical context of the compound defined and substituent used, an optionally substituted aryl or heteroaryl group or an optionally substituted heterocyclic group as otherwise described herein.
  • Alkylene groups may also be substituted as otherwise disclosed herein, preferably with optionally substituted C 1 -C 6 alkyl groups (methyl, ethyl or hydroxymethyl or hydroxyethyl is preferred, thus providing a chiral center), a sidechain of an amino acid group as otherwise described herein, an amido group as described hereinabove, or a urethane group O-C(O)-NR 1 R 2 group wherein R 1 and R 2 are as otherwise described herein, although numerous other groups may also be used as substituents.
  • Various optionally substituted moieties may be substituted with 3 or more substituents, preferably no more than 3 substituents and preferably with 1 or 2 substituents.
  • aryl or“aromatic”, in context, refers to a substituted (as otherwise described herein) or unsubstituted monovalent aromatic radical having a single ring (e.g., benzene, phenyl, benzyl) or condensed rings (e.g., naphthyl, anthracenyl, phenanthrenyl, etc.) and can be bound to the compound according to the present invention at any available stable position on the ring(s) or as otherwise indicated in the chemical structure presented.
  • aryl groups in context, may include heterocyclic aromatic ring systems“heteroaryl” groups having one or more nitrogen, oxygen, or sulfur atoms in the ring (moncyclic) such as imidazole, furyl, pyrrole, furanyl, thiene, thiazole, pyridine, pyrimidine, pyrazine, triazole, oxazole or fused ring systems such as indole, quinoline, indolizine, azaindolizine, benzofurazan, etc., among others, which may be optionally substituted as described above.
  • heterocyclic aromatic ring systems“heteroaryl” groups having one or more nitrogen, oxygen, or sulfur atoms in the ring (moncyclic) such as imidazole, furyl, pyrrole, furanyl, thiene, thiazole, pyridine, pyrimidine, pyrazine, triazole, oxazole or fused ring systems such as indo
  • heteroaryl groups which may be mentioned include nitrogen-containing heteroaryl groups such as pyrrole, pyridine, pyridone, pyridazine, pyrimidine, pyrazine, pyrazole, imidazole, triazole, triazine, tetrazole, indole, isoindole, indolizine, azaindolizine, purine, indazole, quinoline, dihydroquinoline, tetrahydroquinoline, isoquinoline, dihydroisoquinoline,
  • nitrogen-containing heteroaryl groups such as pyrrole, pyridine, pyridone, pyridazine, pyrimidine, pyrazine, pyrazole, imidazole, triazole, triazine, tetrazole, indole, isoindole, indolizine, azaindolizine, purine, indazole, quinoline, di
  • phenanthridine carbazole, carbazoline, perimidine, phenanthroline, phenacene, oxadiazole, benzimidazole, pyrrolopyridine, pyrrolopyrimidine and pyridopyrimidine; sulfur-containing aromatic heterocycles such as thiophene and benzothiophene; oxygen-containing aromatic heterocycles such as furan, pyran, cyclopentapyran, benzofuran and isobenzofuran; and aromatic heterocycles comprising 2 or more hetero atoms selected from among nitrogen, sulfur and oxygen, such as thiazole, thiadizole, isothiazole, benzoxazole, benzothiazole, benzothiadiazole, phenothiazine, isoxazole, furazan, phenoxazine, pyrazoloxazole, imidazothiazole, thienofuran, furopyrrole, pyridoxazine, furopyridine, fur
  • substituted aryl refers to an aromatic carbocyclic group comprised of at least one aromatic ring or of multiple condensed rings at least one of which being aromatic, wherein the ring(s) are substituted with one or more substituents.
  • an aryl group can comprise a substituent(s) selected from: -(CH 2 ) n OH, -(CH 2 ) n -O-(C 1 -C 6 )alkyl, -(CH 2 ) n -O-(CH 2 ) n - (C 1 -C 6 )alkyl, -(CH 2 ) n -C(O)(C 0 -C 6 ) alkyl, -(CH 2 ) n -C(O)O(C 0 -C 6 )alkyl, -(CH 2 ) n -OC(O)(C 0 - C 6 )alkyl, amine, mono- or di-(C 1 -C 6 alkyl) amine wherein the alkyl group on the amine is optionally substituted with 1 or 2 hydroxyl groups or up to three halo (preferably F, Cl) groups, OH, COOH, C 1 -C 6 alkyl,
  • Carboxyl denotes the group -C(O)OR, wherein R is hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl or substituted heteroaryl , whereas these generic substituents have meanings which are identical with definitions of the corresponding groups defined herein.
  • heteroaryl or“hetaryl” can mean but is in no way limited to an optionally substituted quinoline (which may be attached to the pharmacophore or substituted on any carbon atom within the quinoline ring), an optionally substituted indole (including dihydroindole), an optionally substituted indolizine, an optionally substituted azaindolizine (2, 3 or 4-azaindolizine) an optionally substituted benzimidazole, benzodiazole, benzoxofuran, an optionally substituted imidazole, an optionally substituted isoxazole, an optionally substituted oxazole (preferably methyl substituted), an optionally substituted diazole, an optionally substituted triazole, a tetrazole, an optionally substituted benzofuran, an optionally substituted thiophene, an optionally substituted thiazole (preferably methyl and/or thiol substituted), an optionally substituted quinoline (including di
  • S c is CHR SS , NR URE , or O;
  • R HET is H, CN, NO 2 , halo (preferably Cl or F), optionally substituted C 1 -C 6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups (e.g. CF 3 ), optionally substituted O(C 1 -C 6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted acetylenic group–C ⁇ C-R a wherein R a is H or a C 1 -C 6 alkyl group (preferably C 1 -C 3 alkyl);
  • R SS is H, CN, NO 2 , halo (preferably F or Cl), optionally substituted C 1 -C 6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups), optionally substituted O-(C 1 -C 6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted -C(O)(C 1 -C 6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups);
  • R URE is H, a C 1 -C 6 alkyl (preferably H or C 1 -C 3 alkyl) or a–C(O)(C 1 -C 6 alkyl), each of which groups is optionally substituted with one or two hydroxyl groups or up to three halogen, preferably fluorine groups, or an optionally substituted heterocycle, for example piperidine, morpholine, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, piperidine, piperazine, each of which is optionally substituted, and
  • Y C is N or C-R YC , wherein R YC is H, OH, CN, NO 2 , halo (preferably Cl or F), optionally substituted C 1 -C 6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups (e.g. CF 3 ), optionally substituted O(C 1 -C 6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted acetylenic group–C ⁇ C-R a wherein R a is H or a C 1 -C 6 alkyl group (preferably C 1 -C 3 alkyl).
  • arylkyl and“heteroarylalkyl” refer to groups that comprise both aryl or, respectively, heteroaryl as well as alkyl and/or heteroalkyl and/or carbocyclic and/or heterocycloalkyl ring systems according to the above definitions.
  • arylalkyl refers to an aryl group as defined above appended to an alkyl group defined above.
  • the arylalkyl group is attached to the parent moiety through an alkyl group wherein the alkyl group is one to six carbon atoms.
  • the aryl group in the arylalkyl group may be substituted as defined above.
  • heterocycle refers to a cyclic group which contains at least one heteroatom, i.e., O, N or S, and may be aromatic (heteroaryl) or non-aromatic.
  • heteroaryl moieties are subsumed under the definition of heterocycle, depending on the context of its use.
  • heterocyclics include: azetidinyl, benzimidazolyl, 1,4- benzodioxanyl, 1,3- benzodioxolyl, benzoxazolyl, benzothiazolyl, benzothienyl, dihydroimidazolyl, dihydropyranyl, dihydrofuranyl, dioxanyl, dioxolanyl, ethyleneurea, 1,3-dioxolane, 1,3-dioxane, 1,4-dioxane, furyl, homopiperidinyl, imidazolyl, imidazolinyl, imidazolidinyl, indolinyl, indolyl,
  • Heterocyclic groups can be optionally substituted with a member selected from the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxy, carboxyalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, hydroxyamino, alkoxyamino, nitro,—SO-alkyl,—SO-substituted alkyl,—SOary
  • heterocyclic groups can have a single ring or multiple condensed rings.
  • nitrogen heterocycles and heteroaryls include, but are not limited to, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthylpyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, phenanthroline, isothiazole, phenazine, isoxazole, phenoxazine, phenothiazine, imidazolidine, imidazoline, piperidine, piperazine, indoline, morpholino, piperidinyl, tetrahydrofur
  • heterocyclic also includes bicyclic groups in which any of the heterocyclic rings is fused to a benzene ring or a cyclohexane ring or another heterocyclic ring (for example, indolyl, quinolyl, isoquinolyl, tetrahydroquinolyl, and the like).
  • cycloalkyl can mean but is in no way limited to univalent groups derived from monocyclic or polycyclic alkyl groups or cycloalkanes, as defnied herein, e.g., saturated monocyclic hydrocarbon groups having from three to twenty carbon atoms in the ring, including, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and the like.
  • substituted cycloalkyl can mean but is in no way limited to a monocyclic or polycyclic alkyl group and being substituted by one or more substituents, for example, amino, halogen, alkyl, substituted alkyl, carbyloxy, carbylmercapto, aryl, nitro, mercapto or sulfo, whereas these generic substituent groups have meanings which are identical with definitions of the corresponding groups as defined in this legend.
  • Heterocycloalkyl refers to a monocyclic or polycyclic alkyl group in which at least one ring carbon atom of its cyclic structure being replaced with a heteroatom selected from the group consisting of N, O, S or P.
  • Substituted heterocycloalkyl refers to a monocyclic or polycyclic alkyl group in which at least one ring carbon atom of its cyclic structure being replaced with a heteroatom selected from the group consisting of N, O, S or P and the group is containing one or more substituents selected from the group consisting of halogen, alkyl, substituted alkyl, carbyloxy, carbylmercapto, aryl, nitro, mercapto or sulfo, whereas these generic substituent group have meanings which are identical with definitions of the
  • Exemplary AR-PROTAC Compounds [0169] As described above, in certain aspects, the description provides bifuctional PROTAC compounds comprising at least one ABM group, a linker, and at least one ULM (or VLM) group as described herein.
  • the compound is selected from the group consisting of compounds 1-864 (as described in Tables 2-30), and salts and polymorphs thereof.
  • the com ound is selected from the rou consisting of:
  • the present invention provides a library of compounds.
  • the library comprises more than one compound wherein each compound has a formula of ABM- L-ULM, wherein ULM is a ubiquitin pathway protein binding moiety (preferably, an E3 ubiquitin ligase moiety as otherwise disclosed herein), e.g., a VLM, and ABM is an AR protein binding moiety, wherein ABM is coupled (preferably, through a linker moiety) to ULM, and wherein the ubiquitin pathway protein binding moiety recognizes an ubiquitin pathway protein, in particular, an E3 ubiquitin ligase.
  • ULM is a ubiquitin pathway protein binding moiety (preferably, an E3 ubiquitin ligase moiety as otherwise disclosed herein), e.g., a VLM
  • ABM is an AR protein binding moiety
  • ABM is coupled (preferably, through a linker moiety) to ULM
  • the ubiquitin pathway protein binding moiety recognizes an ubiquitin pathway
  • compositions comprising the pharmaceutically acceptable salts, in particular, acid or base addition salts of compounds of the present invention.
  • salt is used throughout the specification to describe, where applicable, a salt form of one or more of the compounds described herein which are presented to increase the solubility of the compound in the gastic juices of the patient's gastrointestinal tract in order to promote dissolution and the bioavailability of the compounds.
  • Pharmaceutically acceptable salts include those derived from pharmaceutically acceptable inorganic or organic bases and acids, where applicable. Suitable salts include those derived from alkali metals such as potassium and sodium, alkaline earth metals such as calcium, magnesium and ammonium salts, among numerous other acids and bases well known in the pharmaceutical art. Sodium and potassium salts are particularly preferred as neutralization salts of the phosphates according to the present invention.
  • the acids which are used to prepare the pharmaceutically acceptable acid addition salts of the aforementioned base compounds useful in this invention are those which form non- toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions, such as the hydrochloride, hydrobromide, hydroiodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, acetate, lactate, citrate, acid citrate, tartrate, bitartrate, succinate, maleate, fumarate, gluconate, saccharate, benzoate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and pamoate [i.e., 1,1'-methylene-bis-(2-hydroxy-3 naphthoate)]salts, among numerous others.
  • non- toxic acid addition salts i.e., salts containing pharmacologically acceptable anions,
  • Pharmaceutically acceptable base addition salts may also be used to produce pharmaceutically acceptable salt forms of the compounds or derivatives according to the present invention.
  • the chemical bases that may be used as reagents to prepare pharmaceutically acceptable base salts of the present compounds that are acidic in nature are those that form non- toxic base salts with such compounds.
  • Such non-toxic base salts include, but are not limited to those derived from such pharmacologically acceptable cations such as alkali metal cations (eg., potassium and sodium) and alkaline earth metal cations (eg, calcium, zinc and magnesium), ammonium or water-soluble amine addition salts such as N-methylglucamine-(meglumine), and the lower alkanolammonium and other base salts of pharmaceutically acceptable organic amines, among others.
  • alkali metal cations eg., potassium and sodium
  • alkaline earth metal cations eg, calcium, zinc and magnesium
  • ammonium or water-soluble amine addition salts such as N-methylglucamine-(meglumine)
  • the lower alkanolammonium and other base salts of pharmaceutically acceptable organic amines among others.
  • compositions comprising compounds as described herein, including salts thereof, and a pharmaceutically acceptable carrier.
  • the compositions are therapeutic or pharmaceutical compositions comprising an effective amount of a compound as described herein and a pharmaceutally acceptable carrier.
  • the amount of compound in a pharmaceutical composition of the instant invention that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host and disease treated, the particular mode of administration.
  • an amount between 0.1 mg/kg and 1000 mg/kg body weight/day of active ingredients is administered dependent upon potency of the agent.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50.
  • Compounds that exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture.
  • IC50 i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms
  • levels in plasma may be measured, for example, by high performance liquid chromatography.
  • compositions of the present invention may be formulated in a conventional manner using one or more pharmaceutically acceptable carriers and may also be administered in controlled-release formulations.
  • Pharmaceutically acceptable carriers that may be used in these pharmaceutical compositions include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as prolamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
  • the active compound is included in the pharmaceutically acceptable carrier or diluent in an amount sufficient to deliver to a patient a therapeutically effective amount for the desired indication, without causing serious toxic effects in the patient treated.
  • a preferred dose of the active compound for all of the herein-mentioned conditions is in the range from about 10 ng/kg to 300 mg/kg, preferably 0.1 to 100 mg/kg per day, more generally 0.5 to about 25 mg per kilogram body weight of the recipient/patient per day.
  • a typical topical dosage will range from 0.01-5% wt/wt in a suitable carrier.
  • the compound is conveniently administered in any suitable unit dosage form, including but not limited to one containing less than 1mg, 1 mg to 3000 mg, preferably 5 to 500 mg of active ingredient per unit dosage form.
  • An oral dosage of about 25-250 mg is often convenient.
  • the active ingredient is preferably administered to achieve peak plasma concentrations of the active compound of about 0.00001-30 mM, preferably about 0.1-30 ⁇ M. This may be achieved, for example, by the intravenous injection of a solution or formulation of the active ingredient, optionally in saline, or an aqueous medium or administered as a bolus of the active ingredient. Oral administration is also appropriate to generate effective plasma concentrations of active agent.
  • the concentration of active compound in the drug composition will depend on absorption, distribution, inactivation, and excretion rates of the drug as well as other factors known to those of skill in the art. It is to be noted that dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition.
  • the active ingredient may be administered at once, or may be divided into a number of smaller doses to be administered at varying intervals of time.
  • preferred carriers are physiological saline or phosphate buffered saline (PBS).
  • the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • a controlled release formulation including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides,
  • polyglycolic acid collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art.
  • Liposomal suspensions may also be pharmaceutically acceptable carriers. These may be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No.4,522,811 (which is incorporated herein by reference in its entirety).
  • liposome formulations may be prepared by dissolving appropriate lipid(s) (such as stearoyl phosphatidyl ethanolamine, stearoyl phosphatidyl choline, arachadoyl phosphatidyl choline, and cholesterol) in an inorganic solvent that is then evaporated, leaving behind a thin film of dried lipid on the surface of the container. An aqueous solution of the active compound are then introduced into the container. The container is then swirled by hand to free lipid material from the sides of the container and to disperse lipid aggregates, thereby forming the liposomal suspension.
  • appropriate lipid(s) such as stearoyl phosphatidyl ethanolamine, stearoyl phosphati
  • compositions comprising compounds described herein can be in any suitable dosage form configured to be delivered by any suitable route.
  • the compounds can be any suitable dosage form configured to be delivered by any suitable route.
  • the compounds can be any suitable dosage form configured to be delivered by any suitable route.
  • the compounds can be any suitable dosage form configured to be delivered by any suitable route.
  • the compounds can be any suitable dosage form configured to be delivered by any suitable route.
  • the compounds can be any suitable dosage form configured to be delivered by any suitable route.
  • the compounds can be any suitable dosage form configured to be delivered by any suitable route.
  • the compounds can be any suitable dosage form configured to be delivered by any suitable route.
  • the compounds can be any suitable route.
  • any appropriate route for example, orally, parenterally, intravenously, intradermally, subcutaneously, or topically, including transdermally, in liquid, cream, gel, or solid form, rectally, nasally, buccally, vaginally or via an implanted reservoir or by aerosol form.
  • parenteral includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • the compositions are administered orally, intraperitoneally or intravenously.
  • the compounds as described herein may be administered in single or divided doses by the oral, parenteral or topical routes.
  • Administration of the active compound may range from continuous (intravenous drip) to several oral administrations per day (for example, Q.I.D.) and may include oral, topical, parenteral, intramuscular, intravenous, sub-cutaneous, transdermal (which may include a penetration enhancement agent), buccal, sublingual and suppository administration, among other routes of administration.
  • Enteric coated oral tablets may also be used to enhance bioavailability of the compounds from an oral route of administration. The most effective dosage form will depend upon the pharmacokinetics of the particular agent chosen as well as the severity of disease in the patient.
  • Administration of compounds as sprays, mists, or aerosols for intra-nasal, intra- tracheal or pulmonary administration may also be used.
  • Compounds as described herein may be administered in immediate release, intermediate release or sustained or controlled release forms. Sustained or controlled release forms are preferably administered orally, but also in suppository and transdermal or other topical forms. Intramuscular injections in liposomal form may also be used to control or sustain the release of compound at an injection site.
  • Sterile injectable forms of the compositions as described herein may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally- acceptable diluent or solvent, for example as a solution in 1, 3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or di-glycerides.
  • Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • oils such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as Ph. Helv or similar alcohol.
  • compositions as described herein may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions.
  • carriers which are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried corn starch.
  • aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
  • Oral compositions will generally include an inert diluent or an edible carrier. They may be enclosed in gelatin capsules or compressed into tablets.
  • the active compound or its prodrug derivative can be incorporated with excipients and used in the form of tablets, troches, or capsules.
  • compositions are included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a dispersing agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a dispersing agent such as alginic acid, Primogel, or corn starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • dosage unit form When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil.
  • dosage unit forms can contain various other materials which modify the physical form of the dosage unit, for example, coatings of sugar, shellac, or enteric agents.
  • the active compound or pharmaceutically acceptable salt thereof can be administered as a component of an elixir, suspension, syrup, wafer, chewing gum or the like.
  • a syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.
  • compositions as described herein may be administered in the form of suppositories for rectal administration.
  • suppositories for rectal administration.
  • a suitable non-irritating excipient which is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter, beeswax and polyethylene glycols.
  • compositions of this invention may also be administered topically. Suitable topical formulations are readily prepared for each of these areas or organs. Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-acceptable transdermal patches may also be used.
  • the pharmaceutical compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers.
  • Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • the compounds may be coated onto a stent which is to be surgically implanted into a patient in order to inhibit or reduce the likelihood of occlusion occurring in the stent in the patient.
  • the pharmaceutical compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers.
  • suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • the pharmaceutical compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with our without a preservative such as
  • compositions may be formulated in an ointment such as petrolatum.
  • compositions of this invention may also be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
  • Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid;
  • a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents
  • antibacterial agents such as benzyl alcohol or methyl parabens
  • antioxidants such as ascorbic acid or sodium bisulfite
  • chelating agents such as ethylenediaminetetraacetic acid
  • buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the parental preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • a patient or subject in need of therapy using compounds as described herein can be treated by administering to the patient (subject) an effective amount of the compound including pharmaceutically acceptable salts, solvates or polymorphs, thereof optionally in a pharmaceutically acceptable carrier or diluent, either alone, or in combination with other known agents.
  • compositions according to the present description include, but not limited to, for example, cancer (e.g., prostate cancer), and Kennedy’s disease.
  • the therapeutic or pharmaceutical compositions comprise an effective amount of an additional biologically or bioactive active agent, e.g., an agent effective for the treatment of cancer, that is co-administered.
  • coadministration shall mean that at least two compounds or compositions are administered to the patient at the same time, such that effective amounts or concentrations of each of the two or more compounds may be found in the patient at a given point in time.
  • compounds according to the present invention may be co-administered to a patient at the same time, the term embraces both administration of two or more agents at the same time or at different times, provided that effective concentrations of all coadministered compounds or compositions are found in the subject at a given time.
  • one or more of the present compounds described above are coadministered in combination with at least one additional bioactive agent, especially including an anticancer agent.
  • the co- administration of compounds results in synergistic therapeutic, including anticancer therapy.
  • the description provides a composition comprising an effective amount of two or more of the PROTAC compounds as described herein, and a pharmaceutically acceptable carrier.
  • the composition further comprises an effective or synergistic amount of another bioactive agent that is not a PROTAC compound.
  • compositions comprising combinations of an effective amount of at least one bifunctional compound according to the present invention, and one or more of the compounds otherwise described herein, all in effective amounts, in combination with a pharmaceutically effective amount of a carrier, additive or excipient, represents a further aspect of the present invention.
  • bioactive agent is used to describe an agent, other than the PROTAC compounds described herein, which is used in combination with the present compounds as an agent with biological activity to assist in effecting an intended therapy, inhibition and/or prevention/prophylaxis for which the present compounds are used.
  • Preferred bioactive agents for use herein include those agents which have pharmacological activity similar to that for which the present compounds are used or administered and include for example, anti-cancer agents.
  • the term“additional anti-cancer agent” is used to describe an anti-cancer agent, which may be combined with PROTAC compounds according to the present description to treat cancer.
  • these agents include, for example, everolimus, trabectedin, abraxane, TLK 286, AV- 299, DN-101, pazopanib, GSK690693, RTA 744, ON 0910.Na, AZD 6244 (ARRY-142886), AMN-107, TKI-258, GSK461364, AZD 1152, enzastaurin, vandetanib, ARQ-197, MK-0457, MLN8054, PHA-739358, R-763, AT-9263, a FLT-3 inhibitor, an androgen receptor inhibitor, a VEGFR inhibitor, an EGFR TK inhibitor, an aurora kinase inhibitor, a PIK-1 modulator, a Bcl-2 inhibitor, an HDAC inhbitor, a c-MET
  • the disclosure provides methods of modulating protein ubiquitination and degradation in a subject, e.g., a cell, a tissue, mammal, or human patient, the method comprising administering an effective amount of a PROTAC compound as described herein or a composition comprising an effective amount of the same to a subject, wherein the compound or composition comprising the same is effective in modulating protein ubquitination and degration of the protein in the subject.
  • the protein is androgen receptor (AR).
  • the description provides a method for regulating protein activity of the androgen receptor in a patient in need comprising administering to said patient an amount of a compound as described herein to a patient.
  • the description provides a method of treating a disease state or condition in a patient wherein dysregulated protein activity is responsible for said disease state or condition, said method comprising administering to said patient an effective amount of a compound as described herein to said patient in order to regulate said protein activity in said patient.
  • the protein is AR.
  • the terms“treat”,“treating”, and“treatment”, etc., as used herein, refer to any action providing a benefit to a patient for which the present compounds may be administered, including the treatment of any disease state or condition which is modulated through the protein to which the present compounds bind.
  • Disease states or conditions, including cancer, which may be treated using compounds according to the present invention are set forth hereinabove.
  • the disclosure provides methods of modulating AR protein ubiquitination and degradation in a subject, e.g., a cell, a tissue, mammal, or human patient, the method comprising administering an effective amount of a compound as described herein or a composition comprising an effective amount of the same to a subject, wherein the compound or composition comprising the same is effective in modulating AR protein ubquitination and degration of the protein in the subject.
  • the disclosure provides methods of treating or ameliorating a symptom of a disease related to AR activity in a subject, e.g., a cell, a tissue, mammal, or human patient, the method comprising administering an effective amount of a compound as described herein or a composition comprising an effective amount of the same to a subject in need thereof, wherein the compound or composition comprising the same is effective in treating or
  • the disease or disorder is asthma, multiple sclerosis, cancer, prostate cancer, Kenney’s disease, ciliopathies, cleft palate, diabetes, heart disease, hypertension, inflammatory bowel disease, mental retardation, mood disorder, obesity, refractive error, infertility, Angelman syndrome, Canavan disease, Coeliac disease, Charcot–Marie–Tooth disease, Cystic fibrosis, Duchenne muscular dystrophy, Haemochromatosis, Haemophilia, Klinefelter's syndrome, Neurofibromatosis, Phenylketonuria, Polycystic kidney disease, (PKD1) or 4 (PKD2) Prader–Willi syndrome, Sickle-cell disease, Tay–Sachs disease, Turner syndrome.
  • cancer is squamous-cell carcinoma, basal cell carcinoma, adenocarcinoma, hepatocellular carcinomas, and renal cell carcinomas, cancer of the bladder, bowel, breast, cervix, colon, esophagus, head, kidney, liver, lung, neck, ovary, pancreas, prostate, and stomach; leukemias; benign and malignant lymphomas, particularly Burkitt's lymphoma and Non-Hodgkin's lymphoma; benign and malignant melanomas; myeloproliferative diseases; sarcomas, including Ewing's sarcoma, hemangiosarcoma, Kaposi's sarcoma, liposarcoma, myosarcomas, peripheral neuroepithelioma, synovial sarcoma, gliomas, astrocytomas, oligodendrogliomas, ependymomas, gliobasto
  • the disease to be treated is cancer, e.g., prostate cancer, or Kennedy’s Disease.
  • the subject is a human.
  • the disclosure provides methods of treating or ameliorating a symptom of a disease related to AR activity in a subject, e.g., a cell, a tissue, mammal, or human patient, the method comprising administering an effective amount of a compound as described herein or a composition comprising an effective amount of the same and an effective or synergistic amount of another bioactive agent to a subject in need thereof, wherein the composition comprising the same is effective in treating or ameliorating a symptom of a disease related to AR activity in the subject.
  • the disease to be treated is cancer, e.g., prostate cancer, or Kennedy’s Disease.
  • the subject is a human.
  • the additional bioactive agent is an anti-cancer agent.
  • the present invention relates to a method for treating a disease state by degrading a protein or polypeptide through which a disease state or condition is modulated comprising administering to said patient or subject an effective amount of at least one compound as described hereinabove, optionally in combination with an additional bioactive agent.
  • the method according to the present invention may be used to treat a large number of disease states or conditions including cancer, by virtue of the administration of effective amounts of at least one compound described herein.
  • the disclosure provides methods for identifying the effects of the degradation of proteins of interest in a biological system using compounds according to the present invention.
  • kits comprising compounds or compositions as described herein.
  • the kit may be promoted, distributed, or sold as a unit for performing the methods of the present invention.
  • the kits of the present invention may preferably contain instructions which describe a suitable use.
  • Such kits can be conveniently used, e.g., in clinical settings, to treat patients exhibiting symptoms of, e.g., cancer or Kennedy’s Disease.
  • Preparative HPLC purifications were performed on a Waters ® UV-Directed Purification System equipped with 2545 Binary Gradient Module, 2767 Sample Manager and 2489 UV/Visible Detector, controlled by MassLynx V4.1 software. All purification work was completed using the following columns: Atlantis Prep T3 OBD Column, SunFire Prep C18 OBD Column and XBridge Prep Phenyl OBD Column. The mobile phases were water (with 0.1%TFA or 0.01% NH 4 HCO 3 ) and acetonitrile; all reagents used were of HPLC grade. The flow rate was 30ml/min.
  • a 1:1000 LC packings flow splitter allowed transfer of a small portion of the eluent into the UV detector.
  • the electrospray source was set at 3.0 kV capillary voltage, 30 V conevoltage, 110°C source temperature, 350°C desolvation temperature, 600L/h desolvation gas flow, and 60L/h cone gas flow.
  • the multiplier was set at 550 for preparative tune method.
  • Analytical LC-MS data was collected on a Shimadzu LCMS-2020 with a mobile phase of 0.05% TFA in Acetonitrile (A) and 0.05% TFA in HPLC grade water (B); 0.1% FA in Acetonitrile (A) and 0.1% FA in HPLC grade water (B); Acetonitrile (A) and 5 mM ammonium bicarbonate in HPLC grade water (B).
  • A 0.1% v/v solution of formic acid in water.
  • B 0.1% v/v solution of formic acid in acetonitrile.
  • the UV detection is an averaged signal from wavelength of 210nm to 350nm and mass spectra are recorded on a mass spectrometer using positive mode electrospray ionization.
  • a PROTAC of ABM-L-ULM may be prepared by the general approaches described below (scheme 3-4), together with synthetic methods known in the art of organic chemistry, or modifications and derivatizations that are familiar to those of ordinary skill in the art.
  • the compounds of the Formula I, or their pharmaceutically acceptable salts may be prepared by the general approaches described below (scheme 5-6), together with synthetic methods known in the art of organic chemistry, or modifications and derivatizations that are familiar to those of ordinary skill in the art.
  • L, ABM, ULM groups, W 1 , W 2 , W 3 , W 4 , X 1 , X 2 , Y 1 , Y 2 , R 1 , R 2 , and R P are as define above.
  • stage 1 and stage 2 transformations in scheme 5 and scheme 6 may involve 1 or multiple synthetic steps. These are routine methods known in the art such as those methods disclosed in standard reference books such as the Compendium of Organic Synthetic Methods, Vol. I-VI (Wiley-lnterscience); or the Comprehensive Organic Transformations, by R.C. Larock (Wiley-lnterscience). Unless otherwise indicated, the substituents in the schemes are defined as above. Isolation and purification of the products is accomplished by standard procedures, which are known to a chemist of ordinary skill.
  • RG 1 is a moiety with a suitable nucleophile such as -OH and RG 2 is a moiety with a suitable leaving group such as halogen, -OMs, or–OTs.
  • a RG 1 containing intermediate is reacted with a RG 2 containing intermediate in a suitable solvent.
  • suitable solvents include, but are not limited to, water, ethers such as THF, glyme, and the like; chlorinated solvents such as DCM, 1,2- dichloroethane (DCE) or CHCI3 and the like, toluene, benzene and the like, DMF, DMSO, MeCN.
  • a base may be added to the reaction to facilitate the reaction. Suitable bases include, but are not limited to, Cs 2 CO 3 , K 2 CO 3 , and the like.
  • the above process may be carried out at temperatures between about -78 °C and about 150 °C. Preferably, the reaction is carried out between about 20 °C and about 120 °C.
  • RG 3 is a moiety contains a–COOH group and RG 4 is a moiety contains a suitable amine group.
  • a RG 3 containing intermediate is reacted with a RG 4 containing intermediate in a suitable solvent in the presence of a suitable amide coupling reagent.
  • suitable solvents include, but are not limited to, water, ethers such as THF, glyme, and the like; chlorinated solvents such as DCM, 1,2-dichloroethane (DCE) or CHCl 3 and the like, toluene, benzene and the like, DMF, DMSO, MeCN.
  • a suitable amide coupling reagent include, but are not limited to, DCC, EDC, HATU, HBTU, PyBOP and the like.
  • a base is often added to the reaction. Suitable bases include, but are not limited to, TEA, DIPEA, and the like.
  • the above process may be carried out at temperatures between about -78 °C and about 150 °C. Preferably, the reaction is carried out between about 0 °C and about 100 °C.
  • the process to produce compounds of the present invention is preferably carried out at about atmospheric pressure although higher or lower pressures can be used if desired. Substantially equimolar amounts of reactants are preferably used although higher or lower amounts can also be used.
  • the compounds of Formulae II-IV may be prepared by the methods similar to chemistry illustrated above for synthesis of compounds of Formula I (scheme 3-6), together with synthetic methods known in the art of organic chemistry, or modifications and derivatizations that are familiar to those of ordinary skill in the art: ; and
  • ABM compounds are active without forming
  • ABM-1 2-chloro-4-(3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1- l benzonitrile
  • Step 1 Synthesis of 2-chloro-4-isothiocyanatobenzonitrile (B).
  • Step 2 Synthesis of 2-chloro-4-[3-(4-hydroxyphenyl)-5-imino-4, 4-dimethyl-2- sulfanylideneimidazolidin-1-yl]benzonitrile (D).
  • Step 3 Synthesis of 2-chloro-4-[3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo- ⁇ 2- sulfanylideneimidazolidin-1-yl]benzonitrile (ABM-1).
  • ABM-2 2-fluoro-4-(3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)benzonitrile:
  • ABM-3 4-(3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1- yl)-2-(trifluoromethyl)benzonitrile:
  • ABM-4 5-(3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1- yl)-3-(trifluoromethyl)picolinoni ril
  • ABM-5 4-(3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1- yl)-2-methoxybenzonitrile:
  • ABM-6 4-(3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1- yl)-2-methylbenzonitrile:
  • ABM-7 3-chloro-5-(3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)picolinonitrile: .
  • ABM-8 4-(1-(4-hydroxyphenyl)-4-oxo-2-thioxo-8-oxa-1,3- diazaspiro[4.5]decan-3-yl)-2-(trifluorometh l benzonitrile:
  • ABM-9 4-(1-(4-hydroxyphenyl)-8-methyl-4-oxo-2-thioxo-1,3,8- triazaspiro[4.5]decan-3-yl)-2-(trifluorometh l benzonitrile:
  • ABM-10 4-(5-(4-hydroxyphenyl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7- yl)-2-(trifluoromethyl)benzonitrile
  • ABM-11 5-(5-(4-hydroxyphenyl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7- yl)-3-(trifluoromethyl)picolinonitrile:
  • ABM-12 4-(4-(3-(4-cyano-3-(trifluoromethyl)phenyl)-5,5-dimethyl-4-oxo-2- thioxoimidazolidin-1-yl)phenyl)butanoic acid: .
  • ABM-13 2-chloro-4-(3-(4'-hydroxybiphenyl-4-yl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)benzonitrile:
  • ABM-14 4-(3-(4'-hydroxybiphenyl-4-yl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)-2-
  • ABM-15 5-(3-(4'-hydroxybiphenyl-4-yl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)-3-(trifluoromethyl)picolinonitrile:
  • ABM-16 4-(3-(3-fluoro-4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile:
  • ABM-17 1-(4-hydroxyphenyl)-5,5-dimethyl-3-(4-nitro-3- (trifluoromethyl)phenyl)-2-thioxoimidazolidin-4-one: .
  • ABM-18 4-(3-(3,5-difluoro-4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile:
  • ABM-19 4-(3-(4-hydroxyphenyl)-4,4-dimethyl-2,5-dioxoimidazolidin-1-yl)-2- (trifluoromethyl)benzonitrile:
  • ABM-20 4-(3-(6-hydroxypyridin-3-yl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile:
  • ABM-21 2-chloro-4-(3-(3-fluoro-4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)benzonitrile:
  • ABM-22 4-(3-(3-fluoro-4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)-2-methoxybenzonitrile: .
  • ABM-23 5-(3-(3-fluoro-4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)-3-(trifluoromethyl)picolinonitrile:
  • ABM-24 5-(3-(2-fluoro-4'-hydroxybiphenyl-4-yl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)-3-(trifluorometh l icolinonitrile:
  • ABM-25 4-(4,4-dimethyl-5-oxo-3-(4-(piperidin-4-yl)phenyl)-2- thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile:
  • ABM-26 trans-2-Chloro-4-[3-amino-2,2,4,4- tetramethylcyclobutoxy]benzonitrile.
  • ABM-27 cis-2-Chloro-4-[3-amino-2,2,4,4- tetramethylcyclobutoxy]benzonitrile
  • Step 1 Synthesis of tot-butyl (4-((tra «5-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)carbamoyl)phenyl)carbamate.
  • Step 2 Synthesis of trans 4-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetram ethyl ey cl obutyl ]b enzami de .
  • ABM-31 trans 5-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]pyrazine ⁇ 2-carboxamide
  • ABM-32 trans 2-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyljpyrimidine-5-carboxamid
  • ABM-33 4-Methoxy-N-[(lr,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyljbenzamide
  • ABM-34 trans 1-(2-Hydroxyethyl)-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]-1H-pyrazole-4-carboxamide
  • ABM-35 trans 6-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]pyridine-3-carboxamide.
  • ABM-36 trans 4-[(5-Hydroxypentyl)amino]-N-[3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetrameth lc clobut l benzamide
  • ABM-37 trans tert-Butyl 2-( ⁇ 5-[(4- ⁇ [3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamo l hen l amino ent l ox acetate
  • ABM-38 tert-butyl trans-(3-(3-chloro-4-cyanophenoxy)-2,2- dimethylcyclobutyl)carbamate] and ABM-39: tert-butyl cis-(3-(3-chloro-4-cyanophenoxy)- 2,2-dimethylcyclobutyl)carbamate.
  • Step 1 Synthesis of ((vinyloxy)methyl)benzene (B).
  • Step 2 Synthesis of 3-(benzyloxy)-2,2-dimethylcyclobutanone (C).
  • Step 5 Synthesis of tert-butyl (3-(benzyloxy)-2,2-dimethylcyclobutyl)carbamate (F).
  • Step 6 Synthesis of tert-butyl (3-hydroxy-2,2-dimethylcyclobutyl)carbamate (G).
  • Step 7 Synthesis of tert-butyl trans-(3-(3-chloro-4-cyanophenoxy)-2,2- dimethylcyclobutyl)carbamate (ABM-38) and tert-butyl cis-(3-(3-chloro-4-cyanophenoxy)-2,2- dimethylcyclobutyl)carbamate (ABM-39).
  • Step 3 Synthesis of tert-butyl (2S,4R)-4-hydroxy-2-( ⁇ [4-(4-methyl-1,3-thiazol- 5-yl)phenyl]methyl ⁇ carbamoyl)pyrrolidine-1-carboxylate (J)
  • reaction mixture was diluted with water (20 mL) and extracted with ethyl acetate (50 mL x 3). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (50 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent:
  • Step 4 Synthesis of (2S,4R)-4-hydroxy-N- ⁇ [4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl ⁇ pyrrolidine-2-carboxamide hydrochloride (K)
  • Step 5 Synthesis of tert-butyl N-[(2S)-1-[(2S,4R)-4-hydroxy-2-( ⁇ [4-(4-methyl- 1,3-thiazol-5-yl)phenyl]methyl ⁇ carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamate (M)
  • Step 6 Synthesis of (2S,4R)-1-[(2S)-2-amino-3,3-dimethylbutanoyl]-4-hydroxy- N- ⁇ [4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl ⁇ pyrrolidine-2-carboxamide hydrochloride (ULM-1)
  • ULM-2 (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4- (thiazol-5-yl)benzyl)pyrrolidine-2-carboxamide:
  • ULM-2 was synthesized according to similar procedure described above for the synthesis of ULM-1, utilizing 4-bromobenzonitrile and 1,3-thiazole as starting materials.
  • LC-MS (ES + ): m/z 417.10 [MH + ], t R 0.51 min (2.0 minute run).
  • ULM-3 (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-((S)-1- (4-(4-methylthiazol-5-yl)phenyl)ethyl)pyrrolidine-2-carboxamide:
  • Step 1 Synthesis of tert-butyl N-[(1S)-1-(4-bromophenyl)ethyl]carbamate (O)
  • Step 2 Synthesis of tert-butyl N-[(1S)-1-[4-(4-methyl-1,3-thiazol-5- yl)phenyl]ethyl]carbamate (P)
  • Step 1 1. Synthesis of 4-(1,3-oxazol-5-yl)benzonitrile (S)
  • ULM-6 was synthesized according to similar procedure described above for the synthesis of ULM-1, utilizing 4-chlorobenzonitrile as the starting material.
  • ULM-7 was synthesized according to similar procedure described above for the synthesis of ULM-1, utilizing 4-cyanobenzonitrile as the starting material.
  • ULM-8 was synthesized according to similar procedure described above for the synthesis of ULM-1, utilizing (S)-2-(tert-butoxycarbonylamino)-3-methylbutanoic acid and 4- methyl-1,3-thiazole (F) as starting materials.
  • ULM-9 was synthesized according to similar procedure described above for the synthesis of ULM-1, utilizing (S)-2-(tert-butoxycarbonylamino)-3-methylbutanoic acid and 1,3- thiazole as starting materials.
  • ULM-10 was synthesized according to similar procedure described above for the synthesis of ULM-5, utilizing (S)-2-(tert-butoxycarbonylamino)-3-methylbutanoic acid as starting material.
  • ULM-11 was synthesized according to similar procedure described above for the synthesis of ULM-1, utilizing 1-methylpyrazole as the starting material.
  • Step 1 2-(aminomethyl)-5-(4-methyl-1,3-thiazol-5-yl)phenol (BI)
  • Step 3 Synthesis of 9H-fluoren-9-ylmethyl (2S,4R)-4-(tert-butoxy)-2-( ⁇ [2- hydroxy-4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl ⁇ carbamoyl)pyrrolidine-1-carboxylate (BJ)
  • Step 4 Synthesis of (2S,4R)-4-(tert-butoxy)-N- ⁇ [2-hydroxy-4-(4-methyl-1,3- thiazol-5-yl)phenyl]methyl ⁇ pyrrolidine-2-carboxamide (BK)
  • Step 5 Synthesis of (2S,4R)-4-(tert-butoxy)-N- ⁇ [2-hydroxy-4-(4-methyl-1,3- thiazol-5-yl)phenyl]methyl ⁇ -1-[(2S)-3-methyl-2-(1-oxo-2,3-dihydro-1H-isoindol-2- yl)butanoyl]pyrrolidine-2-carboxamide ULM-12) [0386] To a stirred solution of (2S)-3-methyl-2-(1-oxo-2,3-dihydro-1H-isoindol-2- yl)butanoic acid (3.6 g, 15.43 mmol) in N,N-dimethylformamide (50 mL) was added DIEA (2.7 g, 20.93 mmol), HATU (5.89 g, 15.49 mmol) and (2S,4R)-4-(tert-butoxy)-N- ⁇ [2-hydroxy-4-(4
  • ULM 15 (2S,4R)-1-((S)-2-Amino-3,3-dimethylbutanoyl)-4-hydroxy-N-((R)- 2-hydroxy-1-(4-(4-methylthiazol-5-yl)phenyl)ethyl)pyrrolidine-2-carboxamide
  • Step 1 Synthesis of 1-(4-Bromophenyl)-2-hydroxyethanone (B).
  • Step 2 Synthesis of 1-(4-Bromophenyl)-2-(tert-butyldimethylsilyloxy)ethanone (C).
  • Step 3 Synthesis of (S,Z)-N-(1-(4-Bromophenyl)-2-(tert- butyldimethylsilyloxy)ethylidene)-2-methylpropane-2-sulfinamide (D).
  • Step 4 Synthesis of (S)-N-((R)-1-(4-Bromophenyl)-2-(tert- butyldimethylsilyloxy)ethyl)-2-methylpropane-2-sulfinamide. (E).
  • Step 5 Synthesis of (R)-Tert-butyl 1-(4-bromophenyl)-2-hydroxyethylcarbamate (F).
  • Step 6 Synthesis of (R)-Tert-butyl 2-hydroxy-1-(4-(4-methylthiazol-5- yl)phenyl)ethylcarbamate (G).
  • Step 7 Synthesis of (R)-2-Amino-2-(4-(4-methylthiazol-5-yl)phenyl)ethanol hydrochloride (H).
  • Step 8 Synthesis of tert-butyl (S)-1-((2S,4R)-2-(((R)-2-Hydroxy-1-(4-(4- methylthiazol-5-yl) phenyl)ethyl)carbamoyl)-4-hydroxypyrrolidin-1-yl)-3,3-dimethyl-1- oxobutan-2-ylcarbamate (I).
  • Step 9 Synthesis of (2S,4R)-1-((S)-2-Amino-3,3-dimethylbutanoyl)-4-hydroxy- N-((R)-2-hydroxy-1-(4-(4-methylthiazol-5-yl)phenyl)ethyl)pyrrolidine-2-carboxamide hydrochloride (ULM-15).
  • Step 1 Synthesis of ( ⁇ [5-(prop-2-en-1-yloxy)pentyl]oxy ⁇ methyl)benzene
  • Step 3 Synthesis of tert-butyl 2-(3- ⁇ [5-(benzyloxy)pentyl]oxy ⁇ propoxy)acetate (Z)
  • Step 4 Synthesis of tert-butyl 2-[3-[(5-hydroxypentyl)oxy]propoxy]acetate (AA)
  • Step 5 Synthesis of tert-butyl 2-[3-( ⁇ 5-[(4- methylbenzenesulfonyl)oxy]pentyl ⁇ oxy)propoxy]acetate (AB) [0421] To a stirred solution of tert-butyl 2-[3-[(5-hydroxypentyl)oxy]propoxy]acetate (AA, 2.3 g, 8.32 mmol) in dichloromethane (30 mL) was added 4-methylbenzene-1-sulfonyl chloride (3.17 g, 16.63 mmol), triethylamine (2.52 g, 24.90 mmol) and 4-dimethylaminopyridine (203 mg, 1.66 mmol) at room temperature.
  • Step 1 Synthesis of 2-[3-( ⁇ 5-[(4-methylbenzenesulfonyl)oxy]pentyl ⁇ oxy)propoxy]acetic acid (L-1)
  • L-2 2-(3-(3,3-dimethyl-5-(tosyloxy)pentyloxy)propoxy)acetic acid .
  • L-3 2-(3-(3-hydroxy-5-(tosyloxy)pentyloxy)propoxy)acetic acid .
  • L-6 2-(2-((2S,3S)-3-(2-(tosyloxy)ethoxy)butan-2-yloxy)ethoxy)acetic acid
  • Step 1 Synthesis of 4- ⁇ 4-[(4-methylbenzenesulfonyl)oxy]butoxy ⁇ butan-1-ol (AE)
  • Step 2 Synthesis of ethyl 2-(4- ⁇ 4-[(4-methylbenzenesulfonyl)oxy]butoxy ⁇ butoxy)acetate (AF)
  • Step 3 Synthesis of 2-(4- ⁇ 4-[(4-methylbenzenesulfonyl)oxy]butoxy ⁇ butoxy)acetic acid (L-7)
  • L-8 tert-but l 2- 3- 4- tos lox butox ro ox acetate
  • Step 4 Synthesis of tert-butyl 2-(3- ⁇ 4-[(4- methylbenzenesulfonyl)oxy]butoxy ⁇ propoxy)acetate (L-8) [0447] To a stirred solution of tert-butyl 2-[3-(4-hydroxybutoxy)propoxy]acetate (AJ, 700 mg, 2.67 mmol) in dichloromethane (10 mL) was added 4-methylbenzene-1-sulfonyl chloride (558.4 mg, 2.93 mmol), TEA (539.5 mg, 5.33 mmol) and 4-dimethylaminopyridine (32.6 mg, 0.27 mmol). The resulting mixture was stirred overnight at room temperature.
  • L-9 was prepared in a similar manner as that used to prepare L-8, except that AK was used in place of AG.
  • Step1 Synthesis of tert-butyl 2-[(6-hydroxyhexa-2,4-diyn-1-yl)oxy]acetate (AP)
  • L-12 tert-butyl 4-(6-(tos lox hexa-24-di n lox butanoate
  • L-13 ethyl 2-(2-(2-aminoethoxy)ethoxy)acetate hydrochloride .
  • Step 1 Synthesis of tert-butyl N-[2-(2-hydroxyethoxy)ethyl]carbamate (AR)
  • Step 2 Synthesis of ethyl 2-[2-(2- ⁇ [(tert-butoxy)carbonyl]amino ⁇ ethoxy)ethoxy]acetate (AS)
  • Step 3 Synthesis of ethyl 2-[2-(2-aminoethoxy)ethoxy]acetate hydrochloride (L-13)
  • Step 2 Synthesis of ethyl 2-[(5- ⁇ [(tert-butoxy)carbonyl]amino ⁇ pentyl)oxy]acetate (AV)
  • Step 3 Synthesis of ethyl 2-(5-aminopentyloxy)acetate (L-14)
  • L-15 meth l 2- 2- 2- meth lamino ethox ethox acetate
  • Step 2 Synthesis of 2- ⁇ 2-[benzyl(methyl)amino]ethoxy ⁇ ethan-1-ol (AY)
  • Step 3 Synthesis of methyl 2-(2- ⁇ 2-[benzyl(methyl)amino]ethoxy ⁇ ethoxy)acetate (AZ) [0479] To a stirred solution of 2- ⁇ 2-[benzyl(methyl)amino]ethoxy ⁇ ethan-1-ol (AY, 2 g) in dichloromethane (20 mL) was added a solution of sodium hydroxide (37%) in water (20 mL) followed by tert-butyl 2-bromoacetate (7.76 g) and TBAC (2.78 g) at room temperature. The resulting mixture was stirred at room temperature for 15 hours. The aqueous layer was separated, and to which aq.
  • Step 4 Synthesis of methyl 2- ⁇ 2-[2-(methylamino)ethoxy]ethoxy ⁇ acetate (L-15)
  • L-16 ethyl 2-(5-(methylamino)pentyloxy)acetate .
  • Step 1 Synthesis of ethyl 2-[(5- ⁇ [(tert- butoxy)carbonyl](methyl)amino ⁇ pentyl)oxy]acetate (BB)
  • Step 2 Synthesis of ethyl 2- ⁇ [5-(methylamino)pentyl]oxy ⁇ acetate (L-16)
  • Step 1 Synthesis of tert-butyl 2- ⁇ 3-[2-(benzyloxy)ethoxy]propoxy ⁇ acetate (BD)
  • Step 2 Synthesis of tert-butyl 2-[3-(2-hydroxyethoxy)propoxy]acetate (BE)
  • Step 3 Synthesis of tert-butyl 2-(3- ⁇ 2-[(4- methylbenzenesulfonyl)oxy]ethoxy ⁇ propoxy)acetate (BF)
  • Step 4 Synthesis of 2-(3- ⁇ 2-[(4-methylbenzenesulfonyl)oxy]ethoxy ⁇ propoxy)acetic acid (L-17)
  • L-18 2-(2-hydroxyethoxy)ethyl 4-methylbenzenesulfonate .
  • L-20 ethyl 3-(2-(2-(tosyloxy)ethoxy)ethoxy)propanoate .
  • L-21 ethyl 5-(tosyloxy)pentanoate .
  • L-22 ethyl 3-(2-(tosyloxy)ethoxy)propanoate .
  • L-24 ethyl 3-(5-(tosyloxy)pentyloxy)propanoate .
  • L-28 ethyl 2-(2-(tosyloxy)ethoxy)acetate .
  • L-29 ethyl 2-(4-(2-(tosyloxy)ethoxy)butoxy)acetate .
  • L-31 2-((2R,3R)-3-(2-hydroxyethoxy)butan-2-yloxy)ethyl 4-methylbenzenesulfonate .
  • L-33 methyl 6-(4-(2-(2-(tert-butoxy)-2-oxoethoxy)ethyl)piperazin-1-yl)nicotinate
  • Step 1 Synthesis of tert-butyl 4-(5-(methoxycarbonyl)pyridin-2-yl)piperazine-1- carboxylate:
  • Step 2 Synthesis of methyl 6-(piperazin-1-yl)nicotinate
  • Step 3 Synthesis of methyl 6-(4-(2-(2-(tert-butoxy)-2-oxoethoxy)ethyl)piperazin-1- yl)nicotinate.
  • Example 1 (2S,4R)-1-((S)-2-(2-(3-(5-(4-(3-(4-cyano-3-(trifluoromethyl)phenyl)-5,5- dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)phenoxy)pentyloxy)propoxy)acetamido)-3,3- dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2- carboxamide:
  • Step 1 Synthesis of tert-butyl 2-(3- ⁇ [5-(4- ⁇ 3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl ⁇ phenoxy)pentyl]oxy ⁇ propoxy)acetate (BG)
  • Step 2 Synthesis of 2-(3-[[5-(4-[3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5-dimethyl-4- oxo-2-sulfanylideneimidazolidin-1-yl]phenoxy)pentyl]oxy]propoxy)acetic acid (BH)
  • Example 2 (2S,4R)-1-((S)-2-(2-(3-(5-(4-(3-(6-cyano-5-(trifluoromethyl)pyridin-3-yl)- 5,5-dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)phenoxy)pentyloxy)propoxy)acetamido)-3,3- dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2- carboxamide:
  • Step 1 Synthesis of 2-[3-( ⁇ 5-[(4-methylbenzenesulfonyl)oxy]pentyl ⁇ oxy)propoxy]acetic acid (L-1)
  • Step 2 Synthesis of (2S,4R)-1-[(2S)-3,3-dimethyl-2- ⁇ 2-[3-( ⁇ 5-[(4- methylbenzenesulfonyl)oxy]pentyl ⁇ oxy)propoxy]acetamido ⁇ butanoyl]-4-hydroxy-N- ⁇ [4-(4- methyl-1,3-thiazol-5-yl)phenyl]methyl ⁇ pyrrolidine-2-carboxamide (BI)
  • Step 3 Synthesis of (2S,4R)-1-[(2S)-2-[2-(3- ⁇ [5-(4- ⁇ 3-[6-cyano-5- (trifluoromethyl)pyridin-3-yl]-5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1- yl ⁇ phenoxy)pentyl]oxy ⁇ propoxy)acetamido]-3,3-dimethylbutanoyl]-4-hydroxy-N- ⁇ [4-(4-methyl- 1,3-thiazol-5-yl)phenyl]methyl ⁇ pyrrolidine-2-carboxamide (Example 2)
  • Example # compound 1 (Table 2) is also referred to as Example 1.
  • Example 54 (2S,4R)-1-((S)-2-(2-(6-(4-(3-(4-cyano-3-(trifluoromethyl)phenyl)-5,5- dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)phenoxy)hexa-2,4-diynyloxy)acetamido)-3,3- dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2- carboxamide:
  • Step 1 Synthesis of tert-butyl 2- ⁇ [6-(4- ⁇ 3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl ⁇ phenoxy)hexa-2,4-diyn-1-yl]oxy ⁇ acetate (BJ)
  • Step 2 Synthesis of 2- ⁇ [6-(4- ⁇ 3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5-dimethyl-4- oxo-2-sulfanylideneimidazolidin-1-yl ⁇ phenoxy)hexa-2,4-diyn-1-yl]oxy ⁇ acetic acid (BK)
  • Step 3 Synthesis of (2S,4R)-1-[(2S)-2-(2- ⁇ [6-(4- ⁇ 3-[4-cyano-3- (trifluoromethyl)phenyl]-5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl ⁇ phenoxy)hexa- 2,4-diyn-1-yl]oxy ⁇ acetamido)-3,3-dimethylbutanoyl]-4-hydroxy-N- ⁇ [4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl ⁇ pyrrolidine-2 ⁇ carboxamide (Example 54) [0542] This material was synthesized according to a similar procedure described in reaction step 3 for the synthesis of Example 1.
  • Example 62 (2S,4R)-1-((S)-2-tert-butyl-16-(4-(3-(4-cyano-3- (trifluoromethyl)phenyl)-5,5-dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)phenyl)-4,13-dioxo- 6,9-dioxa-3,12-diazahexadecane)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine- 2-carboxamide:
  • Step 1 Synthesis of ethyl 2-(2- ⁇ 2-[4-(4- ⁇ 3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl ⁇ phenyl)butanamido]ethoxy ⁇ ethoxy)acetate (BL)
  • Step 2 Synthesis of 2-(2- ⁇ 2-[4-(4- ⁇ 3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5-dimethyl- 4-oxo-2-sulfanylideneimidazolidin-1-yl ⁇ phenyl)butanamido]ethoxy ⁇ ethoxy)acetic acid (BM)
  • Step 3 Synthesis of (2S,4R)-1-[(2S)-2-[2-(2- ⁇ 2-[4-(4- ⁇ 3-[4-cyano-3- (trifluoromethyl)phenyl]-5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1- yl ⁇ phenyl)butanamido]ethoxy ⁇ ethoxy)acetamido]-3,3-dimethylbutanoyl]-4-hydroxy-N- ⁇ [4-(4- methyl-1,3-thiazol-5-yl)phenyl]methyl ⁇ pyrrolidine-2-carboxamide (Example 62)
  • Examples 63-65 were synthesized according to similar procedure described for synthesis of example 62, by using corresponding starting materials and intermediates.
  • Example 66 2-(2-(4'-(3-(4-cyano-3-(trifluoromethyl)phenyl)-5,5-dimethyl-4-oxo-2- thioxoimidazolidin-1-yl)biphenyl-4-yloxy)ethoxy)ethyl (S)-1-((2S,4R)-4-hydroxy-2-(4-(4- methylthiazol-5-yl)benzylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2- ylcarbamate:
  • Step 1 Synthesis of 4-[3-(4- ⁇ 4-[2-(2-hydroxyethoxy)ethoxy]phenyl ⁇ phenyl)-4,4- dimethyl-5-oxo-2--ulfanylideneimidazolidin-1-yl]-2-(trifluoromethyl)benzonitrile (BN)
  • Step 2 synthesis of 2- ⁇ 2-[4-(4- ⁇ 3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5-dimethyl-4- oxo-2-sulfanylideneimidazolidin-1-yl ⁇ phenyl)phenoxy]ethoxy ⁇ ethyl N-[(2S)-1-[(2S,4R)-4- hydroxy-2-( ⁇ [4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl ⁇ carbamoyl)pyrrolidin-1-yl]-3,3- dimethyl-1-oxobutan-2-yl]carbamate (Example 66)
  • Example 66 Yield: 6% as a white solid.
  • Step 1 Synthesis of ethyl 2-(2- ⁇ 2-[4-(4- ⁇ 3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl ⁇ phenyl)phenoxy]ethoxy ⁇ ethoxy)acetate (BO)
  • Step 2 Synthesis of 2-(2- ⁇ 2-[4-(4- ⁇ 3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5-dimethyl- 4-oxo-2-sulfanylideneimidazolidin-1-yl ⁇ phenyl)phenoxy]ethoxy ⁇ ethoxy)acetic acid (BP)
  • Step 3 Synthesis of (2S,4R)-1-[(2S)-2-[2-(2- ⁇ 2-[4-(4- ⁇ 3-[4-cyano-3- (trifluoromethyl)phenyl]-5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1- yl ⁇ phenyl)phenoxy]ethoxy ⁇ ethoxy)acetamido]-3,3-dimethylbutanoyl]-4-hydroxy-N- ⁇ [4-(4- methyl-1,3-thiazol-5-yl)phenyl]methyl ⁇ pyrrolidine-2-carboxamide (Example 67)
  • Examples 74 and 76 were synthesized according to similar procedure described for synthesis of Example 66, by using corresponding starting materials and intermediates.
  • Examples 68-73, 75, 77-79 were synthesized according to similar procedure described for synthesis of Example 67, by using corresponding starting materials and intermediates.
  • Example 80 (2S,4R)-1-((S)-2-(2-(3-(2-(4-(4-(3-(4-cyano-3-(trifluoromethyl)phenyl)- 5,5-dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)phenyl)piperidin-1- yl)ethoxy)propoxy)acetamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5- yl)benzyl)pyrrolidine-2-carboxamide:
  • Step 1 synthesis of (2S,4R)-1-[(2S)-3,3-dimethyl-2-[2-(3- ⁇ 2-[(4-methylbenzenesulfonyl) oxy]ethoxy ⁇ propoxy)acetamido]butanoyl]-4-hydroxy-N- ⁇ [4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl ⁇ pyrrolidine-2-carboxamide (BQ)
  • Step 2 Synthesis of (2S,4R)-1-[(2S)-2-[2-(3- ⁇ 2-[4-(4- ⁇ 3-[4-cyano-3- (trifluoromethyl)phenyl]-5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl ⁇ phenyl)piperidin- 1-yl]ethoxy ⁇ propoxy)acetamido]-3,3-dimethylbutanoyl]-4-hydroxy-N- ⁇ [4-(4-methyl-1,3-thiazol- 5-yl)phenyl]methyl ⁇ pyrrolidine-2-carboxamide (Exampl 80)
  • Example 80 Yield: 7% as a white solid.
  • Example 81 was synthesized according to similar procedure described for synthesis of Example 80, by using corresponding starting materials and intermediates.
  • Example 81 (2S,4R)-1-((S)-2-(2-(4-(2-(4-(4-(4-(3-(4-cyano-3-(trifluoromethyl)phenyl)- 5,5-dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)phenyl)piperidin-1- yl)ethoxy)butoxy)acetamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5- yl)benzyl)pyrrolidine-2-carboxamide
  • Example 82 (2S,4R)-N-(2-(2-(2-(2-(2-(4-(3-(4-cyano-3-(trifluoromethyl)phenyl)-5,5- dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)phenoxy)ethoxy)ethoxy)-4-(4- methylthiazol-5-yl)benzyl)-4-hydroxy-1-((S)-3-methyl-2-(1-oxoisoindolin-2- yl)butanoyl)pyrrolidine-2-carboxamide:
  • Step 1 Synthesis of 4-[3-(4- ⁇ 2-[2-(2-hydroxyethoxy)ethoxy]ethoxy ⁇ phenyl)-4,4- dimethyl-5-oxo-2--ulfanylideneimidazolidin-1-yl]-2-(trifluoromethyl)benzonitrile (BR)
  • Step 2 Synthesis of 2- ⁇ 2-[2-(4- ⁇ 3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5-dimethyl-4- oxo-2-sulfanylideneimidazolidin-1-yl ⁇ phenoxy)ethoxy]ethoxy ⁇ ethyl 4-methylbenzene-1- sulfonate (BS)
  • Step 3 Synthesis of (2S,4R)-4-(tert-butoxy)-N- ⁇ [2-(2- ⁇ 2-[2-(4- ⁇ 3-[4-cyano-3- (trifluoromethyl)phenyl]-5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1- yl ⁇ phenoxy)ethoxy]ethoxy ⁇ ethoxy)-4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl ⁇ -1-[(2S)-3- methyl-2-(1-oxo-2,3-dihydro-1H-isoindol-2-yl)butanoyl]pyrrolidine-2-carboxamide (BT)
  • Step 4 Synthesis of (2S,4R)-N- ⁇ [2-(2- ⁇ 2-[2-(4- ⁇ 3-[4-cyano-3-(trifluoromethyl)phenyl]- 5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl ⁇ phenoxy)ethoxy]ethoxy ⁇ ethoxy)-4-(4- methyl-1,3-thiazol-5-yl)phenyl]methyl ⁇ -4-hydroxy-1-[(2S)-3-methyl-2-(1-oxo-2,3-dihydro-1H- isoindol-2-yl)butanoyl]pyrrolidine-2-carboxamide (Example 82)

Abstract

The present disclosure relates to bifunctional compounds, which find utility to degrade (and inhibit) Androgen Receptor. In particular, the present invention is directed to compounds, which contain on one end a VHL ligand which binds to the ubiquitin ligase and on the other end a moiety which binds Androgen Receptor such that Androgen Receptor is placed in proximity to the ubiquitin ligase to effect degradation (and inhibition) of Androgen Receptor. The present invention exhibits a broad range of pharmacological activities associated with compounds according to the present invention, consistent with the degradation/inhibition of Androgen Receptor.

Description

COMPOUNDS AND METHODS FOR THE TARGETED DEGRADATION OF
ANDROGEN RECEPTOR CROSS-REFERENCE TO RELATED APPLICATIONS
[001] The present disclosure is an International Patent Application claiming priority to U.S. Patent Application Serial No.15/663,273, which is a Continuation-In-Part of U.S.
Nonprovisional Application Serial No.15/002,303, filed 20 January 2016, which claims the benefit of, and priority to, U.S. Provisional Patent Application Serial No.62/105,210, filed 20 January 2015 and entitled: Compounds and Methods for the Targeted Degradation of the Androgen Receptor, all of which are incorporated herein by reference in their entireties. BACKGROUND
[002] 1. Field of the Discovery. The present description relates to bifunctional compounds, which are useful for the modifying the ubiquitination and subsequent degradation of target polypeptides and proteins, in particular, androgen receptor. In certain aspects, the compounds comprise a Von Hippel-Lindau (VHL) binding moiety, which binds to the VHL E3 ubiquitin ligase, a target protein binding moiety, which binds to the target protein (e.g., androgen receptor), and optionally a linker moiety which links the VHL binding moiety and target protein binding moiety.These compounds work in such way that the target protein/polypeptide is placed in proximity to the ubiquitin ligase to effect degradation (and inhibition) of that protein (e.g., androgen receptor).
[003] 2. Background Information. Androgen Receptor (AR) belongs to a nuclear hormone receptor family that is activated by androgens, such as testosterone and
dihydrotestosterone (Pharmacol. Rev.2006, 58(4), 782-97; Vitam. Horm.1999, 55:309-52.). In the absence of androgens, AR is bound by Heat Shock Protein 90 (Hsp90) in the cytosol. When an androgen binds AR, its conformation changes to release AR from Hsp90 and to expose the Nuclear Localization Signal (NLS). The latter enables AR to translocate into the nucleus where AR acts as a transcription factor to promote gene expression responsible for male sexual characteristics (Endocr. Rev.1987, 8(1):1-28; Mol. Endocrinol.2002, 16(10), 2181-7). AR deficiency leads to Androgen Insensitivity Syndrome, formerly termed testicular feminization. [004] While AR is responsible for development of male sexual characteristics, it is also a well-documented oncogene in certain forms cancers including prostate cancers (Endocr. Rev. 2004, 25(2), 276-308). A commonly measured target gene of AR activity is the secreted Prostate Specific Antigen (PSA) protein. The current treatment regimen for prostate cancer involves inhibiting the androgen-AR axis by two methods. The first approach relies on reduction of androgens, while the second strategy aims to inhibit AR function (Nat. Rev. Drug Discovery, 2013, 12,823–824). Despite the development of effective targeted therapies, most patients develop resistance and the disease progresses. An alternative approach for the treatment of prostate cancer involves eliminating the AR protein. Because AR is a critical driver of tumorigenesis in many forms of prostate cancers, its elimination should lead to therapeutically benefical response.
[005] There exists an ongoing need in the art for effective treatments for diseases and conditions that are related to aberrant AR regulation or activity, such as, for example, cancer, prostate cancer, and Kennedy’s Disease. SUMMARY
[006] The present disclosure describes compounds, including compositions comprising the same, which function to recruit endogenous proteins to an E3 ubiquitin ligase, e.g., Von Hippel-Lindau (VHL) E3 ubiquitin ligase, for ubiquitination and subsequent degradation, and methods of using the same. In particular, the present disclosure provides bifunctional or proteolysis targeting chimeric (PROTAC) compounds, which find utility as modulators of targeted ubiquitination and degradation of androgen receptor (AR). In addition, the description provides methods of using an effective amount of the compounds as described herein for the treatment or amelioration of a disease condition including cancer, e.g., prostate cancer, and Kennedy’s Disease.
[007] Thus, in one aspect, the disclosure provides compounds which function to recruit endogenous proteins, e.g., AR proteins, to E3 Ubiquitin Ligase for ubiquintination and degradation. In certain embodiments, the compounds have the following general structure:
[008] ABM– L-ULM (I),
[009] wherein ABM is an AR binding moiety, ULM is an E3 ligase binding moiety, e.g., a VHL E3 ligase binding moiety (VLM), and L is a bond or a linker moiety which links the ABM and ULM. As such, in certain embodiments, the description provides compounds having the following general structure:
[010] ABM– L-VLM (II),
[011] wherein ABM is an AR binding moiety, VLM is a VHL E3 ligase binding moiety and L is a bond or a linker moiety which links the ABM and VLM. In certain embodiments, the VLM comprises a hydroxyl prolyl moiety.
[012] In certain embodiments, the ULM is a moiety specific for an E3 ubiquitin ligase such as, e.g., cereblon, mouse double minute 2 homolog (Mdm2), or inhibitor of apoptosis (IAP), wherein the ULM moiety is coupled to an ABM as described herein.
[013] It will be understood that the general structures are exemplary and the respective moieties can be arranged spatially in any desired order or configuration, e.g., ULM-L-ABM, and VLM-L-ABM respectively.
[014] In another aspect, the description provides AR binding moieties (ABM). In an additional embodiment, the description provides compounds having the following general structure: ABM-L, wherein ABM is an AR binding moiety as described herein, and L is a chemical linker moiety, or optionally a bond. In certain embodiments, the ABM and/or L are coupled to a ULM as described herein.
[015] In any of the aspects or embodiments described herein, the ABM is selected from following structures:
Figure imgf000004_0001
wherein W1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CN, C^CH, CF3, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
Y1, Y2 are each independently NRY1, O, S;
Y3, Y4, Y5 are each independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2; Q is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, optionally substituted with 0-6 RQ, each RQ is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
R1, R2, Ra, Rb, RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or R1, R2 together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms;
W2 is a bond, C1-6 alkyl, alicyclic (e.g., C1-6 alicyclic), heterocyclic, aryl, heteroaryl, bicyclic, biaryl, biheteroaryl, biheterocyclic, each optionally substituted by 1, 2 or 3 RW2; and each RW2 is independently H, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1-3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN.
[016] In any of the aspects or embodiments described herein, the ABM can comprise or consist of a structure as set forth herein, in particular in any of the ABMs as provided in
Examples 1-870.
[017] In certain embodiments, the ULM (derivatized or configured to be linked or coupled to an ABM via a linker (as indicated by the dashed line)) has the structure,
Figure imgf000005_0001
wherein, W3 is optionally substituted aryl, optionally substituted heteroaryl, or
Figure imgf000006_0001
each R9 and R10 is independently hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted hydroxyalkyl, optionally substituted heteroaryl, or haloalkyl; or R9, R10, and the carbon atom to which they are attached form an optionally substituted cycloalkyl;
R11 is optionally substituted heterocyclic, optionally substituted alkoxy, optionally
substituted heteroaryl, optionally substituted aryl,
Figure imgf000006_0002
or
Figure imgf000006_0003
R12 is H or optionally substituted alkyl;
R13 is H, optionally substituted alkyl, optionally substituted alkylcarbonyl, optionally substituted (cycloalkyl)alkylcarbonyl, optionally substituted aralkylcarbonyl, optionally substituted arylcarbonyl, optionally substituted (heterocyclyl)carbonyl, or optionally substituted aralkyl;
R14a, R14b, is each independently H, haloalkyl, or optionally substituted alkyl;
W5 is a phenyl or a 5-10 membered heteroaryl,
R15 is H, halogen, CN, OH, NO2, N R14aR14b, OR14a, CONR14aR14b, NR14aCOR14b, SO2NR14aR14b, NR14a SO2R14b, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted haloalkoxy, optionally substituted aryl, optionally substituted heteroaryl,
optionally substituted cycloalkyl, optionally substituted cycloheteroalkyl, or
Figure imgf000006_0004
wherein R17 is H, halo, optionally substituted C3-6cycloalkyl, optionally substituted C1-6alkyl, optionally substituted C1-6alkenyl, or C1-6haloalkyl; Xa is S or O; each R16 is independently halo, optionally substituted alkyl, optionally substituted haloalkyl, hydroxy, or optionally substituted haloalkoxy;
o is 0, 1, 2, 3, or 4;
each R18 is independently halo, optionally substituted alkoxy, cyano, optionally substituted alkyl, haloalkyl, haloalkoxy or a linker; and
p is 0, 1, 2, 3, or 4.
[018] In another embodiments, the ULM has the structure
wherein:
Figure imgf000007_0001
R9 is H;
R10 is isopropyl, tert-butyl, sec-butyl, cyclopentyl, or cyclohexyl;
R11 is
R12 is H;
R13 is H, optionally substituted alkyl, optionally substituted alkylcarbonyl, optionally
substituted (cycloalkyl)alkylcarbonyl, optionally substituted aralkylcarbonyl, optionally substituted arylcarbonyl, optionally substituted (heterocyclyl)carbonyl, or optionally substituted arylalkyl;
R14a is H, haloalkyl, methyl, ethyl, isopropyl, cyclopropyl, or C1-C6 alkyl (linear, branched, optionally substituted), each optionally substituted with 1 or more halo, hydroxyl, nitro, CN, C1-C6 alkyl (linear, branched, optionally substituted), or C1-C6 alkoxyl (linear, branched, optionally substituted); and R15 is
Figure imgf000008_0001
wherein R17 is H, halo, optionally substituted C3-6cycloalkyl, optionally substituted C1-6alkyl, optionally substituted C1-6alkenyl, and C1-6haloalkyl; and Xa is S or O.
[019] In certain embodiments, an androgen receptor binding moiety has a structure of
Figure imgf000008_0002
wherein:
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, CF3, hydroxyl, nitro, CN, C^CH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
Y1, Y2 are each independently NRY1, O, S;
Y3, Y4, Y5 are each independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2;
Q is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, optionally substituted with 0-6 RQ, each RQ is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl);
W2 is a bond, C1-6 alkyl, alicyclic, heterocyclic, aryl, heteroaryl, bicyclic, biaryl,
biheteroaryl,or biheterocyclic, each optionally substituted by 1, 2 or 3 RW2; and each RW2 is independently H, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1- 3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN.
[020] In certain additional embodiments, the compounds comprise a plurality of E3 ligase binding moieties and/or a plurality of ABMs. [021] In certain embodiments, the linker group (L) comprises a chemical structural unit represented by the formula:
-Aq- wherein
q is an integer greater than 1; and
A is independently selected from the group consisting of a bond, CRL1RL2, O, S, SO, SO2, NRL3, SO2NRL3, SONRL3, CONRL3, NRL3CONRL4, NRL3SO2NRL4, CO, CRL1=CRL2, C^C, SiRL1RL2, P(O)RL1, P(O)ORL1, NRL3C(=NCN)NRL4, NRL3C(=NCN),
NRL3C(=CNO2)NRL4, C3-11cycloalkyl optionally substituted with 0-6 RL1 and/or RL2 groups, C3-11heteocyclyl optionally substituted with 0-6 RL1 and/or RL2 groups, aryl optionally substituted with 0-6 RL1 and/or RL2 groups, heteroaryl optionally substituted with 0-6 RL1 and/or RL2 groups;
wherein RL1, RL2, RL3, RL4 and RL5 are each, independently, selected from the group consisting of H, halo, C1-8alkyl, OC1-8alkyl, SC1-8alkyl, NHC1-8alkyl, N(C1-8alkyl)2, C3- 11cycloalkyl, aryl, heteroaryl, C3-11heterocyclyl, OC1-8cycloalkyl, SC1-8cycloalkyl, NHC1- 8cycloalkyl, N(C1-8cycloalkyl)2, N(C1-8cycloalkyl)(C1-8alkyl), OH, NH2, SH, SO2C1-8alkyl, P(O)(OC1-8alkyl)(C1-8alkyl), P(O)(OC1-8alkyl)2, CC-C1-8alkyl, CCH, CH=CH(C1-8alkyl), C(C1-8alkyl)=CH(C1-8alkyl), C(C1-8alkyl)=C(C1-8alkyl)2, Si(OH)3, Si(C1-8alkyl)3, Si(OH)(C1- 8alkyl)2, COC1-8alkyl, CO2H, halogen, CN, CF3, CHF2, CH2F, NO2, SF5, SO2NHC1-8alkyl, SO2N(C1-8alkyl)2, SONHC1-8alkyl, SON(C1-8alkyl)2, CONHC1-8alkyl, CON(C1-8alkyl)2, N(C1-8alkyl)CONH(C1-8alkyl), N(C1-8alkyl)CON(C1-8alkyl)2, NHCONH(C1-8alkyl),
NHCON(C1-8alkyl)2, NHCONH2, N(C1-8alkyl)SO2NH(C1-8alkyl), N(C1-8alkyl) SO2N(C1- 8alkyl)2, NH SO2NH(C1-8alkyl), NH SO2N(C1-8alkyl)2, and NH SO2NH2; and
wherein when q is greater than 1, RL1 or RL2 each, independently, can be linked to another A group to form cycloalkyl and/or heterocyclyl moeity that can be further substituted with 0-4 RL5 groups.
[022] In certain embodiments, the description provides a bifunctional compound having a structure selected from the group consisting of Examples 1-870, a salt, a polymorph, and a prodrug thereof.
[023] In another aspect, the description provides compositions comprising compounds as described herein, and a pharmaceutically acceptable carrier. In certain embodiments, the compositions are therapeutic or pharmaceutical compositions comprising an effective amount of a compound as described herein and a pharmaceutally acceptable carrier. In certain
embodiments, the therapeutic or pharmaceutical compositions comprise an additional biologically active agent, e.g., an agent effective for the treatment of cancer.
[024] In any of the aspects or embodiments described herein, the therapeutic compositions comprising compounds described herein can be in any suitable dosage form, e.g., solid, or liquid, and configured to be delivered by any suitable route, e.g., oral, parenteral, intravenous, intraperitoneal, subcutaneous, intramuscular, etc., and in any desired unit dosage form. For example, in certain embodimetns, the therapeutic composition as described herein is configured to be administered or consumed by a subject one or more times over a descired time period, e.g., day, week, month, etc.
[025] In another aspect, the disclosure provides methods of modulating protein ubiquitination and degradation in a subject, e.g., a cell, a tissue, mammal, or human patient, the method comprising administering an effective amount of a compound as described herein or a composition comprising an effective amount of the same to a subject, wherein the compound or composition comprising the same is effective in modulating protein ubquitination and degration of the protiein in the subject. In certain embodiments, the protein is androgen receptor (AR).
[026] In another aspect, the disclosure provides methods of modulating AR protein ubiquitination and degradation in a subject, e.g., a cell, a tissue, mammal, or human patient, the method comprising administering an effective amount of a compound as described herein or a composition comprising an effective amount of the same to a subject, wherein the compound or composition comprising the same is effective in modulating AR protein ubquitination and degration of the protiein in the subject.
[027] In another aspect, the disclosure provides methods of treating or ameliorating a symptom of a disease related to AR activity in a subject, e.g., a cell, a tissue, mammal, or human patient, the method comprising administering an effective amount of a compound as described herein or a composition comprising an effective amount of the same to a subject in need thereof, wherein the compound or composition comprising the same is effective in treating or ameliorating a symptom of a disease related to AR activity in the subject. In certain
embodiments, the disease to be treated is cancer, e.g., prostate cancer, or Kennedy’s Disease. In a preferred embodiment, the subject is a human. [028] In another aspect, the disclosure provides methods for identifying the effects of the degradation of proteins of interest in a biological system using compounds according to the present invention.
[029] In another aspect, the description provides kits comprising compounds or compositions as described herein. The kit may be promoted, distributed, or sold as a unit for performing the methods of the present invention. In addition, the kits of the present invention may preferably contain instructions which describe a suitable use. Such kits can be conveniently used, e.g., in clinical settings, to treat patients exhibiting symptoms of, e.g., cancer or Kennedy’s Disease.
[030] Where applicable or not specifically disclaimed, any one of the embodiments described herein are contemplated to be able to combine with any other one or more
embodiments, even though the embodiments are described under different aspects of the invention. As such, the preceding general areas of utility are given by way of example only and are not intended to be limiting on the scope of the present disclosure and appended claims.
Additional objects and advantages associated with the compositions, methods, and processes of the present invention will be appreciated by one of ordinary skill in the art in light of the instant claims, description, and examples. For example, the various aspects and embodiments of the invention may be utilized in numerous combinations, all of which are expressly contemplated by the present description. These additional advantages objects and embodiments are expressly included within the scope of the present invention. The publications and other materials used herein to illuminate the background of the invention, and in particular cases, to provide additional details respecting the practice, are incorporated by reference. BRIEF DESCRIPTION OF THE DRAWINGS
[031] The accompanying drawings, which are incorporated into and form a part of the specification, illustrate several embodiments of the present invention and, together with the description, serve to explain the principles of the invention. The drawings are only for the purpose of illustrating an embodiment of the invention and are not to be construed as limiting the invention. Further objects, features and advantages of the invention will become apparent from the following detailed description taken in conjunction with the accompanying figures showing illustrative embodiments of the invention, in which: [032] Figure 1A and Figure 1B. Illustration of general principle for PROTAC function. Figure 1A: Exemplary PROTACs comprise an androgen receptor targeting moiety (ABM; darkly shaded rectangle), a Von Hippel-Lindau (VHL) E3 ubiquitin ligase binding moiety (VLM; lightly shaded triangle), and a linker moiety (L; black line) coupling or tethering the ABM to the VLM (as described herein, L can be absent or a bond or a chemical linker moiety). Figure 1B Illustrates the functional use of the PROTACs as described herein. Briefly, the VLM recognizes and binds to Von Hippel-Lindau (VHL) E3 ubiquitin ligase, and the ABM binds and recruits androgen receptor and brings it into close proximity to the Von Hippel-Lindau (VHL) E3 ubiquitin ligase. Typically, the E3 ubiquitin ligase is complexed with an E2 ubiquitin- conjugating protein, and either alone or via the E2 protein catalyzes attachment of ubiquitin (dark circles) to a lysine on the target protein via an isopeptide bond. The poly-ubiquitinated protein (far right) is then targeted for degration by the proteosomal machinery of the cell.
[033] Figure 2. Apoptosis in VCaP cells. VCaP cells were cultured in Charcoal Stripped Serum containing media supplemented with 0.1 nM R1881 for 48 hrs. The degree of apoptosis was ascertained with CaspaseGlo assay (Promega). These results demonstrated that PROTACs are much more potent in inducing apoptosis than an AR antagonist enzalutamide. Further, the degree of AR degradation correlates with their ability to induce apoptosis in VCaP cells.
[034] Figure 3. Anti-proliferation in LNCaP F876L. Anti-proliferation in LNCaP F876L cells observed with treatment with Example 1 as compared to enzalutamide. LNCaP cells transduced with AR F876L construct were cultured in Charcoal Stripped Serum containing media.
[035] Figure 4. PSA suppression in LNCaP F876L. LNCaP cells transduced with AR F876L construct were cultured in Charcoal Stripped Serum containing media supplemented with 0.1 nM R1881 for 7 days. The results demonstrated that AR PROTAC is able to suppress the transcriptional activity of AR in F876L containing cells.
[036] Figure 5. Prostate involution in C57B6 mouse model. 12-week old male C57BL/6 mice were treated with AR PROTAC Example 163 and its inactive epimer analog Compound A which is unable to bind to VHL E3 ligase. Enzalutamide (PO, QD, 30 mpk), Example 163 (IP, QD, 1 and 3 mpk) and Compound A (IP, QD, 1 and 3 mpk) were administered for 10 days, upon which the prostates were isolated and weighed. These results demonstrated that the ability of PROTAC Example 163 to degrade AR leads to significant prostate involution in mice at very low doses.
[037] Figure 6. Tumor growth inhibition in VCaP xenograft model. VCaP cells were implanted into CB17 scid mice subcutaneously. Once the tumors were palpable, the mice were castrated, leading to temporary tumor stasis. Upon regrowth of tumors, the mice were dosed with enzalutamide (PO, QD, 30 mpk) or AR PROTAC Example 163 (IP, QD, at 30, 10 and 3 mpk) as indicated. Tumor growth inhibition was observed in all treatment arms.
[038] Figure 7A and Figure 7B. AR degradation of PROTAC is E3 ligase dependent. Figure 7A: AR PROTAC Example 1 was added to LNCaP cells at indicated concentrations for 24 hours in the presence or absence of 10 uM VHL E3 ligase ligand compound B. Figure 7B: LNCaP cells were treated with AR PROTAC Example 1 and its inactive epimer analog compound C which is unable to bind to VHL E3 ligase.” DETAILED DESCRIPTION
[039] The following is a detailed description provided to aid those skilled in the art in practicing the present invention. Those of ordinary skill in the art may make modifications and variations in the embodiments described herein without departing from the spirit or scope of the present disclosure. All publications, patent applications, patents, figures and other references mentioned herein are expressly incorporated by reference in their entirety.
[040] The present description relates to the surprising and unexpected discovery that an E3 ubiquitin ligase protein can ubiquitinate a target protein, in particular androgen receptor, once the E3 ubiquitin ligase protein and the target protein are brought into proximity by a chimeric construct (e.g., PROTAC) as described herein, in which a moiety that binds the E3 ubiquitin ligase protein is coupled, e.g., covalently, to a moiety that bind the androgen receptortarget protein. Accordingly, the present description provides compounds, compositions comprising the same, and associated methods of use for ubiquitination and degradation of a chosen target protein, e.g., androgen receptor (See Figure 1A and Figure 1B).
[041] The present description is related in certain aspects to U.S. Patent Publication 2014/0356322A1, which is incorporated herein by reference in its entirety for all purposes.
[042] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. The terminology used in the description is for describing particular embodiments only and is not intended to be limiting of the invention.
[043] Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise (such as in the case of a group containing a number of carbon atoms in which case each carbon atom number falling within the range is provided), between the upper and lower limit of that range and any other stated or intervening value in that stated range is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges is also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either both of those included limits are also included in the invention.
[044] The following terms are used to describe the present invention. In instances where a term is not specifically defined herein, that term is given an art-recognized meaning by those of ordinary skill applying that term in context to its use in describing the present invention.
[045] The articles "a" and "an" as used herein and in the appended claims are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article unless the context clearly indicates otherwise. By way of example, "an element" means one element or more than one element.
[046] The phrase "and/or," as used herein in the specification and in the claims, should be understood to mean "either or both" of the elements so conjoined, i.e., elements that are conjunctively present in some cases and disjunctively present in other cases. Multiple elements listed with "and/or" should be construed in the same fashion, i.e., "one or more" of the elements so conjoined. Other elements may optionally be present other than the elements specifically identified by the "and/or" clause, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, a reference to "A and/or B", when used in conjunction with open-ended language such as "comprising" can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.
[047] As used herein in the specification and in the claims, "or" should be understood to have the same meaning as "and/or" as defined above. For example, when separating items in a list, "or" or "and/or" shall be interpreted as being inclusive, i.e., the inclusion of at least one, but also including more than one, of a number or list of elements, and, optionally, additional unlisted items. Only terms clearly indicated to the contrary, such as "only one of' or "exactly one of," or, when used in the claims, "consisting of," will refer to the inclusion of exactly one element of a number or list of elements. In general, the term "or" as used herein shall only be interpreted as indicating exclusive alternatives (i.e., "one or the other but not both") when preceded by terms of exclusivity, such as "either," "one of," "only one of," or "exactly one of."
[048] The term“about” and the like, as used herein, in association with numeric values or ranges, reflects the fact that there is a certain level of variation that is recognized and tolerated in the art due to practical and/or theoretical limitations. For example, minor variation is tolerated due to inherent variances in the manner in which certain devices operate and/or measurements are taken. In accordance with the above, the phrase“about” is normally used to encompass values within the standard deviation or standard error.
[049] In the claims, as well as in the specification above, all transitional phrases such as "comprising," "including," "carrying," "having," "containing," "involving," "holding,"
"composed of," and the like are to be understood to be open-ended, i.e., to mean including but not limited to. Only the transitional phrases "consisting of' and "consisting essentially of' shall be closed or semi-closed transitional phrases, respectively, as set forth in the United States Patent Office Manual of Patent Examining Procedures, Section 2111.03.
[050] As used herein in the specification and in the claims, the phrase "at least one," in reference to a list of one or more elements, should be understood to mean at least one element selected from anyone or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements. This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase "at least one" refers, whether related or unrelated to those elements specifically identified. Thus, as a nonlimiting example, "at least one of A and B" (or, equivalently, "at least one of A or B," or, equivalently "at least one of A and/or B") can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.
[051] It should also be understood that, in certain methods described herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited unless the context indicates otherwise.
[052] The terms "co-administration" and "co-administering" or“combination therapy” can refer to both concurrent administration (administration of two or more therapeutic agents at the same time) and time varied administration (administration of one or more therapeutic agents at a time different from that of the administration of an additional therapeutic agent or agents), as long as the therapeutic agents are present in the patient to some extent, preferably at effective amounts, at the same time. In certain preferred aspects, one or more of the present compounds described herein, are coadministered in combination with at least one additional bioactive agent, especially including an anticancer agent. In particularly preferred aspects, the co-administration of compounds results in synergistic activity and/or therapy, including anticancer activity.
[053] The term“effective” can mean, but is in no way limited to, that amount/dose of the active pharmaceutical ingredient, which, when used in the context of its intended use, effectuates or is sufficient to prevent, inhibit the occurrence, ameliorate, delay or treat (alleviate a symptom to some extent, preferably all) the symptoms of a condition, disorder or disease state in a subject in need of such treatment or receiving such treatment. The term effective subsumes all other effective amount or effective concentration terms, e.g.,“effective amount/dose,” “pharmaceutically effective amount/dose” or“therapeutically effective amount/dose,” which are otherwise described or used in the present application.
[054] The effective amount depends on the type and severity of disease, the
composition used, the route of administration, the type of mammal being treated, the physical characteristics of the specific mammal under consideration, concurrent medication, and other factors which those skilled in the medical arts will recognize. The exact amount can be ascertainable by one skilled in the art using known techniques (see, e.g., Lieberman,
Pharmaceutical Dosage Forms (vols. 1-3, 1992); Lloyd, The Art, Science and Technology of Pharmaceutical Compounding (1999); Pickar, Dosage Calculations (1999); and Remington: The Science and Practice of Pharmacy, 20th Edition, 2003, Gennaro, Ed., Lippincott, Williams & Wilkins).
[055] The term“pharmacological composition,”“therapeutic composition,”
“therapeutic formulation” or“pharmaceutically acceptable formulation” can mean, but is in no way limited to, a composition or formulation that allows for the effective distribution of an agent provided by the invention, which is in a form suitable for administration to the physical location most suitable for their desired activity, e.g., systemic administration.
[056] The term "pharmaceutically acceptable" or“pharmacologically acceptable” can mean, but is in no way limited to, entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal, or a human, as appropriate.
[057] The term "pharmaceutically acceptable carrier" or“pharmacologically acceptable carrier” can mean, but is in no way limited to, any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Suitable carriers are described in the most recent edition of Remington's Pharmaceutical Sciences, a standard reference text in the field, which is incorporated herein by reference. Preferred examples of such carriers or diluents include, but are not limited to, water, saline, finger's solutions, dextrose solution, and 5% human serum albumin. Liposomes and non-aqueous vehicles such as fixed oils may also be used. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.
[058] The term“systemic administration" refers to a route of administration that is, e.g., enteral or parenteral, and results in the systemic distribution of an agent leading to systemic absorption or accumulation of drugs in the blood stream followed by distribution throughout the entire body. Suitable forms, in part, depend upon the use or the route of entry, for example oral, transdermal, or by injection. Such forms should not prevent the composition or formulation from reaching a target cell (i.e., a cell to which the negatively charged polymer is desired to be delivered to). For example, pharmacological compositions injected into the blood stream should be soluble. Other factors are known in the art, and include considerations such as toxicity and forms which prevent the composition or formulation from exerting its effect. Administration routes which lead to systemic absorption include, without limitations: intravenous, subcutaneous, intraperitoneal, inhalation, oral, intrapulmonary and intramuscular. The rate of entry of a drug into the circulation has been shown to be a function of molecular weight or size. The use of a liposome or other drug carrier comprising the compounds of the instant invention can potentially localize the drug, for example, in certain tissue types, such as the tissues of the reticular endothelial system (RES). A liposome formulation which can facilitate the association of drug with the surface of cells, such as, lymphocytes and macrophages is also useful.
[059] The term“local administration" refers to a route of administration in which the agent is delivered to a site that is apposite or proximal, e.g., within about 10 cm, to the site of the lesion or disease.
[060] The term“compound”, as used herein, unless otherwise indicated, refers to any specific chemical compound disclosed herein and includes tautomers, regioisomers, geometric isomers, and where applicable, stereoisomers, including optical isomers (enantiomers) and other steroisomers (diastereomers) thereof, as well as pharmaceutically acceptable salts and derivatives (including prodrug forms) thereof where applicable, in context. Within its use in context, the term compound generally refers to a single compound, but also may include other compounds such as stereoisomers, regioisomers and/or optical isomers (including racemic mixtures) as well as specific enantiomers or enantiomerically enriched mixtures of disclosed compounds. The term also refers, in context to prodrug forms of compounds which have been modified to facilitate the administration and delivery of compounds to a site of activity. It is noted that in describing the present compounds, numerous substituents and variables associated with same, among others, are described.
[061] It is understood by those of ordinary skill that molecules which are described herein are stable compounds as generally described hereunder. When the bond is
Figure imgf000018_0001
shown, both a double bond and single bond are represented within the context of the compound shown.
[062] As used herein, "derivatives" can mean compositions formed from the native compounds either directly, by modification, or by partial substitution. As used herein, "analogs" can mean compositions that have a structure similar to, but not identical to, the native compound. [063] The term“Ubiquitin Ligase” refers to a family of proteins that facilitate the transfer of ubiquitin to a specific substrate protein, targeting the substrate protein for
degradation. By way of example, Von Hippel-Lindau E3 Ubiquitin Ligase or VCB E3 Ubiquitin Ligase is protein that alone or in combination with an E2 ubiquitin-conjugating enzyme causes the attachment of ubiquitin to a lysine on a target protein, and subsequently targets the specific protein substrates for degradation by the proteasome. Thus, E3 ubiquitin ligase alone or in complex with an E2 ubiquitin conjugating enzyme is responsible for the transfer of ubiquitin to targeted proteins. In general, the ubiquitin ligase is involved in polyubiquitination such that a second ubiquitin is attached to the first; a third is attached to the second, and so forth.
Polyubiquitination marks proteins for degradation by the proteasome. However, there are some ubiquitination events that are limited to mono-ubiquitination, in which only a single ubiquitin is added by the ubiquitin ligase to a substrate molecule. Mono-ubiquitinated proteins are not targeted to the proteasome for degradation, but may instead be altered in their cellular location or function, for example, via binding other proteins that have domains capable of binding ubiquitin. Further complicating matters, different lysines on ubiquitin can be targeted by an E3 to make chains. The most common lysine is Lys48 on the ubiquitin chain. This is the lysine used to make polyubiquitin, which is recognized by the proteasome.
[064] The term“subject” is used throughout the specification to describe a cell, tissue, or animal, preferably a mammal, e.g., a human or a domesticated animal, to whom treatment, including prophylactic treatment, with the compositions according to the present invention is provided. For treatment of those infections, conditions or disease states which are specific for a specific animal such as a human patient, the term patient refers to that specific animal, including a domesticated animal such as a dog or cat or a farm animal such as a horse, cow, sheep, etc. In general, in the present invention, the term patient refers to a human patient unless otherwise stated or implied from the context of the use of the term.
[065] Compounds
[066] In one aspect, the present invention provides compounds useful for regulating protein activity. The composition comprises a ubiquitin pathway protein binding moiety
(preferably for an E3 ubiquitin ligase, alone or in complex with an E2 ubiquitin conjugating enzyme which is responsible for the transfer of ubiquitin to targeted proteins) according to a defined chemical structure and a protein targeting moiety which are linked or coupled together, preferably through a linker, wherein the ubiquitin pathway protein binding moiety recognizes an ubiquitin pathway protein and the targeting moiety recognizes a target protein (e.g., androgen receptor). Such compounds may be referred to herein as PROTAC compounds or PROTACs.
[067] In one aspect, the description provides AR binding moieties (ABM). In certain embodiments, the compounds having the following general structure: ABM-L, wherein ABM is an AR binding moiety as described herein, and L is a chemical linker moiety, e.g., a linker as described herein, or optionally a bond. In certain embodiments, the ABM and/or L are coupled to a ULM as described herein below.
[068] In another aspect, the disclosure provides compounds which function to recruit androgen receptor (AR) proteins to E3 Ubiquitin Ligase for ubiquintination and degradation. In certain embodiments, the compounds have the following general structure:
[069] ABM–L–ULM (I),
[070] wherein ULM is an E3 ligase binding moiety, ABM is an AR binding moiety, which binds to an AR protein and L is a bond or a chemical linker moiety which links the ABM and ULM.
[071] In certain embodiments, the ULM is a moiety specific for an E3 ubiquitin ligase such as, e.g., Von Hippel-Lindau E3 ubiquitin ligase (VHL), cereblon, mouse double minute 2 homolog (Mdm2), or inhibitor of apoptosis (IAP), wherein the ULM moiety is coupled to an ABM as described herein.
[072] Without being bound by any particular theory, it is hypothesized that due at least in part to the proximity of AR and the E3 ubiquitin ligase, the AR is ubiquitinated by the ubiquitin ligase and degraded. In certain embodiments, the ABM is chemically linked or coupled directly to the ULM group. In certain additional embodiments, the ABM is chemically linked or coupled to the ULM via a chemical linker moiety. In additional embodiments, the description provides compounds having the following general structure:
[073] ABM–L–VLM (II),
[074] wherein ABM is an AR binding moiety and VLM is a Von Hippel-Lindau E3 Ubiquitin Ligase (VHL) binding moiety,and L is a bond or a chemical linker moiety which links the ABM and VLM. The ULM or VLM group and ABM group may be covalently linked to the linker group through any covalent bond which is appropriate and stable to the chemistry of the linker. [075] In certain embodiments, the ULM or VLM comprises a hydroxyprolyl moiety. The hydroxyl prolyl moiety has been shown to be importantn for binding and recruiting of the VHL protein.
[076] It will be understood that the general structures are exemplary and the respective moieties can be arranged in any desired order or configuration, e.g., ULM-L-ABM, and VLM-L- ABM respectively. In certain additional embodiments, the compounds comprise a plurality of E3 ligase binding moieties and/or a plurality of ABMs.
[077] In certain embodiments, ABM alone, without forming ABM-L-ULM, provides desired properties in regulating protein activity.
[078] In any of the aspects or embodiments of compounds described herein, unless indicated otherwise, the compounds are intended to encompass pharmaceutically acceptable salts, enantiomers, stereoisomers, solvates or polymorphs thereof.
[079] Exemplary ULMs
[080] In certain embodiments of the compounds as described herein, the ULM comprises a chemical structure selected from the group ULM-a:
wherein:
Figure imgf000021_0001
a dashed line indicates the attachment of at least one ABM, another ULM or VLM (i.e., ULM’ or VLM’), or a chemical linker moiety coupling at least one ABM, a ULM’ or VLM’ to the other end of the linker;
X1, X2 are each independently a bond, O, NRY3, CRY3RY4, C=O, C=S, SO, SO2;
RY3, RY4 are each independently H, linear or branched C1-6 alkyl, optionally substituted by 1 or more halo, optionally substituted C P
1-6 alkoxyl (e.g., optionally substituted with 0-3 R groups);
RP is 0, 1, 2, or 3, groups, each independently selected from H, halo, -OH, C1-3 alkyl, C=O; W3 is an optionally substituted–T-N(R1aR1b),–T-Aryl, an optionally substituted–T- Heteroaryl, an optionally substituted–T-Heterocycle, an optionally substituted -NR1-T- Aryl, an optionally substituted -NR1-T-Heteroaryl or an optionally substituted -NR1-T- Heterocycle, where T is covalently bonded to X1;
each R1 , R1a , R1b is independently H, a C1-C6 alkyl group (linear, branched, optionally
substituted by 1 or more halo, -OH), RY3C=O, RY3C=S, RY3SO, RY3SO2, N(RY3RY4)C=O, N(RY3RY4)C=S, N(RY3RY4)SO, N(RY3RY4)SO2;
W4 is an optionally substituted -NR1-T-Aryl, an optionally substituted -NR1-T-Heteroaryl group or an optionally substituted -NR1-T-Heterocycle, wherein -NR1 is covalently bonded to X2
; R1 is H or CH3, preferably H; and
T is an optionally substituted–(CH2)n- group, wherein each one of the methylene groups may be optionally substituted with one or two substituents, preferably selected from halogen, a C1-C6 alkyl group (linear, branched, optionally substituted by 1 or more halogen, -OH) or the sidechain of an amino acid as otherwise described herein, preferably methyl, which may be optionally substituted; and n is 0 to 6, often 0, 1, 2, or 3, preferably 0.
[081] Alternatively, T may also be a–(CH2O)n- group, a–(OCH2)n- group, a–
(CH2CH2O)n- group, a–(OCH2CH2)n- group, each of which groups is optionally substituted; and
[082] Alternatively, T is an optionally substituted–(CH2)n- group, wherein each one of the methylene groups may be optionally substituted with one or two substituents, preferably selected from halogen, an amino acid sidechain as otherwise described herein or a C1-C6 alkyl group (linear, branched, optionally substituted by 1 or more halo, -OH), preferably one or two methyl groups, which may be optionally substituted; and n is 0 to 6, often 0, 1, 2 or 3, preferably 0 or 1.
[083] Alternatively, T may also be a–(CH2O)n- group, a–(OCH2)n- group, a–
(CH2CH2O)n- group, a–(OCH2CH2)n- group, all of which groups are optionally substituted.
[084] In any of the embodiments described herein, W3 and/or W4 can be attached to a linker moiety as described herein.
[085] In certain embodiments, aryl groups for W3 include optionally substituted phenyl or naphthyl groups, preferably phenyl groups, wherein the phenyl or naphthyl group is optionally substituted with a linker group to which is attached a ABM group (including a ULM’ group) and/or a halogen (preferably F or Cl), an amine, monoalkyl- or dialkyl amine (preferably, dimethylamine), an amido group (preferably a–(CH2)m-NR1C(O)R2 group wherein m, R1 and R2 are the same as for R1), a halogen (often F or Cl), OH, CH3, CF3, OMe, OCF3, NO2, CN or a S(O)2RS group (RS is a a C1-C6 alkyl group, an optionally substituted aryl, heteroaryl or heterocycle group or a -(CH2)mNR1R2 group), each of which may be substituted in ortho-, meta- and/or para- positions of the phenyl ring, preferably para-), or an Aryl (preferably phenyl), heteroaryl or heterocycle. Preferably said substituent phenyl group is an optionally substituted phenyl group (i.e., the substituent phenyl group itself is preferably substituted with at least one of F, Cl, OH, SH, COOH, CH3, CF3, OMe, OCF3, NO2, CN or a linker group to which is attached a ABM group (including a ULM’ group), wherein the substitution occurs in ortho-, meta- and/or para- positions of the phenyl ring, preferably para-), a naphthyl group, which may be optionally substituted including as described above, an optionally substituted heteroaryl (preferably an optionally substituted isoxazole including a methylsubstituted isoxazole, an optionally substituted oxazole including a methylsubstituted oxazole, an optionally substituted thiazole including a methyl substituted thiazole, an optionally substituted pyrrole including a
methylsubstituted pyrrole, an optionally substituted imidazole including a methylimidazole, a benzylimidazole or methoxybenzylimidazole, an oximidazole or methyloximidazole, an optionally substituted diazole group, including a methyldiazole group, an optionally substituted triazole group, including a methylsubstituted triazole group, a pyridine group, including a halo- (preferably, F) or methylsubstitutedpyridine group or an oxapyridine group (wherein the pyridine group is linked to the phenyl group by an oxygen) or an optionally substituted heterocycle (tetrahydrofuran, tetrahydrothiophene, pyrrolidine, piperidine, morpholine, piperazine, tetrahydroquinoline, oxane or thiane. Each of the aryl, heteroaryl or heterocyclic groups may be optionally substituted with a linker group to which is attached a ABM group (including a ULM’ group).
[086] In certain embodiments, heteroaryl groups for W3 include an optionally substituted quinoline (which may be attached to the pharmacophore or substituted on any carbon atom within the quinoline ring), an optionally substituted indole (including dihydroindole), an optionally substituted indolizine, an optionally substituted azaindolizine (2, 3 or 4-azaindolizine) an optionally substituted benzimidazole, benzodiazole, benzoxofuran, an optionally substituted imidazole, an optionally substituted isoxazole, an optionally substituted oxazole (preferably methyl substituted), an optionally substituted diazole, an optionally substituted triazole, a tetrazole, an optionally substituted benzofuran, an optionally substituted thiophene, an optionally substituted thiazole (preferably methyl and/or thiol substituted), an optionally substituted isothiazole, an optionally substituted triazole (preferably a 1,2,3-triazole substituted with a methyl group, a triisopropylsilyl group, an optionally substituted -(CH2)m-O-C1-C6 alkyl group or an optionally substituted -(CH2)m-C(O)-O-C1-C6 alkyl group), an optionally substituted pyridine (2-, 3, or 4-pyridine) or a group according to the chemical structure:
Figure imgf000024_0001
, wherein:
Sc is CHRSS, NRURE, or O;
RHET is H, CN, NO2, halo (preferably Cl or F), optionally substituted C1-C6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups (e.g. CF3), optionally substituted O(C1-C6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted acetylenic group–C^C-Ra wherein Ra is H or a C1-C6 alkyl group (preferably C1-C3 alkyl);
RSS is H, CN, NO2, halo (preferably F or Cl), optionally substituted C1-C6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups), optionally substituted O-(C1-C6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted -C(O)(C1-C6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups);
RURE is H, a C1-C6 alkyl (preferably H or C1-C3 alkyl) or a–C(O)(C1-C6 alkyl), each of
which groups is optionally substituted with one or two hydroxyl groups or up to three halogen, preferably fluorine groups, or an optionally substituted heterocycle, for example piperidine, morpholine, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, piperidine, piperazine, each of which is optionally substituted; and
YC is N or C-RYC, wherein RYC is H, OH, CN, NO2, halo (preferably Cl or F), optionally substituted C1-C6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups (e.g. CF3), optionally substituted O(C1-C6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted acetylenic group–C^C-Ra wherein Ra is H or a C1-C6 alkyl group (preferably C1-C3 alkyl). Each of said heteroaryl groups may be optionally substituted with a linker group to which is attached a ABM group (including a ULM’ group).
[087] In additional embodiments, heterocycle groups for W3 include
tetrahydroquinoline, piperidine, piperazine, pyrrollidine, morpholine, tetrahydrofuran, tetrahydrothiophene, oxane and thiane, each of which groups may be optionally substituted or a group according to the chemical structure:
Figure imgf000025_0001
wherein:
RPRO is H, optionally substituted C1-C6 alkyl or an optionally substituted aryl (phenyl or napthyl), heteroaryl or heterocyclic group selected from the group consisting of oxazole, isoxazole, thiazole, isothiazole, imidazole, diazole, oximidazole, pyrrole, pyrollidine, furan, dihydrofuran, tetrahydrofuran, thiene, dihydrothiene, tetrahydrothiene, pyridine, piperidine, piperazine, morpholine, quinoline, (each preferably substituted with a C1-C3 alkyl group, preferably methyl or a halo group, preferably F or Cl), benzofuran, indole, indolizine, azaindolizine; RPRO1 and RPRO2 are each independently H, an optionally subsituted C1-C3 alkyl group or together form a keto group, and
each n is 0, 1, 2, 3, 4, 5, or 6 (preferably 0 or 1), wherein each of said Heteocycle groups may be optionally substituted with a linker group to which is attached a ABM group
(including a ULM’ group) or a pharmaceutically acceptable salt, stereoisomer, solvate or polymorph thereof.
[088] In certain embodiments, W3 substituents for use in the present invention also include specifically (and without limitation to the specific compound disclosed) the W3 substituents which are found in the identified compounds disclosed herein (which includes the specific compounds which are disclosed in the present specification, and the figures which are attached hereto). Each of these W3 substituents may be used in conjunction with any number of W4 substituents, which are also disclosed herein.
[089] In certain embodiments, Aryl groups for W4 include optionally substituted phenyl or naphthyl groups, preferably phenyl groups, wherein the phenyl group is optionally substituted with a linker group to which is attached an ABM group (including a ULM’ group), a halogen (preferably F or Cl), an amine, monoalkyl- or dialkyl amine (preferably, dimethylamine), F, Cl, OH, COOH, C1-C6 alkyl, preferably CH3, CF3, OMe, OCF3, NO2, or CN group (each of which may be substituted in ortho-, meta- and/or para- positions of the phenyl ring, preferably para-), an optionally substituted phenyl group (the phenyl group itself is preferably substituted with a linker group attached to a ABM group, including a ULM’ group), and/or at least one of F, Cl, OH, COOH, CH3, CF3, OMe, OCF3, NO2, or CN group (in ortho-, meta- and/or para- positions of the phenyl ring, preferably para-), a naphthyl group, which may be optionally substituted, an optionally substituted heteroaryl, preferably an optionally substituted isoxazole including a methylsubstituted isoxazole, an optionally substituted oxazole including a methylsubstituted oxazole, an optionally substituted thiazole including a methyl substituted thiazole, an optionally substituted isothiazole including a methyl substituted isothiazole, an optionally substituted pyrrole including a methylsubstituted pyrrole, an optionally substituted imidazole including a methylimidazole, an optionally substituted benzimidazole or methoxybenzylimidazole, an optionally substituted oximidazole or methyloximidazole, an optionally substituted diazole group, including a methyldiazole group, an optionally substituted triazole group, including a methylsubstituted triazole group, an optionally substituted pyridine group, including a halo- (preferably, F) or methylsubstitutedpyridine group or an oxapyridine group (wherein the pyridine group is linked to the phenyl group by an oxygen), an optionally substituted furan, an optionally substituted benzofuran, an optionally substituted dihydrobenzofuran, an optionally substituted indole, indolizine or azaindolizine (2, 3, or 4-azaindolizine), an optionally substituted quinoline, an optionally substituted group according to the chemical structure:
Figure imgf000027_0001
, wherein:
Sc is CHRSS, NRURE, or O;
RHET is H, CN, NO2, halo (preferably Cl or F), optionally substituted C1-C6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups (e.g. CF3), optionally substituted O(C1-C6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted acetylenic group–C^C-Ra wherein Ra is H or a C1-C6 alkyl group (preferably C1-C3 alkyl);
RSS is H, CN, NO2, halo (preferably F or Cl), optionally substituted C1-C6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups), optionally substituted O-(C1-C6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted -C(O)(C1-C6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups);
RURE is H, a C1-C6 alkyl (preferably H or C1-C3 alkyl) or a–C(O)(C1-C6 alkyl) each of which groups is optionally substituted with one or two hydroxyl groups or up to three halogen, preferably fluorine groups, or an optionally substituted phenyl group, an optionally substituted heteroaryl, or an optionally substituted heterocycle, preferably for example piperidine, morpholine, pyrrolidine, tetrahydrofuran);
RPRO is H, optionally substituted C1-C6 alkyl or an optionally substituted aryl (phenyl or napthyl), heteroaryl or heterocyclic group selected from the group consisting of oxazole, isoxazole, thiazole, isothiazole, imidazole, diazole, oximidazole, pyrrole, pyrollidine, furan, dihydrofuran, tetrahydrofuran, thiene, dihydrothiene, tetrahydrothiene, pyridine, piperidine, piperazine, morpholine, quinoline, (each preferably substituted with a C1-C3 alkyl group, preferably methyl or a halo group, preferably F or Cl), benzofuran, indole, indolizine, azaindolizine;
RPRO1 and RPRO2 are each independently H, an optionally subsituted C1-C3 alkyl group or together form a keto group; and
each n is independently 0, 1, 2, 3, 4, 5, or 6 (preferably 0 or 1), or an optionally substituted heterocycle, preferably tetrahydrofuran, tetrahydrothiene, piperidine, piperazine or morpholine (each of which groups when substituted, are preferably substituted with a methyl or halo (F, Br, Cl), each of which groups may be optionally substituted with a linker group to which is attached a ABM group (including a ULM’ group).
[090] In certain preferred aspects,
Figure imgf000028_0002
is a
[091]
Figure imgf000028_0001
[092] wherein RPRO and n are the same as above.
[093] In certain embodiments, heteroaryl groups for W4 include an optionally substituted quinoline (which may be attached to the pharmacophore or substituted on any carbon atom within the quinoline ring), an optionally substituted indole, an optionally substituted indolizine, an optionally substituted azaindolizine, an optionally substituted benzofuran, including an optionally substituted benzofuran, an optionally substituted isoxazole, an optionally substituted thiazole, an optionally substituted isothiazole, an optionally substituted thiophene, an optionally substituted pyridine (2-, 3, or 4-pyridine), an optionally substituted imidazole, an optionally substituted pyrrole, an optionally substituted diazole, an optionally substituted triazole, a tetrazole, an optionally substituted oximidazole, or a group according to the chemical structure:
Figure imgf000029_0001
wherein:
Sc is CHRSS, NRURE, or O;
RHET is H, CN, NO2, halo (preferably Cl or F), optionally substituted C1-C6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups (e.g. CF3), optionally substituted O(C1-C6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted acetylenic group–C^C-Ra wherein Ra is H or a C1-C6 alkyl group (preferably C1-C3 alkyl);
RSS is H, CN, NO2, halo (preferably F or Cl), optionally substituted C1-C6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups), optionally substituted O-(C1-C6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted -C(O)(C1-C6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups);
RURE is H, a C1-C6 alkyl (preferably H or C1-C3 alkyl) or a–C(O)(C1-C6 alkyl), each of which groups is optionally substituted with one or two hydroxyl groups or up to three halogen, preferably fluorine groups, or an optionally substituted heterocycle, for example piperidine, morpholine, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, piperidine, piperazine, each of which is optionally substituted, and
YC is N or C-RYC, wherein RYC is H, OH, CN, NO2, halo (preferably Cl or F), optionally substituted C1-C6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups (e.g. CF3), optionally substituted O(C1-C6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted acetylenic group–C^C-Ra wherein Ra is H or a C1-C6 alkyl group (preferably C1-C3 alkyl), each of which groups may be optionally substituted with a linker group to which is attached a ABM group (including a ULM’ group).
[094] In certain embodiments, heterocycle groups for W4 include tetrahydrofuran, tetrahydrothiene, tetrahydroquinoline, piperidine, piperazine, pyrrollidine, morpholine, oxane or thiane, each of which groups may be optionally substituted, or a group according to the chemical structure:
Figure imgf000030_0001
preferably, a group,
Figure imgf000030_0002
wherein:
RPRO is H, optionally substituted C1-C6 alkyl or an optionally substituted aryl, heteroaryl or heterocyclic group;
RPRO1 and RPRO2 are each independently H, an optionally subsituted C1-C3 alkyl group or together form a keto group and
each n is independently 0, 1, 2, 3, 4, 5, or 6 (often 0 or 1), each of which groups may be
optionally substituted with a linker group to which is attached a ABM group (including a ULM’ group) In additional embodiments, W4 substituents for use in the present invention also include specifically (and without limitation to the specific compound disclosed) the W4 substituents which are found in the identified compounds disclosed herein (which includes the specific compounds which are disclosed in the present specification, and the figures which are attached hereto).Each of these W4 substituents may be used in conjunction with any number of W3 substituents which are also disclosed herein.
[095] In certain additional embodiments, ULM-a, is optionally substituted by 1-3 RP groups in the pyrrolidine moiety. Each RP is independently H, halo, -OH, C1-3alkyl.
[096] In any of the embodiments described herein, the W3, W4 can independently be covalently coupled to a linker which is attached one or more ABM groups.
[097] In certain embodiments, ULM is a group (derivatized or configured to be linked or coupled to an ABM via a linker (as indicated by the dashed line) according to the chemical structure:
Figure imgf000031_0001
[098] wherein, W3 is optionally substituted aryl, optionally substituted heteroaryl, or
Figure imgf000031_0002
[099] each R9 and R10 is independently hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted hydroxyalkyl, optionally substituted heteroaryl, or haloalkyl; or R9, R10, and the carbon atom to which they are attached form an optionally substituted cycloalkyl; [0100] R11 is optionally substituted heterocyclic, optionally substituted alkoxy, optionally
substituted heteroaryl, optionally substituted aryl,
Figure imgf000032_0001
or
Figure imgf000032_0002
;
[0101] R12 is H or optionally substituted alkyl;
[0102] R13 is H, optionally substituted alkyl, optionally substituted alkylcarbonyl, optionally substituted (cycloalkyl)alkylcarbonyl, optionally substituted aralkylcarbonyl, optionally substituted arylcarbonyl, optionally substituted (heterocyclyl)carbonyl, or optionally substituted aralkyl;
[0103] R14a, R14b, is each independently H, haloalkyl, or optionally substituted alkyl;
[0104] W5 is a phenyl or a 5-10 membered heteroaryl,
[0105] R15 is H, halogen, CN, OH, NO2, NR14aR14b, OR14a, CONR14aR14b, NR14aCOR14b, SO2NR14aR14b, NR14aSO2R14b, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted haloalkoxy; optionally substituted aryl; optionally substituted heteroaryl; optionally substituted cycloalkyl; optionally substituted cycloheteroalkyl;
[0106] each R16 is independently halo, optionally substituted alkyl, haloalkyl, hydroxy, optionally substituted alkoxy, or haloalkoxy;
[0107] o is 0, 1, 2, 3, or 4;
[0108] each R18 is independently halo, optionally substituted alkoxy, cyano, optionally substituted alkyl, haloalkyl, haloalkoxy or a linker; and
[0109] p is 0, 1, 2, 3, or 4.
[0110] In certain embodiments, R15
Figure imgf000032_0003
wherein R17 is H, halo, optionally substituted C3-6cycloalkyl, optionally substituted C1-6alkyl, optionally substituted C1-6alkenyl, and C1-6haloalkyl; and
[0111] Xa is S or O. [0112] In certain embodiments, R17 is selected from the group methyl, ethyl, isopropyl, and cyclopropyl.
0113 In certain additional embodiments R15 is selected from the rou consistin of:
Figure imgf000033_0001
[0114] In certain embodiments, R11 is selected from the group consisting of:
Figure imgf000033_0002
;
Figure imgf000034_0001
[0115] In certain embodiments, the ULM (derivatized or configured to be linked or coupled to an ABM via a linker (as indicated by the dashed line)) has the structure:
Figure imgf000034_0002
,
[0116] wherein
[0117] R9 is H;
[0118] R10 is isopropyl, tert-butyl, sec-butyl, cyclopentyl, or cyclohexyl;
[0119] R11 is
Figure imgf000034_0003
;
[0120] R12 is H;
[0121] R13 is H, optionally substituted alkyl, optionally substituted alkylcarbonyl, optionally substituted (cycloalkyl)alkylcarbonyl, optionally substituted aralkylcarbonyl, optionally substituted arylcarbonyl, optionally substituted (heterocyclyl)carbonyl, or optionally substituted aralkyl; [0122] R14a is H, haloalkyl, methyl, ethyl, isopropyl, cyclopropyl, or C1-C6 alkyl (linear, branched, optionally substituted), each optionally substituted with 1 or more halo, hydroxyl, nitro, CN, C1-C6 alkyl (linear, branched, optionally substituted), or C1-C6 alkoxyl (linear, branched, optionally substituted);and
[0123] R15 is
Figure imgf000035_0001
wherein R17 is H, halo, optionally substituted C3-6cycloalkyl, optionally substituted C1-6alkyl, optionally substituted C1-6alkenyl, or C1-6haloalkyl; and Xa is S or O.
[0124] In certain embodiments, the ULM or VLM is selected from the group consisting of:
Figure imgf000035_0002
Figure imgf000036_0001
attached to the linker moiety at the position indicated.
[0125] In certain embodiments the ULM is selected from the group consisting of:
(2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5- yl)benzyl)pyrrolidine-2-carboxamide; (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4- hydroxy-N-(4-(thiazol-5-yl)benzyl)pyrrolidine-2-carboxamide; (2S,4R)-1-((S)-2-amino-3,3- dimethylbutanoyl)-4-hydroxy-N-((S)-1-(4-(4-methylthiazol-5-yl)phenyl)ethyl)pyrrolidine-2- carboxamide; (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(oxazol-5- yl)benzyl)pyrrolidine-2-carboxamide hydrochloride; (2S,4R)-1-((S)-2-amino-3,3- dimethylbutanoyl)-4-hydroxy-N-(4-(4-methyloxazol-5-yl)benzyl)pyrrolidine-2-carboxamide; (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-N-(4-chlorobenzyl)-4-hydroxypyrrolidine-2- carboxamide hydrochloride; (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-N-(4-cyanobenzyl)- 4-hydroxypyrrolidine-2-carboxamide hydrochloride; (2S,4R)-1-((S)-2-amino-3-methylbutanoyl)- 4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide hydrochloride;
(2S,4R)-1-((S)-2-amino-3-methylbutanoyl)-4-hydroxy-N-(4-(thiazol-5-yl)benzyl)pyrrolidine-2- carboxamide hydrochloride; (2S,4R)-1-((S)-2-amino-3-methylbutanoyl)-4-hydroxy-N-(4-(4- methyloxazol-5-yl)benzyl)pyrrolidine-2-carboxamide hydrochloride; (2S,4R)-1-((S)-2-amino- 3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(1-methyl-1H-pyrazol-5-yl)benzyl)pyrrolidine-2- carboxamide hydrochloride; (2S,4R)-4-tert-butoxy-N-(2-hydroxy-4-(4-methylthiazol-5- yl)benzyl)-1-((S)-3-methyl-2-(1-oxoisoindolin-2-yl)butanoyl)pyrrolidine-2-carboxamide;
(2S,4R)-4-tert-butoxy-1-((S)-2-(6-fluoro-1-oxoisoindolin-2-yl)-3-methylbutanoyl)-N-(2- hydroxy-4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide; (2S,4R)-4-tert-butoxy-1- ((S)-2-(7-cyano-1-oxoisoindolin-2-yl)-3-methylbutanoyl)-N-(2-hydroxy-4-(4-methylthiazol-5- yl)benzyl)pyrrolidine-2-carboxamide; and (2S,4R)-1-((S)-2-Amino-3,3-dimethylbutanoyl)-4- hydroxy-N-((R)-2-hydroxy-1-(4-(4-methylthiazol-5-yl)phenyl)ethyl)pyrrolidine-2-carboxamide hydrochloride.
[0126] Exemplary Linkers
[0127] In certain embodiments, the compounds as described herein include one or more ABM chemically linked or coupled to one or more ULMs or VLMs via a chemical linker (L). In certain embodiments, the linker group L is a group comprises one or more covalently connected structural units of A (e.g. -A1…Aq- ), wherein A1 is coupled to an ABM moiety, and q is an integer greater than or equal to 0. In certain embodiments, q is an integer greater than or equal to 1.
[0128] In certain embodiments, e. g., wherein q is greater than 2, Aq is a group which is connected to a ULM or VLM moiety, and A1 and Aq are connected via structural units of A (number of such structural units of A: q-2).
[0129] In certain embodiments, e. g., wherein q is 2, Aq is a group which is connected to A1 and to a ULM or VLM moiety.
[0130] In certain embodiments, e. g., wherein q is 1, the structure of the linker group L is –A1–, and A1 is a group which is connected to a ULM or VLM moiety and an ABM moiety.
[0131] In additional embodiments, q is an integer from 1 to 100, 1 to 90, 1 to 80, 1 to 70, 1 to 60, 1 to 50, 1 to 40, 1 to 30, 1 to 20, or 1 to 10.
[0132] In certain embodiments, A1 to Aq are, each independently, a bond, CRL1RL2, O, S, SO, SO2, NRL3, SO2NRL3, SONRL3, CONRL3, NRL3CONRL4, NRL3SO2NRL4, CO, CRL1=CRL2, C^C, SiRL1RL2, P(O)RL1, P(O)ORL1, NRL3C(=NCN)NRL4, NRL3C(=NCN),
NRL3C(=CNO2)NRL4, C3-11cycloalkyl optionally substituted with 0-6 RL1 and/or RL2 groups , C3- 11heteocyclyl optionally substituted with 0-6 RL1 and/or RL2 groups, aryl optionally substituted with 0-6 RL1 and/or RL2 groups, heteroaryl optionally substituted with 0-6 RL1 and/or RL2 groups, wherein RL1 or RL
2, each independently, can be linked to other A groups to form cycloalkyl and/or heterocyclyl moeity which can be further substituted with 0-4 RL5 groups; [0133] wherein RL1, RL2, RL3, RL4 and RL5 are, each independently, H, halo, C1-8alkyl, OC1-8alkyl, SC1-8alkyl, NHC1-8alkyl, N(C1-8alkyl)2, C3-11cycloalkyl, aryl, heteroaryl, C3- 11heterocyclyl, OC1-8cycloalkyl, SC1-8cycloalkyl, NHC1-8cycloalkyl, N(C1-8cycloalkyl)2, N(C1- 8cycloalkyl)(C1-8alkyl), OH, NH2, SH, SO2C1-8alkyl, P(O)(OC1-8alkyl)(C1-8alkyl), P(O)(OC1- 8alkyl)2, CC-C1-8alkyl, CCH, CH=CH(C1-8alkyl), C(C1-8alkyl)=CH(C1-8alkyl), C(C1- 8alkyl)=C(C1-8alkyl)2, Si(OH)3, Si(C1-8alkyl)3, Si(OH)(C1-8alkyl)2, COC1-8alkyl, CO2H, halogen, CN, CF3, CHF2, CH2F, NO2, SF5, SO2NHC1-8alkyl, SO2N(C1-8alkyl)2, SONHC1-8alkyl, SON(C1- 8alkyl)2, CONHC1-8alkyl, CON(C1-8alkyl)2, N(C1-8alkyl)CONH(C1-8alkyl), N(C1-8alkyl)CON(C1- 8alkyl)2, NHCONH(C1-8alkyl), NHCON(C1-8alkyl)2, NHCONH2, N(C1-8alkyl)SO2NH(C1-8alkyl), N(C1-8alkyl) SO2N(C1-8alkyl)2, NH SO2NH(C1-8alkyl), NH SO2N(C1-8alkyl)2, NH SO2NH2.
[0134] In certain embodiments, the linker (L) is selected from the group consisting of:
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
[0135] In additional embodiments, the linker (L) comprises a structure selected from, but not limited to the structure shown below, wherein a dashed line indicates the attachment point to the ABM or ULM moieties: wherein:
Figure imgf000041_0001
WL1 and WL2 are each independently a 4-8 membered ring with 0-4 heteroatoms, optionally substituted with RQ, each RQ is independently a H, halo, OH, CN, CF3, NH2, carboxyl, C1-C6 alkyl (linear, branched, optionally substituted), C1-C6 alkoxy (linear, branched, optionally substituted), or 2 RQ groups taken together with the atom they are attached to, form a 4-8 membered ring system containing 0-4 heteroatoms;
YL1 is each independently a bond, C1-C6 alkyl (linear, branched, optionally substituted) and optionally one or more C atoms are replaced with O; or C1-C6 alkoxy (linear, branched, optionally substituted); and
n is 0-10.
[0136] In additional embodiments, the linker (L) comprises a structure selected from, but not limited to the structure shown below, wherein a dashed line indicates the attachment point to the ABM or ULM moieties:
Figure imgf000041_0002
wherein:
WL1 and WL2 are each independently aryl, heteroaryl, cyclic, heterocyclic, C1-6 alkyl (linear, branched, optionally substituted), C1-C6 alkoxy (linear, branched, optionally substituted), bicyclic, biaryl, biheteroaryl,or biheterocyclic, each optionally substituted with RQ, each RQ is independently a H, halo, OH, CN, CF3, NH2, carboxyl, hydroxyl, nitro, C^CH, C2-6 alkenyl, C2-6 alkynyl, C1-C6 alkyl (linear, branched, optionally substituted), C1-C6 alkoxy (linear, branched, optionally substituted), OC1-3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN, or 2 RQ groups taken together with the atom they are attached to, form a 4-8 membered ring system containing 0-4 heteroatoms; YL1 is each independently a bond, NRY1, O, S, NRY2, CRY1RY2, C=O, C=S, SO, SO2, C1-C6 alkyl (linear, branched, optionally substituted) and optionally one or more C atoms are replaced with O; C1-C6 alkoxy (linear, branched, optionally substituted);
QL is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, biheterocyclic, or bicyclic, optionally bridged, optionally substituted with 0-6 RQ, each RQ is independently H, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or R1, R2 together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms; and
n is 0-10.
[0137] In additional embodiments, the linker group is optionally substituted
(poly)ethyleneglycol having between 1 and about 100 ethylene glycol units, between about 1 and about 50 ethylene glycol units, between 1 and about 25 ethylene glycol units, between about 1 and 10 ethylene glycol units, between 1 and about 8 ethylene glycol units and 1 and 6 ethylene glycol units, between 2 and 4 ethylene glycol units,or optionally substituted alkyl groups interdispersed with optionally substituted, O, N, S, P or Si atoms. In certain embodiments, the linker is substituted with an aryl, phenyl, benzyl, alkyl, alkylene, or heterocycle group. In certain embodiments, the linker may be asymmetric or symmetrical.
[0138] In certain aspects the description provides a PROTAC compound in which the linker is cleavable in vivo into a functional E3 ligase binding moiety, and target protein binding moiety. In this regard, and without being bound by any particular theory, it is hypothesized that such a configuration can potentiate the beneficial effects of the degradation activity of the intact PROTAC molecule. Thus, in certain embodiments, the linker is configured or“tuned” to have the desired kinetics of cleavage into functional component molecules or active metabolites. In certain embodiments, the enzyme responsible for cleavage of the linker is a liver enzyme, such as, e.g., oxidases, peroxidase, reductases, transferases, dehydrogenases, peroxidases. In certain embodiments, the enzyme is at least one of cytochrome P450 oxidase, e.g., CYP3A4, Flavin- containing monooxygenase, alcohol dehydrogenase, aldehyde dehydrogenase, monoamine oxidase, peroxidase, glutathione S-transferase, cytochrome P450 reductase, sulfotransferase, methyltransferase, N-acetyltransferase, glucuronosyltransferase, transpeptidase, or combination thereof.
[0139] Exemplary Androgen Binding Moieties (ABMs)
[0140] In another aspect, the description provides AR binding moieties (ABM), which in certain aspects and embodiments are coupled to a linker and/or a ULM as described herein.
[0141] In any of the compounds described herein, the ABM comprises a chemical moiety that binds to the androgen receptor (AR). Various androgen receptor binding compounds have been described in literature, including various androgen derivatives such as testosterone, dihydrotestosterone, and metribolone (also known as methyltrienolone or R1881), and non- steroidal compounds such as bicalutamide, enzalutamide. Those of ordinary skill in the art would appreciate that these androgen receptor binding compounds could be potentially used as an ABM moiety in a PROTAC compound. Such literature includes, but not limited to, G. F. Allan et. al, Nuclear Receptor Signaling, 2003, 1, e009; R. H. Bradbury et. al, Bioorganic & Medicinal Chemistry Letters, 20115442–5445; C. Guo et. al, Bioorganic & Medicinal Chemistry Letters, 20122572-2578; P. K. Poutiainen et. al, J. Med. Chem.2012, 55, 6316− 6327 A. Pepe et. al, J. Med. Chem.2013, 56, 8280– 8297; M. E. Jung et al, J. Med. Chem.2010, 53, 2779–2796, which are incorporated by reference herein.
[0142] In certain embodiments, the ABM comprises a structure selected from, but not limited to the structures shown below, wherein a dashed line indicates the attachment point of linker moiety:
Figure imgf000043_0001
Figure imgf000044_0001
wherein:
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CF3, CN, C^CH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
Y1, Y2 are each independently NRY1, O, S;
Y3, Y4, Y5 are each independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2;
Q is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, optionally substituted with 0-6 RQ, each RQ is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); R1, R2, Ra, Rb, RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched,
optionally substituted by 1 or more halo, C1-6 alkoxyl), or R1, R2 together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms;
W2 is a bond, C1-6 alkyl, alicyclic, heterocyclic, aryl, heteroaryl, bicyclic, biaryl, biheteroaryl, or biheterocyclic, each optionally substituted by 1, 2 or 3 RW2; and
each RW2 is independently H, halo, OH, NH2, C1-6 alkyl (optionally substituted by 1 or more F), OC1-3alkyl (optionally substituted by 1 or more F), NRY1RY2, or CN.
[0143] In certain embodiments described herein, the ABM comprises a structure shown below, wherein a dashed line indicates the point of attachment to a linker moiety:
Figure imgf000045_0001
wherein:
W1 is aryl or heteroaryl, each optionally substituted by 1 or more halo, hydroxyl, nitro, CN, CF3, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo);
each Y3 is independently a bond, O, NRY2, CRY1RY2, or C=O;
each RQ is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
RY1, RY2 are each independently H, OH, or C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl);
W2 is a bond, C1-6 alkyl, aryl, heteroaryl, alicyclic, heterocyclic, bicyclic, biaryl, biheteroaryl, biheterocyclic, each optionally substituted by 1, 2 or 3 RW2; and
each RW2 is independently H, OH, NH2, CRY1RY2, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1-3alkyl (optionally substituted by 1 or more F).
[0144] In any of the embodiments described herein, the W2 is covalently coupled to one or more ULM or VLM groups, or a linker to which is attached one or more ULM or VLM groups as described herein.
[0145] In certain embodiments, W1 is
Figure imgf000045_0002
wherein each R22 is independently halo, optionally substituted alkyl, haloalkyl, cyano, or nitro; and
each R23 is independently H, halo, optionally substituted alkyl, haloalkyl, cyano, or nitro.
[0146] In certain additional embodiments, W1 is selected from the group consisting of:
Figure imgf000046_0001
[0147] In certain embodiments, ABM is selected from the group consisting of:
Figure imgf000046_0002
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
[0148] In certain embodiments, the ABM comprises the structure:
wherein:
Figure imgf000049_0002
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CF3, CN, C^CH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
Y3, Y4, Y5 are each independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2; Q is a 4 membered alicyclic ring with 0-2 heteroatoms, optionally substituted with 0-6 RQ, each RQ is independently H, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl);
W2 is a bond, C1-6 alkyl, alicyclic (e.g., C1-6 alicyclic), heterocyclic, aryl, heteroaryl, bicyclic, biheterocyclic, biaryl, or biheteroaryl each optionally substituted by 1, 2 or 3 RW2; and each RW2 is independently H, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1- 3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN.
[0149] In an additional aspect, the description provides an androgen receptor
bindingcompound comprising a structure of:
Figure imgf000050_0001
wherein:
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CN, CF3, C^CH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
Y1, Y2 are each independently NRY1, O, S;
Y3, Y4, Y5 are each independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2;
Q is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, optionally substituted with 0-6 RQ, each RQ is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl);
W2 is a bond, C1-6 alkyl, alicyclic, heterocyclic, aryl, heteroaryl, bicyclic, biheterocyclic, biaryl, biheteroaryl, each optionally substituted by 1, 2 or 3 RW2; and each RW2 is independently H, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1- 3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN.
[0150] In certain embodimetns, the androgen receptor binding compound of ABM-e is selected from the group consisting of:
trans-2-Chloro-4-[3-amino-2,2,4,4-tetramethylcyclobutoxy]benzonitrile;
cis-2-Chloro-4-[3-amino-2,2,4,4-tetramethylcyclobutoxy]benzonitrile;
trans 6-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyridazine-3- carboxamide;
trans tert-Butyl N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]carbamate; trans 4-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]benzamide; trans 5-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyrazine-2- carboxamide;
trans 2-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyrimidine-5- carboxamide;
4-Methoxy-N-[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]benzamide;
trans 1-(2-Hydroxyethyl)-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]- 1H-pyrazole-4-carboxamide;
trans 6-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyridine-3- carboxamide;
trans 4-[(5-Hydroxypentyl)amino]-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]benzamide;
trans tert-Butyl 2-({5-[(4-{[3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamoyl}phenyl)aminopentyl}oxy)acetate;
trans-2-Chloro-4-[3-amino-2,2,4,4-tetramethylcyclobutoxy]benzonitrile;
tert-butyl trans-(3-(3-chloro-4-cyanophenoxy)-2,2-dimethylcyclobutyl)carbamate; and
tert-butyl cis-(3-(3-chloro-4-cyanophenoxy)-2,2-dimethylcyclobutyl)carbamate.
[0151] The term“hydrocarbyl” shall mean a compound which contains carbon and hydrogen and which may be fully saturated, partially unsaturated or aromatic and includes aryl groups, alkyl groups, alkenyl groups and alkynyl groups. [0152] The term“alkyl” shall mean within its context a linear, branch-chained or cyclic fully saturated hydrocarbon radical or alkyl group, preferably a C1-C10, more preferably a C1-C6, alternatively a C1-C3 alkyl group, which may be optionally substituted. Examples of alkyl groups are methyl, ethyl, n-butyl, sec-butyl, n-hexyl, n-heptyl, n-octyl, n-nonyl, n-decyl, isopropyl, 2-methylpropyl, cyclopropyl, cyclopropylmethyl, cyclobutyl, cyclopentyl, cyclopen- tylethyl, cyclohexylethyl and cyclohexyl, among others. In certain preferred embodiments, compounds according to the present invention which may be used to covalently bind to dehalogenase enzymes. These compounds generally contain a side chain (often linked through a polyethylene glycol group) which terminates in an alkyl group which has a halogen substituent (often chlorine or bromine) on its distil end which results in covalent binding of the compound containing such a moiety to the protein.
[0153] The term“Alkenyl” refers to linear, branch-chained or cyclic C2-C10 (preferably C2-C6) hydrocarbon radicals containing at least one C=C bond.
[0154] The term“Alkynyl” refers to linear, branch-chained or cyclic C2-C10 (preferably C2-C6) hydrocarbon radicals containing at least one C^C bond.
[0155] The term“alkylene” when used, refers to a–(CH2)n- group (n is an integer generally from 0-6), which may be optionally substituted. When substituted, the alkylene group preferably is substituted on one or more of the methylene groups with a C1-C6 alkyl group (including a cyclopropyl group or a t-butyl group), more preferably a methyl group, but may also be substituted with one or more halo groups, preferably from 1 to 3 halo groups or one or two hydroxyl groups, O-(C1-C6 alkyl) groups or amino acid sidechains as otherwise disclosed herein. In certain embodiments, an alkylene group may be substituted with a urethane or alkoxy group (or other group) which is further substituted with a polyethylene glycol chain (of from 1 to 10, preferably 1 to 6, often 1 to 4 ethylene glycol units) to which is substituted (preferably, but not exclusively on the distal end of the polyethylene glycol chain) an alkyl chain substituted with a single halogen group, preferably a chlorine group. In still other embodiments, the alkylene (often, a methylene) group, may be substituted with an amino acid sidechain group such as a sidechain group of a natural or unnatural amino acid, for example, alanine, ^-alanine, arginine, asparagine, aspartic acid, cysteine, cystine, glutamic acid, glutamine, glycine, phenylalanine, histidine, isoleucine, lysine, leucine, methionine, proline, serine, threonine, valine, tryptophan or tyrosine. [0156] The term“unsubstituted” shall mean substituted only with hydrogen atoms. A range of carbon atoms which includes C0 means that carbon is absent and is replaced with H. Thus, a range of carbon atoms which is C0-C6 includes carbons atoms of 1, 2, 3, 4, 5 and 6 and for C0, H stands in place of carbon. The term“substituted” or“optionally substituted” shall mean independently (i.e., where more than substituent occurs, each substituent is independent of another substituent) one or more substituents (independently up to five substitutents, preferably up to three substituents, often 1 or 2 substituents on a moiety in a compound according to the present invention and may include substituents which themselves may be further substituted) at a carbon (or nitrogen) position anywhere on a molecule within context, and includes as substituents hydroxyl, thiol, carboxyl, cyano (C^N), nitro (NO2), halogen (preferably, 1, 2 or 3 halogens, especially on an alkyl, especially a methyl group such as a trifluoromethyl), an alkyl group (preferably, C1-C10 , more preferably, C1-C6), aryl (especially phenyl and substituted phenyl for example benzyl or benzoyl), alkoxy group (preferably, C1-C6 alkyl or aryl, including phenyl and substituted phenyl), thioether (C1-C6 alkyl or aryl), acyl (preferably, C1-C6 acyl), ester or thioester (preferably, C1-C6 alkyl or aryl) including alkylene ester (such that attachment is on the alkylene group, rather than at the ester function which is preferably substituted with a C1-C6 alkyl or aryl group), preferably, C1-C6 alkyl or aryl, halogen (preferably, F or Cl), amine (including a five- or six-membered cyclic alkylene amine, further including a C1-C6 alkyl amine or a C1-C6 dialkyl amine which alkyl groups may be substituted with one or two hydroxyl groups) or an optionally substituted–N(C0-C6 alkyl)C(O)(O-C1-C6 alkyl) group (which may be optionally substituted with a polyethylene glycol chain to which is further bound an alkyl group containing a single halogen, preferably chlorine substituent), hydrazine, amido, which is preferably substituted with one or two C1-C6 alkyl groups (including a carboxamide which is optionally substituted with one or two C1-C6 alkyl groups), alkanol (preferably, C1-C6 alkyl or aryl), or alkanoic acid (preferably, C1-C6 alkyl or aryl). Substituents according to the present invention may include, for example–SiR1R2R3 groups wherein each of R1 and R2 is as otherwise described herein and R3 is H or a C1-C6 alkyl group, preferably R1, R2, R3 in this context is a C1- C3 alkyl group (including an isopropyl or t-butyl group). Each of the above-described groups may be linked directly to the substituted moiety or alternatively, the substituent may be linked to the substituted moiety (preferably in the case of an aryl or heteraryl moiety) through an optionally substituted–(CH2)m- or alternatively an optionally substituted -(OCH2)m-, - (OCH2CH2)m- or–(CH2CH2O)m- group, which may be substituted with any one or more of the above-described substituents. Alkylene groups–(CH2)m- or–(CH2)n- groups or other chains such as ethylene glycol chains, as identified above, may be substituted anywhere on the chain. Preferred substitutents on alkylene groups include halogen or C1-C6 (preferably C1-C3) alkyl groups, which may be optionally substituted with one or two hydroxyl groups, one or two ether groups (O-C1-C6 groups), up to three halo groups (preferably F), or a sideshain of an amino acid as otherwise described herein and optionally substituted amide (preferably carboxamide substituted as described above) or urethane groups (often with one or two C0-C6 alkyl substitutents, which group(s) may be further substituted). In certain embodiments, the alkylene group (often a single methylene group) is substituted with one or two optionally substituted C1- C6 alkyl groups, preferably C1-C4 alkyl group, most often methyl or O-methyl groups or a sidechain of an amino acid as otherwise described herein. In the present invention, a moiety in a molecule may be optionally substituted with up to five substituents, preferably up to three substituents. Most often, in the present invention moieties which are substituted are substituted with one or two substituents.
[0157] The term“substituted” (each substituent being independent of any other substituent) shall also mean within its context of use C1-C6 alkyl, C1-C6 alkoxy, halogen, amido, carboxamido, sulfone, including sulfonamide, keto, carboxy, C1-C6 ester (oxyester or
carbonylester), C1-C6 keto, urethane -O-C(O)-NR1R2 or–N(R1)-C(O)-O-R1, nitro, cyano and amine (especially including a C1-C6 alkylene-NR1R2, a mono- or di- C1-C6 alkyl substituted amines which may be optionally substituted with one or two hydroxyl groups). Each of these groups contain unless otherwise indicated, within context, between 1 and 6 carbon atoms. In certain embodiments, preferred substituents will include for example,–NH-, -NHC(O)-, -O-, =O, -(CH2)m- (here, m and n are in context, 1, 2, 3, 4, 5 or 6), -S-, -S(O)-, SO2- or–NH-C(O)-NH-, - (CH2)nOH, -(CH2)nSH, -(CH2)nCOOH, C1-C6 alkyl, -(CH2)nO-(C1-C6 alkyl), -(CH2)nC(O)-(C1-C6 alkyl), -(CH2)nOC(O)-(C1-C6 alkyl), -(CH2)nC(O)O-(C1-C6 alkyl), -(CH2)nNHC(O)-R1, - (CH2)nC(O)-NR1R2, -(OCH2)nOH, -(CH2O)nCOOH, C1-C6 alkyl, -(OCH2)nO-(C1-C6 alkyl), - (CH2O)nC(O)-(C1-C6 alkyl), -(OCH2)nNHC(O)-R1, -(CH2O)nC(O)-NR1R2, -S(O)2-RS, -S(O)-RS (RS is C1-C6 alkyl or a–(CH2)m-NR1R2 group), NO2, CN or halogen (F, Cl, Br, I, preferably F or Cl), depending on the context of the use of the substituent. R1 and R2 are each, within context, H or a C1-C6 alkyl group (which may be optionally substituted with one or two hydroxyl groups or up to three halogen groups, preferably fluorine). The term“substituted” shall also mean, within the chemical context of the compound defined and substituent used, an optionally substituted aryl or heteroaryl group or an optionally substituted heterocyclic group as otherwise described herein. Alkylene groups may also be substituted as otherwise disclosed herein, preferably with optionally substituted C1-C6 alkyl groups (methyl, ethyl or hydroxymethyl or hydroxyethyl is preferred, thus providing a chiral center), a sidechain of an amino acid group as otherwise described herein, an amido group as described hereinabove, or a urethane group O-C(O)-NR1R2 group wherein R1 and R2 are as otherwise described herein, although numerous other groups may also be used as substituents. Various optionally substituted moieties may be substituted with 3 or more substituents, preferably no more than 3 substituents and preferably with 1 or 2 substituents. It is noted that in instances where, in a compound at a particular position of the molecule substitution is required (principally, because of valency), but no substitution is indicated, then that substituent is construed or understood to be H, unless the context of the substitution suggests otherwise.
[0158] The term "aryl" or“aromatic”, in context, refers to a substituted (as otherwise described herein) or unsubstituted monovalent aromatic radical having a single ring (e.g., benzene, phenyl, benzyl) or condensed rings (e.g., naphthyl, anthracenyl, phenanthrenyl, etc.) and can be bound to the compound according to the present invention at any available stable position on the ring(s) or as otherwise indicated in the chemical structure presented. Other examples of aryl groups, in context, may include heterocyclic aromatic ring systems“heteroaryl” groups having one or more nitrogen, oxygen, or sulfur atoms in the ring (moncyclic) such as imidazole, furyl, pyrrole, furanyl, thiene, thiazole, pyridine, pyrimidine, pyrazine, triazole, oxazole or fused ring systems such as indole, quinoline, indolizine, azaindolizine, benzofurazan, etc., among others, which may be optionally substituted as described above. Among the heteroaryl groups which may be mentioned include nitrogen-containing heteroaryl groups such as pyrrole, pyridine, pyridone, pyridazine, pyrimidine, pyrazine, pyrazole, imidazole, triazole, triazine, tetrazole, indole, isoindole, indolizine, azaindolizine, purine, indazole, quinoline, dihydroquinoline, tetrahydroquinoline, isoquinoline, dihydroisoquinoline,
tetrahydroisoquinoline, quinolizine, phthalazine, naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine, imidazopyridine, imidazotriazine, pyrazinopyridazine, acridine,
phenanthridine, carbazole, carbazoline, perimidine, phenanthroline, phenacene, oxadiazole, benzimidazole, pyrrolopyridine, pyrrolopyrimidine and pyridopyrimidine; sulfur-containing aromatic heterocycles such as thiophene and benzothiophene; oxygen-containing aromatic heterocycles such as furan, pyran, cyclopentapyran, benzofuran and isobenzofuran; and aromatic heterocycles comprising 2 or more hetero atoms selected from among nitrogen, sulfur and oxygen, such as thiazole, thiadizole, isothiazole, benzoxazole, benzothiazole, benzothiadiazole, phenothiazine, isoxazole, furazan, phenoxazine, pyrazoloxazole, imidazothiazole, thienofuran, furopyrrole, pyridoxazine, furopyridine, furopyrimidine, thienopyrimidine and oxazole, among others, all of which may be optionally substituted.
[0159] The term "substituted aryl" refers to an aromatic carbocyclic group comprised of at least one aromatic ring or of multiple condensed rings at least one of which being aromatic, wherein the ring(s) are substituted with one or more substituents. For example, an aryl group can comprise a substituent(s) selected from: -(CH2)nOH, -(CH2)n-O-(C1-C6)alkyl, -(CH2)n-O-(CH2)n- (C1-C6)alkyl, -(CH2)n-C(O)(C0-C6) alkyl, -(CH2)n-C(O)O(C0-C6)alkyl, -(CH2)n-OC(O)(C0- C6)alkyl, amine, mono- or di-(C1-C6 alkyl) amine wherein the alkyl group on the amine is optionally substituted with 1 or 2 hydroxyl groups or up to three halo (preferably F, Cl) groups, OH, COOH, C1-C6 alkyl, preferably CH3, CF3, OMe, OCF3, NO2, or CN group (each of which may be substituted in ortho-, meta- and/or para- positions of the phenyl ring, preferably para-), an optionally substituted phenyl group (the phenyl group itself is preferably substituted with a linker group attached to a ABM group, including a ULM group), and/or at least one of F, Cl, OH, COOH, CH3, CF3, OMe, OCF3, NO2, or CN group (in ortho-, meta- and/or para- positions of the phenyl ring, preferably para-), a naphthyl group, which may be optionally substituted, an optionally substituted heteroaryl, preferably an optionally substituted isoxazole including a methylsubstituted isoxazole, an optionally substituted oxazole including a methylsubstituted oxazole, an optionally substituted thiazole including a methyl substituted thiazole, an optionally substituted isothiazole including a methyl substituted isothiazole, an optionally substituted pyrrole including a methylsubstituted pyrrole, an optionally substituted imidazole including a methylimidazole, an optionally substituted benzimidazole or methoxybenzylimidazole, an optionally substituted oximidazole or methyloximidazole, an optionally substituted diazole group, including a methyldiazole group, an optionally substituted triazole group, including a methylsubstituted triazole group, an optionally substituted pyridine group, including a halo- (preferably, F) or methylsubstitutedpyridine group or an oxapyridine group (where the pyridine group is linked to the phenyl group by an oxygen), an optionally substituted furan, an optionally substituted benzofuran, an optionally substituted dihydrobenzofuran, an optionally substituted indole, indolizine or azaindolizine (2, 3, or 4-azaindolizine), an optionally substituted quinoline, and combinations thereof.
[0160] "Carboxyl" denotes the group -C(O)OR, wherein R is hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl or substituted heteroaryl , whereas these generic substituents have meanings which are identical with definitions of the corresponding groups defined herein.
[0161] The term“heteroaryl”or“hetaryl” can mean but is in no way limited to an optionally substituted quinoline (which may be attached to the pharmacophore or substituted on any carbon atom within the quinoline ring), an optionally substituted indole (including dihydroindole), an optionally substituted indolizine, an optionally substituted azaindolizine (2, 3 or 4-azaindolizine) an optionally substituted benzimidazole, benzodiazole, benzoxofuran, an optionally substituted imidazole, an optionally substituted isoxazole, an optionally substituted oxazole (preferably methyl substituted), an optionally substituted diazole, an optionally substituted triazole, a tetrazole, an optionally substituted benzofuran, an optionally substituted thiophene, an optionally substituted thiazole (preferably methyl and/or thiol substituted), an optionally substituted isothiazole, an optionally substituted triazole (preferably a 1,2,3-triazole substituted with a methyl group, a triisopropylsilyl group, an optionally substituted -(CH2)m-O- C1-C6 alkyl group or an optionally substituted -(CH2)m-C(O)-O-C1-C6 alkyl group), an optionally substituted pyridine (2-, 3, or 4-pyridine) or a group according to the chemical structure:
Figure imgf000058_0001
, wherein:
Sc is CHRSS, NRURE, or O;
RHET is H, CN, NO2, halo (preferably Cl or F), optionally substituted C1-C6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups (e.g. CF3), optionally substituted O(C1-C6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted acetylenic group–C^C-Ra wherein Ra is H or a C1-C6 alkyl group (preferably C1-C3 alkyl);
RSS is H, CN, NO2, halo (preferably F or Cl), optionally substituted C1-C6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups), optionally substituted O-(C1-C6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted -C(O)(C1-C6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups);
RURE is H, a C1-C6 alkyl (preferably H or C1-C3 alkyl) or a–C(O)(C1-C6 alkyl), each of which groups is optionally substituted with one or two hydroxyl groups or up to three halogen, preferably fluorine groups, or an optionally substituted heterocycle, for example piperidine, morpholine, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, piperidine, piperazine, each of which is optionally substituted, and
YC is N or C-RYC, wherein RYC is H, OH, CN, NO2, halo (preferably Cl or F), optionally substituted C1-C6 alkyl (preferably substituted with one or two hydroxyl groups or up to three halo groups (e.g. CF3), optionally substituted O(C1-C6 alkyl) (preferably substituted with one or two hydroxyl groups or up to three halo groups) or an optionally substituted acetylenic group–C^C-Ra wherein Ra is H or a C1-C6 alkyl group (preferably C1-C3 alkyl).
[0162] The terms“arylkyl” and“heteroarylalkyl” refer to groups that comprise both aryl or, respectively, heteroaryl as well as alkyl and/or heteroalkyl and/or carbocyclic and/or heterocycloalkyl ring systems according to the above definitions.
[0163] The term "arylalkyl" as used herein refers to an aryl group as defined above appended to an alkyl group defined above. The arylalkyl group is attached to the parent moiety through an alkyl group wherein the alkyl group is one to six carbon atoms. The aryl group in the arylalkyl group may be substituted as defined above.
[0164] The term“heterocycle” refers to a cyclic group which contains at least one heteroatom, i.e., O, N or S, and may be aromatic (heteroaryl) or non-aromatic. Thus, the heteroaryl moieties are subsumed under the definition of heterocycle, depending on the context of its use. Exemplary heterocyclics include: azetidinyl, benzimidazolyl, 1,4- benzodioxanyl, 1,3- benzodioxolyl, benzoxazolyl, benzothiazolyl, benzothienyl, dihydroimidazolyl, dihydropyranyl, dihydrofuranyl, dioxanyl, dioxolanyl, ethyleneurea, 1,3-dioxolane, 1,3-dioxane, 1,4-dioxane, furyl, homopiperidinyl, imidazolyl, imidazolinyl, imidazolidinyl, indolinyl, indolyl,
isoquinolinyl, isothiazolidinyl, isothiazolyl, isoxazolidinyl, isoxazolyl, morpholinyl,
naphthyridinyl, oxazolidinyl, oxazolyl, pyridone, 2-pyrrolidone, pyridine, piperazinyl, N- methylpiperazinyl, piperidinyl, phthalimide, succinimide, pyrazinyl, pyrazolinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, pyrrolyl, quinolinyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydroquinoline, thiazolidinyl, thiazolyl, thienyl, tetrahydrothiophene, oxane, oxetanyl, oxathiolanyl, thiane among others.
[0165] Heterocyclic groups can be optionally substituted with a member selected from the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxy, carboxyalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, hydroxyamino, alkoxyamino, nitro,—SO-alkyl,—SO-substituted alkyl,—SOaryl,—SO- heteroaryl,—SO2-alkyl,—SO2-substituted alkyl,—SO2-aryl, oxo (^O), and -SO2-heteroaryl. Such heterocyclic groups can have a single ring or multiple condensed rings. Examples of nitrogen heterocycles and heteroaryls include, but are not limited to, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthylpyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, phenanthroline, isothiazole, phenazine, isoxazole, phenoxazine, phenothiazine, imidazolidine, imidazoline, piperidine, piperazine, indoline, morpholino, piperidinyl, tetrahydrofuranyl, and the like as well as N-alkoxy-nitrogen containing heterocycles. The term "heterocyclic" also includes bicyclic groups in which any of the heterocyclic rings is fused to a benzene ring or a cyclohexane ring or another heterocyclic ring (for example, indolyl, quinolyl, isoquinolyl, tetrahydroquinolyl, and the like).
[0166] The term“cycloalkyl” can mean but is in no way limited to univalent groups derived from monocyclic or polycyclic alkyl groups or cycloalkanes, as defnied herein, e.g., saturated monocyclic hydrocarbon groups having from three to twenty carbon atoms in the ring, including, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and the like. The term "substituted cycloalkyl" can mean but is in no way limited to a monocyclic or polycyclic alkyl group and being substituted by one or more substituents, for example, amino, halogen, alkyl, substituted alkyl, carbyloxy, carbylmercapto, aryl, nitro, mercapto or sulfo, whereas these generic substituent groups have meanings which are identical with definitions of the corresponding groups as defined in this legend.
[0167] "Heterocycloalkyl" refers to a monocyclic or polycyclic alkyl group in which at least one ring carbon atom of its cyclic structure being replaced with a heteroatom selected from the group consisting of N, O, S or P. "Substituted heterocycloalkyl" refers to a monocyclic or polycyclic alkyl group in which at least one ring carbon atom of its cyclic structure being replaced with a heteroatom selected from the group consisting of N, O, S or P and the group is containing one or more substituents selected from the group consisting of halogen, alkyl, substituted alkyl, carbyloxy, carbylmercapto, aryl, nitro, mercapto or sulfo, whereas these generic substituent group have meanings which are identical with definitions of the
corresponding groups as defined in this legend.
[0168] Exemplary AR-PROTAC Compounds [0169] As described above, in certain aspects, the description provides bifuctional PROTAC compounds comprising at least one ABM group, a linker, and at least one ULM (or VLM) group as described herein.
[0170] In certain embodiments, the compound is selected from the group consisting of compounds 1-864 (as described in Tables 2-30), and salts and polymorphs thereof.
0171 In certain embodiments the com ound is selected from the rou consisting of:
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
;
Figure imgf000065_0001
;
Figure imgf000066_0001
;
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
[0172] In another embodiment, the present invention provides a library of compounds. The library comprises more than one compound wherein each compound has a formula of ABM- L-ULM, wherein ULM is a ubiquitin pathway protein binding moiety (preferably, an E3 ubiquitin ligase moiety as otherwise disclosed herein), e.g., a VLM, and ABM is an AR protein binding moiety, wherein ABM is coupled (preferably, through a linker moiety) to ULM, and wherein the ubiquitin pathway protein binding moiety recognizes an ubiquitin pathway protein, in particular, an E3 ubiquitin ligase.
[0173] The present description includes, where applicable, the compositions comprising the pharmaceutically acceptable salts, in particular, acid or base addition salts of compounds of the present invention.
[0174] The term "pharmaceutically acceptable salt" is used throughout the specification to describe, where applicable, a salt form of one or more of the compounds described herein which are presented to increase the solubility of the compound in the gastic juices of the patient's gastrointestinal tract in order to promote dissolution and the bioavailability of the compounds. Pharmaceutically acceptable salts include those derived from pharmaceutically acceptable inorganic or organic bases and acids, where applicable. Suitable salts include those derived from alkali metals such as potassium and sodium, alkaline earth metals such as calcium, magnesium and ammonium salts, among numerous other acids and bases well known in the pharmaceutical art. Sodium and potassium salts are particularly preferred as neutralization salts of the phosphates according to the present invention.
[0175] The acids which are used to prepare the pharmaceutically acceptable acid addition salts of the aforementioned base compounds useful in this invention are those which form non- toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions, such as the hydrochloride, hydrobromide, hydroiodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, acetate, lactate, citrate, acid citrate, tartrate, bitartrate, succinate, maleate, fumarate, gluconate, saccharate, benzoate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and pamoate [i.e., 1,1'-methylene-bis-(2-hydroxy-3 naphthoate)]salts, among numerous others.
[0176] Pharmaceutically acceptable base addition salts may also be used to produce pharmaceutically acceptable salt forms of the compounds or derivatives according to the present invention. The chemical bases that may be used as reagents to prepare pharmaceutically acceptable base salts of the present compounds that are acidic in nature are those that form non- toxic base salts with such compounds. Such non-toxic base salts include, but are not limited to those derived from such pharmacologically acceptable cations such as alkali metal cations (eg., potassium and sodium) and alkaline earth metal cations (eg, calcium, zinc and magnesium), ammonium or water-soluble amine addition salts such as N-methylglucamine-(meglumine), and the lower alkanolammonium and other base salts of pharmaceutically acceptable organic amines, among others.
[0177] Compositions
[0178] In another aspect, the description provides compositions comprising compounds as described herein, including salts thereof, and a pharmaceutically acceptable carrier. In certain embodiments, the compositions are therapeutic or pharmaceutical compositions comprising an effective amount of a compound as described herein and a pharmaceutally acceptable carrier.
[0179] The amount of compound in a pharmaceutical composition of the instant invention that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host and disease treated, the particular mode of administration.
Generally, an amount between 0.1 mg/kg and 1000 mg/kg body weight/day of active ingredients is administered dependent upon potency of the agent. Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds that exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects. The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.
[0180] The compositions of the present invention may be formulated in a conventional manner using one or more pharmaceutically acceptable carriers and may also be administered in controlled-release formulations. Pharmaceutically acceptable carriers that may be used in these pharmaceutical compositions include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as prolamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat. [0181] The active compound is included in the pharmaceutically acceptable carrier or diluent in an amount sufficient to deliver to a patient a therapeutically effective amount for the desired indication, without causing serious toxic effects in the patient treated. A preferred dose of the active compound for all of the herein-mentioned conditions is in the range from about 10 ng/kg to 300 mg/kg, preferably 0.1 to 100 mg/kg per day, more generally 0.5 to about 25 mg per kilogram body weight of the recipient/patient per day. A typical topical dosage will range from 0.01-5% wt/wt in a suitable carrier.
[0182] The compound is conveniently administered in any suitable unit dosage form, including but not limited to one containing less than 1mg, 1 mg to 3000 mg, preferably 5 to 500 mg of active ingredient per unit dosage form. An oral dosage of about 25-250 mg is often convenient.
[0183] The active ingredient is preferably administered to achieve peak plasma concentrations of the active compound of about 0.00001-30 mM, preferably about 0.1-30 ^M. This may be achieved, for example, by the intravenous injection of a solution or formulation of the active ingredient, optionally in saline, or an aqueous medium or administered as a bolus of the active ingredient. Oral administration is also appropriate to generate effective plasma concentrations of active agent.
[0184] The concentration of active compound in the drug composition will depend on absorption, distribution, inactivation, and excretion rates of the drug as well as other factors known to those of skill in the art. It is to be noted that dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition. The active ingredient may be administered at once, or may be divided into a number of smaller doses to be administered at varying intervals of time.
[0185] If administered intravenously, preferred carriers are physiological saline or phosphate buffered saline (PBS).
[0186] In one embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides,
polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art.
[0187] Liposomal suspensions may also be pharmaceutically acceptable carriers. These may be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No.4,522,811 (which is incorporated herein by reference in its entirety). For example, liposome formulations may be prepared by dissolving appropriate lipid(s) (such as stearoyl phosphatidyl ethanolamine, stearoyl phosphatidyl choline, arachadoyl phosphatidyl choline, and cholesterol) in an inorganic solvent that is then evaporated, leaving behind a thin film of dried lipid on the surface of the container. An aqueous solution of the active compound are then introduced into the container. The container is then swirled by hand to free lipid material from the sides of the container and to disperse lipid aggregates, thereby forming the liposomal suspension.
[0188] Modes of Adminstration
[0189] In any of the aspects or embodiments described herein, the therapeutic
compositions comprising compounds described herein can be in any suitable dosage form configured to be delivered by any suitable route. For example, the compounds can be
administered by any appropriate route, for example, orally, parenterally, intravenously, intradermally, subcutaneously, or topically, including transdermally, in liquid, cream, gel, or solid form, rectally, nasally, buccally, vaginally or via an implanted reservoir or by aerosol form.
[0190] The term "parenteral" as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, intraperitoneally or intravenously.
[0191] The compounds as described herein may be administered in single or divided doses by the oral, parenteral or topical routes. Administration of the active compound may range from continuous (intravenous drip) to several oral administrations per day (for example, Q.I.D.) and may include oral, topical, parenteral, intramuscular, intravenous, sub-cutaneous, transdermal (which may include a penetration enhancement agent), buccal, sublingual and suppository administration, among other routes of administration. Enteric coated oral tablets may also be used to enhance bioavailability of the compounds from an oral route of administration. The most effective dosage form will depend upon the pharmacokinetics of the particular agent chosen as well as the severity of disease in the patient.
[0192] Administration of compounds as sprays, mists, or aerosols for intra-nasal, intra- tracheal or pulmonary administration may also be used. Compounds as described herein may be administered in immediate release, intermediate release or sustained or controlled release forms. Sustained or controlled release forms are preferably administered orally, but also in suppository and transdermal or other topical forms. Intramuscular injections in liposomal form may also be used to control or sustain the release of compound at an injection site.
[0193] Sterile injectable forms of the compositions as described herein may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally- acceptable diluent or solvent, for example as a solution in 1, 3-butanediol. 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 are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or di-glycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as Ph. Helv or similar alcohol.
[0194] The pharmaceutical compositions as described herein may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added. Oral compositions will generally include an inert diluent or an edible carrier. They may be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound or its prodrug derivative can be incorporated with excipients and used in the form of tablets, troches, or capsules.
Pharmaceutically compatible binding agents, and/or adjuvant materials are included as part of the composition.
[0195] The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a dispersing agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil. In addition, dosage unit forms can contain various other materials which modify the physical form of the dosage unit, for example, coatings of sugar, shellac, or enteric agents.
[0196] The active compound or pharmaceutically acceptable salt thereof can be administered as a component of an elixir, suspension, syrup, wafer, chewing gum or the like. A syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.
[0197] Alternatively, the pharmaceutical compositions as described herein may be administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient, which is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols.
[0198] The pharmaceutical compositions of this invention may also be administered topically. Suitable topical formulations are readily prepared for each of these areas or organs. Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-acceptable transdermal patches may also be used. For topical applications, the pharmaceutical compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers. Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. In certain preferred aspects of the invention, the compounds may be coated onto a stent which is to be surgically implanted into a patient in order to inhibit or reduce the likelihood of occlusion occurring in the stent in the patient.
[0199] Alternatively, the pharmaceutical compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
[0200] For ophthalmic use, the pharmaceutical compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with our without a preservative such as
benzylalkonium chloride. Alternatively, for ophthalmic uses, the pharmaceutical compositions may be formulated in an ointment such as petrolatum.
[0201] The pharmaceutical compositions of this invention may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
[0202] Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid;
buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The parental preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
[0203] It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease or condition being treated. [0204] A patient or subject in need of therapy using compounds as described herein can be treated by administering to the patient (subject) an effective amount of the compound including pharmaceutically acceptable salts, solvates or polymorphs, thereof optionally in a pharmaceutically acceptable carrier or diluent, either alone, or in combination with other known agents.
[0205] Co-administration
[0206] Disease states of conditions which may be treated using compounds or
compositions according to the present description include, but not limited to, for example, cancer (e.g., prostate cancer), and Kennedy’s disease. In certain embodiments, the therapeutic or pharmaceutical compositions comprise an effective amount of an additional biologically or bioactive active agent, e.g., an agent effective for the treatment of cancer, that is co-administered.
[0207] The term“coadministration” or“combination therapy” shall mean that at least two compounds or compositions are administered to the patient at the same time, such that effective amounts or concentrations of each of the two or more compounds may be found in the patient at a given point in time. Although compounds according to the present invention may be co-administered to a patient at the same time, the term embraces both administration of two or more agents at the same time or at different times, provided that effective concentrations of all coadministered compounds or compositions are found in the subject at a given time. In certain preferred aspects of the present invention, one or more of the present compounds described above, are coadministered in combination with at least one additional bioactive agent, especially including an anticancer agent. In particularly preferred aspects of the invention, the co- administration of compounds results in synergistic therapeutic, including anticancer therapy.
[0208] In another aspect, the description provides a composition comprising an effective amount of two or more of the PROTAC compounds as described herein, and a pharmaceutically acceptable carrier. In certain embodiments, the composition further comprises an effective or synergistic amount of another bioactive agent that is not a PROTAC compound.
[0209] Pharmaceutical compositions comprising combinations of an effective amount of at least one bifunctional compound according to the present invention, and one or more of the compounds otherwise described herein, all in effective amounts, in combination with a pharmaceutically effective amount of a carrier, additive or excipient, represents a further aspect of the present invention. [0210] The term“bioactive agent” is used to describe an agent, other than the PROTAC compounds described herein, which is used in combination with the present compounds as an agent with biological activity to assist in effecting an intended therapy, inhibition and/or prevention/prophylaxis for which the present compounds are used. Preferred bioactive agents for use herein include those agents which have pharmacological activity similar to that for which the present compounds are used or administered and include for example, anti-cancer agents.
[0211] The term“additional anti-cancer agent” is used to describe an anti-cancer agent, which may be combined with PROTAC compounds according to the present description to treat cancer. These agents include, for example, everolimus, trabectedin, abraxane, TLK 286, AV- 299, DN-101, pazopanib, GSK690693, RTA 744, ON 0910.Na, AZD 6244 (ARRY-142886), AMN-107, TKI-258, GSK461364, AZD 1152, enzastaurin, vandetanib, ARQ-197, MK-0457, MLN8054, PHA-739358, R-763, AT-9263, a FLT-3 inhibitor, an androgen receptor inhibitor, a VEGFR inhibitor, an EGFR TK inhibitor, an aurora kinase inhibitor, a PIK-1 modulator, a Bcl-2 inhibitor, an HDAC inhbitor, a c-MET inhibitor, a PARP inhibitor, a Cdk inhibitor, an EGFR TK inhibitor, an IGFR-TK inhibitor, an anti-HGF antibody, a PI3 kinase inhibitors, an AKT inhibitor, a JAK/STAT inhibitor, a checkpoint-1 or 2 inhibitor, a focal adhesion kinase inhibitor, a Map kinase kinase (mek) inhibitor, a VEGF trap antibody, pemetrexed, erlotinib, dasatanib, nilotinib, decatanib, panitumumab, amrubicin, oregovomab, Lep-etu, nolatrexed, azd2171, batabulin, ofatumumab, zanolimumab, edotecarin, tetrandrine, rubitecan, tesmilifene, oblimersen, ticilimumab, ipilimumab, gossypol, Bio 111, 131-I-TM-601, ALT-110, BIO 140, CC 8490, cilengitide, gimatecan, IL13-PE38QQR, INO 1001, IPdR1 KRX-0402, lucanthone, LY317615, neuradiab, vitespan, Rta 744, Sdx 102, talampanel, atrasentan, Xr 311, romidepsin, ADS-100380, sunitinib, 5-fluorouracil, vorinostat, etoposide, gemcitabine, doxorubicin, liposomal doxorubicin, 5'-deoxy-5-fluorouridine, vincristine, temozolomide, ZK-304709, seliciclib; PD0325901, AZD-6244, capecitabine, L-Glutamic acid, N-[4-[2-(2-amino-4,7- dihydro-4-oxo-1 H - pyrrolo[2,3- d]pyrimidin-5-yl)ethyl]benzoyl]-, disodium salt, heptahydrate, camptothecin, PEG-labeled irinotecan, tamoxifen, toremifene citrate, anastrazole, exemestane, letrozole, DES(diethylstilbestrol), estradiol, estrogen, conjugated estrogen, bevacizumab, IMC- 1C11, CHIR-258); 3-[5-(methylsulfonylpiperadinemethyl)- indolylj-quinolone, vatalanib, AG- 013736, AVE-0005, the acetate salt of [D- Ser(Bu t ) 6 ,Azgly 10 ] (pyro-Glu-His-Trp-Ser-Tyr- D-Ser(Bu t )-Leu-Arg-Pro- Azgly-NH 2 acetate [C59H84N18Oi4 -(C2H4O2)X wherein x = 1 to 2.4], goserelin acetate, leuprolide acetate, triptorelin pamoate, medroxyprogesterone acetate, hydroxyprogesterone caproate, megestrol acetate, raloxifene, bicalutamide, flutamide, nilutamide, megestrol acetate, CP-724714; TAK-165, HKI-272, erlotinib, lapatanib, canertinib, ABX-EGF antibody, erbitux, EKB-569, PKI-166, GW-572016, Ionafarnib, BMS-214662, tipifarnib; amifostine, NVP-LAQ824, suberoyl analide hydroxamic acid, valproic acid, trichostatin A, FK-228, SU11248, sorafenib, KRN951 , aminoglutethimide, arnsacrine, anagrelide, L-asparaginase, Bacillus Calmette-Guerin (BCG) vaccine, adriamycin, bleomycin, buserelin, busulfan, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, clodronate, cyproterone, cytarabine, dacarbazine, dactinomycin, daunorubicin, diethylstilbestrol, epirubicin, fludarabine, fludrocortisone, fluoxymesterone, flutamide, gleevec, gemcitabine, hydroxyurea, idarubicin, ifosfamide, imatinib, leuprolide, levamisole, lomustine, mechlorethamine, melphalan, 6-mercaptopurine, mesna, methotrexate, mitomycin, mitotane, mitoxantrone, nilutamide, octreotide, oxaliplatin, pamidronate, pentostatin, plicamycin, porfimer, procarbazine, raltitrexed, rituximab, streptozocin, teniposide, testosterone, thalidomide, thioguanine, thiotepa, tretinoin, vindesine, 13-cis-retinoic acid, phenylalanine mustard, uracil mustard, estramustine, altretamine, floxuridine, 5-deooxyuridine, cytosine arabinoside, 6-mecaptopurine, deoxycoformycin, calcitriol, valrubicin, mithramycin, vinblastine, vinorelbine, topotecan, razoxin, marimastat, COL-3, neovastat, BMS-275291 , squalamine, endostatin, SU5416, SU6668, EMD121974, interleukin-12, IM862, angiostatin, vitaxin, droloxifene, idoxyfene, spironolactone, finasteride, cimitidine, trastuzumab, denileukin diftitox,gefitinib, bortezimib, paclitaxel, cremophor-free paclitaxel, docetaxel, epithilone B, BMS- 247550, BMS-310705, droloxifene, 4- hydroxytamoxifen, pipendoxifene, ERA-923, arzoxifene, fulvestrant, acolbifene, lasofoxifene, idoxifene, TSE-424, HMR- 3339, ZK186619, topotecan, PTK787/ZK 222584, VX-745, PD 184352, rapamycin, 40-O-(2-hydroxyethyl)-rapamycin, temsirolimus, AP-23573, RAD001, ABT-578, BC-210, LY294002, LY292223, LY292696, LY293684, LY293646, wortmannin, ZM336372, L-779,450, PEG-filgrastim, darbepoetin, erythropoietin, granulocyte colony- stimulating factor, zolendronate, prednisone, cetuximab, granulocyte macrophage colony- stimulating factor, histrelin, pegylated interferon alfa-2a, interferon alfa-2a, pegylated interferon alfa-2b, interferon alfa-2b, azacitidine, PEG-L-asparaginase, lenalidomide, gemtuzumab, hydrocortisone, interleukin-11, dexrazoxane, alemtuzumab, all-transretinoic acid, ketoconazole, interleukin-2, megestrol, immune globulin, nitrogen mustard, methylprednisolone, ibritgumomab tiuxetan, androgens, decitabine, hexamethylmelamine, bexarotene, tositumomab, arsenic trioxide, cortisone, editronate, mitotane, cyclosporine, liposomal daunorubicin, Edwina- asparaginase, strontium 89, casopitant, netupitant, an NK-1 receptor antagonist, palonosetron, aprepitant, diphenhydramine, hydroxyzine, metoclopramide, lorazepam, alprazolam, haloperidol, droperidol, dronabinol, dexamethasone, methylprednisolone, prochlorperazine, granisetron, ondansetron, dolasetron, tropisetron, pegfilgrastim, erythropoietin, epoetin alfa, darbepoetin alfa and mixtures thereof.
[0212] Methods of Treatment
[0213] In another aspect, the disclosure provides methods of modulating protein ubiquitination and degradation in a subject, e.g., a cell, a tissue, mammal, or human patient, the method comprising administering an effective amount of a PROTAC compound as described herein or a composition comprising an effective amount of the same to a subject, wherein the compound or composition comprising the same is effective in modulating protein ubquitination and degration of the protein in the subject. In certain embodiments, the protein is androgen receptor (AR).
[0214] In certain embodiments, the description provides a method for regulating protein activity of the androgen receptor in a patient in need comprising administering to said patient an amount of a compound as described herein to a patient.
[0215] In still additional embodiments, the description provides a method of treating a disease state or condition in a patient wherein dysregulated protein activity is responsible for said disease state or condition, said method comprising administering to said patient an effective amount of a compound as described herein to said patient in order to regulate said protein activity in said patient. In certain embodiments, the protein is AR.
[0216] The terms“treat”,“treating”, and“treatment”, etc., as used herein, refer to any action providing a benefit to a patient for which the present compounds may be administered, including the treatment of any disease state or condition which is modulated through the protein to which the present compounds bind. Disease states or conditions, including cancer, which may be treated using compounds according to the present invention are set forth hereinabove.
[0217] In another aspect, the disclosure provides methods of modulating AR protein ubiquitination and degradation in a subject, e.g., a cell, a tissue, mammal, or human patient, the method comprising administering an effective amount of a compound as described herein or a composition comprising an effective amount of the same to a subject, wherein the compound or composition comprising the same is effective in modulating AR protein ubquitination and degration of the protein in the subject.
[0218] In another aspect, the disclosure provides methods of treating or ameliorating a symptom of a disease related to AR activity in a subject, e.g., a cell, a tissue, mammal, or human patient, the method comprising administering an effective amount of a compound as described herein or a composition comprising an effective amount of the same to a subject in need thereof, wherein the compound or composition comprising the same is effective in treating or
ameliorating a symptom of a disease related to AR activity in the subject.
[0219] In certain embodiments, the disease or disorder is asthma, multiple sclerosis, cancer, prostate cancer, Kenney’s disease, ciliopathies, cleft palate, diabetes, heart disease, hypertension, inflammatory bowel disease, mental retardation, mood disorder, obesity, refractive error, infertility, Angelman syndrome, Canavan disease, Coeliac disease, Charcot–Marie–Tooth disease, Cystic fibrosis, Duchenne muscular dystrophy, Haemochromatosis, Haemophilia, Klinefelter's syndrome, Neurofibromatosis, Phenylketonuria, Polycystic kidney disease, (PKD1) or 4 (PKD2) Prader–Willi syndrome, Sickle-cell disease, Tay–Sachs disease, Turner syndrome. The method according to claim 48 wherein said cancer is squamous-cell carcinoma, basal cell carcinoma, adenocarcinoma, hepatocellular carcinomas, and renal cell carcinomas, cancer of the bladder, bowel, breast, cervix, colon, esophagus, head, kidney, liver, lung, neck, ovary, pancreas, prostate, and stomach; leukemias; benign and malignant lymphomas, particularly Burkitt's lymphoma and Non-Hodgkin's lymphoma; benign and malignant melanomas; myeloproliferative diseases; sarcomas, including Ewing's sarcoma, hemangiosarcoma, Kaposi's sarcoma, liposarcoma, myosarcomas, peripheral neuroepithelioma, synovial sarcoma, gliomas, astrocytomas, oligodendrogliomas, ependymomas, gliobastomas, neuroblastomas,
ganglioneuromas, gangliogliomas, medulloblastomas, pineal cell tumors, meningiomas, meningeal sarcomas, neurofibromas, and Schwannomas; bowel cancer, breast cancer, prostate cancer, cervical cancer, uterine cancer, lung cancer, ovarian cancer, testicular cancer, thyroid cancer, astrocytoma, esophageal cancer, pancreatic cancer, stomach cancer, liver cancer, colon cancer, melanoma; carcinosarcoma, Hodgkin's disease, Wilms' tumor or teratocarcinomas. In certain embodiments, the disease to be treated is cancer, e.g., prostate cancer, or Kennedy’s Disease. In a preferred embodiment, the subject is a human. [0220] In another aspect, the disclosure provides methods of treating or ameliorating a symptom of a disease related to AR activity in a subject, e.g., a cell, a tissue, mammal, or human patient, the method comprising administering an effective amount of a compound as described herein or a composition comprising an effective amount of the same and an effective or synergistic amount of another bioactive agent to a subject in need thereof, wherein the composition comprising the same is effective in treating or ameliorating a symptom of a disease related to AR activity in the subject. In certain embodiments, the disease to be treated is cancer, e.g., prostate cancer, or Kennedy’s Disease. In a preferred embodiment, the subject is a human. In certain additional embodiments, the additional bioactive agent is an anti-cancer agent.
[0221] In alternative aspects, the present invention relates to a method for treating a disease state by degrading a protein or polypeptide through which a disease state or condition is modulated comprising administering to said patient or subject an effective amount of at least one compound as described hereinabove, optionally in combination with an additional bioactive agent. The method according to the present invention may be used to treat a large number of disease states or conditions including cancer, by virtue of the administration of effective amounts of at least one compound described herein.
[0222] In another aspect, the disclosure provides methods for identifying the effects of the degradation of proteins of interest in a biological system using compounds according to the present invention.
[0223] Kits
[0224] In another aspect, the description provides kits comprising compounds or compositions as described herein. The kit may be promoted, distributed, or sold as a unit for performing the methods of the present invention. In addition, the kits of the present invention may preferably contain instructions which describe a suitable use. Such kits can be conveniently used, e.g., in clinical settings, to treat patients exhibiting symptoms of, e.g., cancer or Kennedy’s Disease.
[0225] EXAMPLES
[0226] General Chemistry– Analysis and Synthesis
[0227] Unless otherwise noted, all materials/reagents were obtained from commercial suppliers and used without further purification. Reactions were monitored by LC-MS and/or thin layer chromatography (TLC) on silica gel 60 F254 (0.2mm) pre-coated aluminum foil or glass- backed and visualized using UV light. Flash chromatography (alternatively called "ISCO chromatography") was performed using an ISCO CombiFiash RF 75 PSI or equivalent with RediSep normal-phase silica gel cartridges. Preparative TLC was performed on Whatman LK6F Silica Gel 60A size 20x20 cm plates with a thickness of 1000 ^m or equivalent.
[0228] 1HNMR (300 or 400 MHz) and 13CNMR (100.6 MHz) spectra were recorded on Bruker spectrometers at room temperature with TMS or the residual solvent peak as the internal standard. The line positions or multiples are given in (^) and the coupling constants (J) are given as absolute values in Hertz (Hz). The multiplicities in 1HNMR spectra are abbreviated as follows: s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), br or broad (broadened).
[0229] Preparative HPLC purifications were performed on a Waters® UV-Directed Purification System equipped with 2545 Binary Gradient Module, 2767 Sample Manager and 2489 UV/Visible Detector, controlled by MassLynx V4.1 software. All purification work was completed using the following columns: Atlantis Prep T3 OBD Column, SunFire Prep C18 OBD Column and XBridge Prep Phenyl OBD Column. The mobile phases were water (with 0.1%TFA or 0.01% NH4HCO3) and acetonitrile; all reagents used were of HPLC grade. The flow rate was 30ml/min. After the columns, a 1:1000 LC packings flow splitter allowed transfer of a small portion of the eluent into the UV detector. The electrospray source was set at 3.0 kV capillary voltage, 30 V conevoltage, 110°C source temperature, 350°C desolvation temperature, 600L/h desolvation gas flow, and 60L/h cone gas flow. For the analyzer, the multiplier was set at 550 for preparative tune method.
[0230] Analytical LC-MS data was collected on a Shimadzu LCMS-2020 with a mobile phase of 0.05% TFA in Acetonitrile (A) and 0.05% TFA in HPLC grade water (B); 0.1% FA in Acetonitrile (A) and 0.1% FA in HPLC grade water (B); Acetonitrile (A) and 5 mM ammonium bicarbonate in HPLC grade water (B).
[0231] Shimadzu LCMS-2020 equipped with LC-20AD or 30AD pumps, SPD-M20A PDA and Alltech 3300 ELSD. The system uses the following conditions for 2.0 min, 2.6 min, 3 min, 3.6 min, 5 min or 5.6 min run time.
[0232] 2.0 minute run: Kinetex XB-C 18100A column, 2.6 ^m, 3.0x 50 mm. The flow rate is 1.5 mL/min, the run time is 2.0 min, and the gradient profiles are 0.01 min 10% A, 1.10 min 100% A, 1.60 min 100% A, 1.70 min 10% A, 2.00 min 10% A. [0233] 2.6 minute run: Shim-pack VP-ODS column, 2.2 ^m, 3.0x 50 mm. The flow rate is 1.5 mL/min, the run time is 2.6 min, and the gradient profiles are 0.01 min 5% A, 1.20 min 100% A, 2.20 min 100% A, 2.30 min 5% A, 2.60 min 5% A.
[0234] 3.0 minute run: ACE UltraCore Super C18 column, 2.5 ^m, 3.0x 50 mm. The flow rate is 1.5 mL/min, the run time is 3.0 min, and the gradient profiles are 0.01 min 10% A, 2.00 min 95% A, 2.60 min 95% A, 2.70 min 10% A, 3.00 min 10% A.
[0235] 3.6 minute run: Shim-pack VP-ODS column , 2.2 ^m, 3.0x 50 mm. The flow rate is 1.5 mL/min, the run time is 3.6 min, and the gradient profiles are 0.01 min 5% A, 2.20 min 100% A, 3.20 min 100% A, 3.30 min 5% A, 3.60 min 5% A.
[0236] 5.0 minute run: ACE UltraCore Super C18 column, 2.5 ^m, 3.0x 50 mm. The flow rate is 1.5 mL/min, the run time is 5.0 min, and the gradient profiles are 0.01 min 10% A, 4.00 min 60% A, 4.70 min 60% A, 4.80 min 10% A, 5.00 min 10% A.
[0237] 5.6 minute run: Shim-pack VP-ODS column , 2.2 ^m, 3.0x 50 mm. The flow rate is 1.5 mL/min, the run time is 5.6 min, and the gradient profiles are 0.01 min 5% A, 3.00 min 50% A, 5.00 min 50% A, 5.20 min 5% A, 5.60 min 5% A
[0238] Alternatively, analytical LC-MS data was collected on Agilent infinity 1260 LC, Agilent 6230 TOF mass spectrometer. The analysis is conducted on a Poroshell 120 EC C18 column (50mm x 3.0mm internal diameter 2.7^m packing diameter) at 45oC.
[0239] The solvents employed are:
[0240] A = 0.1% v/v solution of formic acid in water.
[0241] B = 0.1% v/v solution of formic acid in acetonitrile.
[0242] The gradient employed are as follows:
[0243] Table 1. Exemplary Column Gradients.
Figure imgf000092_0001
4.5 1 95 5
[0244]
[0245] The UV detection is an averaged signal from wavelength of 210nm to 350nm and mass spectra are recorded on a mass spectrometer using positive mode electrospray ionization.
[0246] Unless otherwise noted, all compounds were prepared with LC-MS purity >95%.
[0247] Chemical Synthesis
[0248] A PROTAC of ABM-L-ULM, or their pharmaceutically acceptable salts, polymorphic forms, prodrugs, solvate forms and isotope containing derivatives thereof, may be prepared by the general approaches described below (scheme 3-4), together with synthetic methods known in the art of organic chemistry, or modifications and derivatizations that are familiar to those of ordinary skill in the art.
[0249] Scheme 3:
Figure imgf000093_0001
[0250] Scheme 4:
Figure imgf000093_0002
[0251] More specifically, The compounds of the Formula I, or their pharmaceutically acceptable salts, may be prepared by the general approaches described below (scheme 5-6), together with synthetic methods known in the art of organic chemistry, or modifications and derivatizations that are familiar to those of ordinary skill in the art.
[0252] Scheme 5:
Figure imgf000094_0001
Figure imgf000094_0002
[0253] Scheme 6:
Figure imgf000094_0003
Figure imgf000094_0004
[0254] In schemes 3-6, L, ABM, ULM groups, W1, W2, W3, W4, X1, X2, Y1, Y2, R1, R2 , and RP are as define above. RG1, RG2, RG3 and RG4 are moieties with suitable reacting groups that would be necessary to enable the synthetic chemistry to connect intermediate A, intermediate L and intermediate V together into PROTAC compounds of Formula I via covalent bond formation chemistries. These chemistries, depends on specific reacting groups, include but not limited to, amide formation, ester formation, carbamate formation, urea formation, ether formation, amine formation and various C-C, C=C bond formation. The stage 1 and stage 2 transformations in scheme 5 and scheme 6 may involve 1 or multiple synthetic steps. These are routine methods known in the art such as those methods disclosed in standard reference books such as the Compendium of Organic Synthetic Methods, Vol. I-VI (Wiley-lnterscience); or the Comprehensive Organic Transformations, by R.C. Larock (Wiley-lnterscience). Unless otherwise indicated, the substituents in the schemes are defined as above. Isolation and purification of the products is accomplished by standard procedures, which are known to a chemist of ordinary skill.
[0255] In certain examples, for the chemistry described in schemes 3-6, RG1 is a moiety with a suitable nucleophile such as -OH and RG2 is a moiety with a suitable leaving group such as halogen, -OMs, or–OTs. In a typical procedure, a RG1 containing intermediate is reacted with a RG2 containing intermediate in a suitable solvent. Suitable solvents include, but are not limited to, water, ethers such as THF, glyme, and the like; chlorinated solvents such as DCM, 1,2- dichloroethane (DCE) or CHCI3 and the like, toluene, benzene and the like, DMF, DMSO, MeCN. If desired, mixtures of these solvents are used. A base may be added to the reaction to facilitate the reaction. Suitable bases include, but are not limited to, Cs2CO3, K2CO3, and the like. The above process may be carried out at temperatures between about -78 °C and about 150 °C. Preferably, the reaction is carried out between about 20 °C and about 120 °C.
[0256] In another example, chemistry described in in schemes 3-6, RG3 is a moiety contains a–COOH group and RG4 is a moiety contains a suitable amine group. In a typical procedure, a RG3 containing intermediate is reacted with a RG4 containing intermediate in a suitable solvent in the presence of a suitable amide coupling reagent. Suitable solvents include, but are not limited to, water, ethers such as THF, glyme, and the like; chlorinated solvents such as DCM, 1,2-dichloroethane (DCE) or CHCl3 and the like, toluene, benzene and the like, DMF, DMSO, MeCN. If desired, mixtures of these solvents are used. In this case, the preferred solvents are DMF or DCM. A suitable amide coupling reagent include, but are not limited to, DCC, EDC, HATU, HBTU, PyBOP and the like. A base is often added to the reaction. Suitable bases include, but are not limited to, TEA, DIPEA, and the like. The above process may be carried out at temperatures between about -78 °C and about 150 °C. Preferably, the reaction is carried out between about 0 °C and about 100 °C.
[0257] Although not explicitly shown in schemes 3-6, a chemist of ordinary skill would realize that during any of the synthetic sequences it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This can be achieved by means of conventional protecting groups, such as those described in T.W. Greene, Protective Groups in Organic Chemistry, John Wiley & Sons (1981); T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Chemistry, John Wiley & Sons (1991), and T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Chemistry, John Wiley & Sons, 1999, which are hereby incorporated by reference in their entireties.
[0258] When a general or exemplary synthetic procedure is referred to, one skilled in the art can readily determine the appropriate reagents, if not indicated, extrapolating from the general or exemplary procedures. Some of the general procedures are given as examples for preparing specific compounds. One skilled in the art can readily adapt such procedures to the synthesis of other compounds. Representation of an unsubstituted position in structures shown or referred to in the general procedures is for convenience and does not preclude substitution as described elsewhere herein. For specific groups that can be present, either as R groups in the general procedures or as optional substituents not shown, refer to the descriptions in the remainder of this document, including the claims, summary and detailed description.
[0259] The process to produce compounds of the present invention is preferably carried out at about atmospheric pressure although higher or lower pressures can be used if desired. Substantially equimolar amounts of reactants are preferably used although higher or lower amounts can also be used.
[0260] The compounds of Formulae II-IV (below), or their pharmaceutically acceptable salts, may be prepared by the methods similar to chemistry illustrated above for synthesis of compounds of Formula I (scheme 3-6), together with synthetic methods known in the art of organic chemistry, or modifications and derivatizations that are familiar to those of ordinary skill in the art: ; and
Figure imgf000097_0001
.
[0261] For compounds of Formulae II-IV, L, ABM, ULM groups, W1, W2, W3, W4 , X1, X2, Y1, Y2, R1, R2, RP , Ra, Rb, Rc and Rd are as define above.
[0262] In certain embodiments, ABM compounds are active without forming
bifunctional compounds of formular II-IV.
[0263] Synthesis of ABM Moieties
[0264] ABM-1: 2-chloro-4-(3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1- l benzonitrile
[0265]
Figure imgf000097_0002
[0266] Step 1: Synthesis of 2-chloro-4-isothiocyanatobenzonitrile (B).
[0267] To a stirred solution of 4-amino-2-chlorobenzonitrile (A, 1 g, 6.55 mmol) in dichloromethane (9 mL) was added sodium bicarbonate (2.21 g, 26.31 mmol) and water (9 mL). The resulting mixture was cooled to 0 °C, to which thiophosgene (817 mg, 7.11 mmol) was added in drop wise in 30 min at 0 °C. The resulting mixture was then warmed up to room temperature and stirred at room temperature for 1 hour. The reaction mixture was diluted with dichloromethane (200 mL), washed with brine (50 mL x 2), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue. The residue was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v: v = 1: 30)) to give desired product (yield: 71%) 1HNMR (400 MHz, CDCl3): ^ 7.69 (d, J = 8.0 Hz, 1H), 7.38 (s, 1H), 7.28 (m, 1H);
[0268] Step 2: Synthesis of 2-chloro-4-[3-(4-hydroxyphenyl)-5-imino-4, 4-dimethyl-2- sulfanylideneimidazolidin-1-yl]benzonitrile (D).
[0269] To a stirred solution of 2-chloro-4-isothiocyanatobenzonitrile (B, 399 mg, 2.05 mmol) in toluene (5 mL) was added 2-[(4-hydroxyphenyl)amino]-2-methylpropanenitrile (C, 300 mg, 1.70 mmol) and 4-dimethylaminopyridine (312 mg, 2.55 mmol). The resulting solution was then heated in an oil bath to 100 °C and stirred at the same temperature for 16h. LC-MS indicated formation of the desired product. The reaction mixture was concentrated under vacuum to give a crude reside which was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v: v =1:1)) to give desired product (yield: 48%) as a brown solid. LC- MS (ES+): m/z 370.95 [MH+], tR =0.74 min (2.0 minute run);
[0270] Step 3: Synthesis of 2-chloro-4-[3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-^2- sulfanylideneimidazolidin-1-yl]benzonitrile (ABM-1).
[0271] To a stirred solution of 2-chloro-4-[3-(4-hydroxyphenyl)-5-imino-4, 4-dimethyl- 2-sulfanylideneimidazolidin-1-yl]benzonitrile (D, 300 mg, 0.81 mmol) in methanol (6 mL) was added aqueous hydrogen chloride (2N, 3.0 mL). The resulting solution was then heated in an oil bath to 100 °C and stirred at the same temperature for 2h. The reaction mixture was diluted with water (30 mL), extracted with ethyl acetate (60 mL x 3), washed with water (50 mL), dried over anhydrous sodium sulfate and concentrated under vacuum to give titled product (yield: 93%) as a yellow solid, which was used for the next step without any further purifications. LC-MS (ES+): m/z 372.00 [MH+], tR =0.97 min (2.0 minute run).
[0272] Unless otherwise noted, the following intermediates and their analogs (for examples, but not limited to, analogs with substitutions such as halogens) were synthesized according to similar procedures described above for the synthesis of ABM-1, by utilizing corresponding starting materials and reagents.
[0273] ABM-2: 2-fluoro-4-(3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)benzonitrile:
Figure imgf000099_0001
.
[0274] ABM-3: 4-(3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1- yl)-2-(trifluoromethyl)benzonitrile:
Figure imgf000099_0002
.
[0275] ABM-4: 5-(3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1- yl)-3-(trifluoromethyl)picolinoni ril
Figure imgf000099_0003
.
[0276] ABM-5: 4-(3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1- yl)-2-methoxybenzonitrile:
Figure imgf000099_0004
.
[0277] ABM-6: 4-(3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1- yl)-2-methylbenzonitrile:
Figure imgf000099_0005
.
[0278] ABM-7: 3-chloro-5-(3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)picolinonitrile:
Figure imgf000100_0001
.
[0279] ABM-8: 4-(1-(4-hydroxyphenyl)-4-oxo-2-thioxo-8-oxa-1,3- diazaspiro[4.5]decan-3-yl)-2-(trifluorometh l benzonitrile:
Figure imgf000100_0002
.
[0280] ABM-9: 4-(1-(4-hydroxyphenyl)-8-methyl-4-oxo-2-thioxo-1,3,8- triazaspiro[4.5]decan-3-yl)-2-(trifluorometh l benzonitrile:
Figure imgf000100_0003
[0281] ABM-10: 4-(5-(4-hydroxyphenyl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7- yl)-2-(trifluoromethyl)benzonitrile
Figure imgf000100_0004
.
[0282] ABM-11: 5-(5-(4-hydroxyphenyl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7- yl)-3-(trifluoromethyl)picolinonitrile:
Figure imgf000100_0005
.
[0283] ABM-12: 4-(4-(3-(4-cyano-3-(trifluoromethyl)phenyl)-5,5-dimethyl-4-oxo-2- thioxoimidazolidin-1-yl)phenyl)butanoic acid:
Figure imgf000101_0001
.
[0284] ABM-13: 2-chloro-4-(3-(4'-hydroxybiphenyl-4-yl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)benzonitrile:
Figure imgf000101_0002
.
[0285] ABM-14: 4-(3-(4'-hydroxybiphenyl-4-yl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)-2-
Figure imgf000101_0003
.
[0286] ABM-15: 5-(3-(4'-hydroxybiphenyl-4-yl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)-3-(trifluoromethyl)picolinonitrile:
Figure imgf000101_0004
.
[0287] ABM-16: 4-(3-(3-fluoro-4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile:
Figure imgf000101_0005
.
[0288] ABM-17: 1-(4-hydroxyphenyl)-5,5-dimethyl-3-(4-nitro-3- (trifluoromethyl)phenyl)-2-thioxoimidazolidin-4-one:
Figure imgf000102_0001
.
[0289] ABM-18: 4-(3-(3,5-difluoro-4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile:
Figure imgf000102_0002
.
[0290] ABM-19: 4-(3-(4-hydroxyphenyl)-4,4-dimethyl-2,5-dioxoimidazolidin-1-yl)-2- (trifluoromethyl)benzonitrile:
Figure imgf000102_0003
.
[0291] ABM-20: 4-(3-(6-hydroxypyridin-3-yl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile:
Figure imgf000102_0004
.
[0292] ABM-21: 2-chloro-4-(3-(3-fluoro-4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)benzonitrile:
Figure imgf000102_0005
.
[0293] ABM-22: 4-(3-(3-fluoro-4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)-2-methoxybenzonitrile:
Figure imgf000103_0001
.
[0294] ABM-23: 5-(3-(3-fluoro-4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)-3-(trifluoromethyl)picolinonitrile:
Figure imgf000103_0002
.
[0295] ABM-24: 5-(3-(2-fluoro-4'-hydroxybiphenyl-4-yl)-4,4-dimethyl-5-oxo-2- thioxoimidazolidin-1-yl)-3-(trifluorometh l icolinonitrile:
Figure imgf000103_0003
.
[0296] ABM-25: 4-(4,4-dimethyl-5-oxo-3-(4-(piperidin-4-yl)phenyl)-2- thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile:
Figure imgf000103_0004
.
[0297] ABM-26: trans-2-Chloro-4-[3-amino-2,2,4,4- tetramethylcyclobutoxy]benzonitrile.
Figure imgf000103_0005
[0298] ABM-27: cis-2-Chloro-4-[3-amino-2,2,4,4- tetramethylcyclobutoxy]benzonitrile
Figure imgf000104_0001
Step 1 : Synthesis of tot-butyl (4-((tra«5-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)carbamoyl)phenyl)carbamate.
[0303] A suspension of 4-((tert-butoxycarbonyl)amino)benzoic acid (1.50 g, 6.34 mmol) in methylene dichloride (40 mL) was charged with N,N-diisopropylethylarnine (3.30 mL, 19.0 mmol), followed by 4-(tram-3-arrdno-2,2,4,4-teti"amethylcyclobutoxy)-2-chlorobenzonitiile hydrochloride (2.0 g, 6.34 mmol). The mixture was stirred for several minutes and then charged with HATU (2.41 g, 6.34 mmol). The reaction mixture was allowed to stir at room temperature for 2 hours. The mixture was diluted with methylene dichloride (40 mL), washed with aqueous IN HQ (2 x), saturated aqueous sodium bicarbonate (2 x), brine, and dried over anhydrous Na2S04. The crude product was used in next step; [0304] Step 2: Synthesis of trans 4-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetram ethyl ey cl obutyl ]b enzami de .
[0305] 4M HC1 in Dioxane (1.38 mL, 40.0 mmol) was added to a pre-mixed solution of tert-butyl (4-((tra«5-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)carbamoyl)phenyl)carbamate (2.00 g, 4.01 mmol) in MeOH (2 mL) and left to stir at room temperature for 1 hour till completion. The reaction mixture was concentrated in vacuo to a solid, which was dissolved with 5% MeOH in DCM. The organic layer was washed with sodium bicarbonate (2 x), filtered through a Biotage Universal Phase Separator and then concentrated in vacuo to a solid. The crude product was recrystallized from EtOH/Heptanes to afford the desired product as a white solid, 1.2 g, 75% yield. lU NMR (400 MHz,
METHANOL-d4) δ 7.72 (d, J = 8.80 Hz, 1H), 7.61 (d, J = 8.61 Hz, 2H), 7.13 (d, J = 2.35 Hz, 1 H), 6.98 (dd, J = 2.45, 8.71 Hz, 1 H), 6.69 (d, J = 8.61 Hz, 2H), 4.28 (s, IH), 4.12 (s, IH), 1.27 (s, 6H). 1.22 (s, 6H). LC-MS (ES+): mJz 398.16/400.15 | M f Pj.
[0306] Unless otherwise noted, the following intermediates and their analogs (for examples, but not limited to, analogs with substitutions such as halogens) were synthesized according to similar procedures described above for the synthesis of ABM-30, by utilizing corresponding starting materials and reagents.
[0307] ABM-31: trans 5-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]pyrazine~2-carboxamide
Figure imgf000105_0001
[0308] ABM-32: trans 2-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyljpyrimidine-5-carboxamid
Figure imgf000105_0002
[0309] ABM-33: 4-Methoxy-N-[(lr,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyljbenzamide
Figure imgf000106_0001
[0310] ABM-34: trans 1-(2-Hydroxyethyl)-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]-1H-pyrazole-4-carboxamide
Figure imgf000106_0002
[0311] ABM-35: trans 6-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]pyridine-3-carboxamide.
Figure imgf000106_0003
[0312] ABM-36: trans 4-[(5-Hydroxypentyl)amino]-N-[3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetrameth lc clobut l benzamide
Figure imgf000106_0004
[0313] ABM-37: trans tert-Butyl 2-({5-[(4-{[3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamo l hen l amino ent l ox acetate
Figure imgf000106_0005
[0314] ABM-38: tert-butyl trans-(3-(3-chloro-4-cyanophenoxy)-2,2- dimethylcyclobutyl)carbamate] and ABM-39: tert-butyl cis-(3-(3-chloro-4-cyanophenoxy)- 2,2-dimethylcyclobutyl)carbamate.
Figure imgf000107_0001
Figure imgf000107_0002
[0315] Step 1: Synthesis of ((vinyloxy)methyl)benzene (B).
[0316] To a stirred solution of potassium tert-butanolate (A) (23 g, 205 mmol) in tetrahydrofuran (120 ml) was added ((2-bromoethoxy)methyl)benzene (30 g, 140 mmol) in tetrahydrofuran (70 ml) at 0°C. The resulting mixture was allowed to warm to room temperature and stirred for 3 hours. TLC showed the reaction was complete. The mixture was partitioned between anhydrous dichloromethane (300 ml) and water (100 ml). The organic layer was collected, washed with brine (100 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to afford crude ((vinyloxy)methyl)benzene (14.8 g, yield 80%) as colorless oil. 1H NMR (400 Hz, CDCl3): ^ 4.09 (dd, J = 2.0, 6.8 Hz, 1H), 4.29-4.33 (m, 1H), 4.77 (s, 2H), 6.54-6.60 (m, 1H), 7.28-7.39 (m, 5H). Chemical Formula: C9H10O; Molecular Weight: 134.18.
[0317] Step 2: Synthesis of 3-(benzyloxy)-2,2-dimethylcyclobutanone (C).
[0318] To a stirred solution of benzyl vinyl ether (2 g, 15.2 mmol) and triethylamine (1.3 ml, 9.2 mmol) in anhydrous acetonitrile (6 ml) was added slowly a solution of isobutyryl chloride (0.8 ml, 7.6 mmol) in dry acetonitrile (3 ml) at reflux. The resulting mixture was refluxed for 0.5 hour. TLC showed the reaction was complete. The reaction mixture was allowed to cool to room temperature and partitioned between ethyl acetate (30 ml) and water (20 ml). The organic layer was collected, and the aqueous layer was extracted with ethyl acetate (20 ml x 2). The combined organic layers were washed with brine (20 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to afford a crude residue which was purified by silica gel flash chromatography (eluted with 5-10% ethyl acetate in hexane) to afford 3- (benzyloxy)-2,2-dimethylcyclobutanone (1.5 g, yield 50%) as colorless oil. 1H NMR (400 Hz, CDCl3): ^ 1.19 (s, 3H), 1.26 (s, 3H), 3.08-3.24 (m, 2H), 3.96-3.99 (m, 1H), 4.55 (s, 2H), 7.29- 7.39 (m, 5H). Chemical Formula: C13H16O2; Molecular Weight: 204.26.
[0319] Step 3: Synthesis of 3-(benzyloxy)-2,2-dimethylcyclobutanone oxime (D)
[0320] To a solution of hydroxylamine hydrochloride (3.0g, 44.1 mmol) in ethanol (100 ml) was added 3-(benzyloxy)-2,2-dimethylcyclobutanone (7.5 g, 36.7 mmol) and sodium acetate trihydrate (6.5 g, 47.7 mmol). The resulting mixture was stirred at room temperature for 2 hours. TLC showed the reaction was complete. The volatiles were evaporated under reduced pressure. The residue was taken up in ethyl acetate (80 ml) and water (50 ml). The organic layer was collected, and the aqueous layer was extracted with ethyl acetate (50 ml x 2). The combined organic layers were washed with brine (50 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give crude 3-(benzyloxy)-2,2-dimethylcyclobutanone oxime (8.3 g, crude) as colorless oil which was used in next step without further purification. LC_MS: (ES+): m/z 220.2 [M+H]+. tR = 2.550 min.
[0321] Step 4: Synthesis of 3-(benzyloxy)-2,2-dimethylcyclobutanamine (E)
[0322] To a solution of 3-(benzyloxy)-2,2-dimethylcyclobutanone oxime (8.3 g, crude) in tetrahydrofuran (75 ml) was added Ni/Al alloy (13.3 g, 155 mmol) under nitrogen atmosphere. The suspension was stirred at 60°C for 30 minutes. To the resulting mixture was added aqueous sodium hydroxide solution (6.9 g in 75 ml water, 172 mmol) dropwise to keep the mixture refluxing. After addition, the mixture was refluxed for another 2 hours. TLC showed the reaction was complete. The solid was removed through filtration and the filter cake was washed with tetrahydrofuran (10 ml x 2). The organic layer was separated, and the aqueous layer was extracted with ethyl acetate (75 ml). The combined organic layers were washed with brine (50 ml) and dried over sodium sulfate and concentrated to give 3-(benzyloxy)-2,2- dimethylcyclobutanamine (6.3 g, crude) as colorless oil. The crude product was used in next step without further purification.1HNMR shows it was a mixture of trans/cis isomers (ratio: cis: trans: 1:1). LC_MS: (ES+): m/z 206.3 [M+H]+. tR = 1.675 min. 1HNMR (400MHz, CDCl3): Trans isomer: ^ 1.01 (s, 3H), 1.11 (s, 3H), 2.22-2.28 (m, 1H), 2.44-2.53 (m, 1H), 3.11-3.14 (m, 1H), 3.73-3.75 (m, 1H), 4.39-4.74 (m, 2H), 7.25-7.36 (m, 5H). Cis isomer: ^ 1.02 (s, 3H), 1.11 (s, 3H), 1.57-1.62 (m, 1H), 1.80-1.86 (m, 1H), 2.58-2.65 (m, 1H), 3.41-3.45 (m, 1H), 4.39-4.74 (m, 2H), 7.25-7.36 (m, 5H). Chemical Formula: C13H19NO; Molecular Weight: 205.30.
[0323] Step 5: Synthesis of tert-butyl (3-(benzyloxy)-2,2-dimethylcyclobutyl)carbamate (F).
[0324] To a stirred solution of 3-(benzyloxy)-2,2-dimethylcyclobutanamine (6.3 g, crude) in tetrahydrofuran (70 ml) was added di-tert-butyl dicarbonate (7 ml, 30.7 mmol) slowly at room temperature. The reaction mixture was stirred at room temperature for 1 hour. TLC showed the reaction was complete. The volatiles were removed under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluted with 5-10% ethyl acetate in hexane) to afford tert-butyl (3-(benzyloxy)-2,2-dimethylcyclobutyl)carbamate (6.0 g, yield 53% over 3 steps) as colorless oil. LC_MS: (ES+): m/z 306.2 [M+H]+. tR = 3.167 min.
[0325] Step 6: Synthesis of tert-butyl (3-hydroxy-2,2-dimethylcyclobutyl)carbamate (G).
[0326] A mixture of tert-butyl (3-(benzyloxy)-2,2-dimethylcyclobutyl)carbamate (6.3 g, 20.6 mmol) and palladium on carbon (10%, 700 mg) in methanol (110 ml) was stirred at room temperature overnight under hydrogen atmosphere (hydrogen balloon). TLC showed the reaction was complete. Palladium on carbon was removed through filtration and the filter cake was washed with methanol (25 ml x 2). The combined filtrates were concentrated under reduced pressure to afford tert-butyl (3-hydroxy-2,2-dimethylcyclobutyl)carbamate (4.5 g, yield 95%) as white solid.1HNMR shows it was a mixture of trans/cis isomers (ratio: ~ 1:1). 1HNMR
(400MHz, CD3OD): Trans isomer: ^ 0.95 (s, 3H), 1.08 (s, 3H), 1.45 (s, 9H), 2.07-2.19 (m, 1H), 2.43-2.50 (m, 1H), 3.61-3.65 (m, 1H), 3.82-3.85 (m, 1H). Cis isomer: ^ 0.91 (s, 3H), 1.13 (s, 3H), 1.45 (s, 9H), 1.72-1.79 (m, 1H), 2.43-2.50 (m, 1H), 3.35-3.38 (m, 1H), 3.69-3.73 (m, 1H). Chemical Formula: C11H21NO3; Molecular Weight: 215.29.
[0327] Step 7: Synthesis of tert-butyl trans-(3-(3-chloro-4-cyanophenoxy)-2,2- dimethylcyclobutyl)carbamate (ABM-38) and tert-butyl cis-(3-(3-chloro-4-cyanophenoxy)-2,2- dimethylcyclobutyl)carbamate (ABM-39).
[0328] To a stirred solution of tert-butyl (3-hydroxy-2,2-dimethylcyclobutyl)carbamate (280 mg, 1.29 mmol) in N,N-dimethylformamide (6 ml) was added sodium hydride (60% in mineral oil, 103 mg, 2.58 mmol) at 0°C. The mixture was stirred at 0°C for 0.5 hour, followed by addition of 2-Chloro-4-fluorobenzonitrile (200 mg, 1.29 mmol); the resulting mixture was stirred at 0°C for 1 hour. TLC showed the reaction was complete. The reaction mixture was quenched with water (3 ml) at 0°C and extracted with ethyl acetate (5 ml x 3). The combined organic layers were washed with brine (15 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to afford a crude residue which was purified by silica gel flash column chromatography (eluted with 20% tert-Butyl methyl ether in hexane) to afford ABM-38 (110 mg, yield 26%) as white solid and ABM-39 (120 mg, yield 26%) as white solid. ABM-38: LC_MS: (ES+): m/z 351.2 [M+H]+. tR = 3.222 min. 1HNMR (400MHz, CDCl3): ^ 1.15 (s, 3H), 1.18 (s, 3H), 1.45 (s, 9H), 2.17-2.24 (m, 1H), 2.39-2.46 (m, 1H), 3.96-4.07 (m, 1H), 4.29-4.32 (m, 1H), 4.59-4.67 (m, 1H), 6.75 (dd, J = 2.4, 8.8 Hz, 1H), 6.89 (d, J = 2.0 Hz, 1H), 7.54 (d, J = 8.8 Hz, 1H). Chemical Formula: C18H23ClN2O3; Molecular Weight: 350.84. ABM-39: LC_MS: (ES+): m/z 351.2 [M+H]+. tR = 3.173 min. 1HNMR (400MHz, CDCl3): ^ 1.03 (s, 3H), 1.32 (s, 3H), 1.45 (s, 9H), 1.80-1.87 (m, 1H), 2.79-2.86 (m, 1H), 3.64-3.72 (m, 1H), 4.16-4.20 (m, 1H), 4.57-4.59 (m, 1H), 6.77 (dd, J = 2.0, 8.8 Hz, 1H), 6.89 (d, J = 2.4 Hz, 1H), 7.54 (d, J = 8.4 Hz, 1H). Chemical Formula: C18H23ClN2O3; Molecular Weight: 350.84.
[0329] The experimental procedure used to make ABM-30, may be used to synthesize the free amine, which is used for further coupling. [0330] Synthesis of ULM Moieties
[0331] ULM-1: (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4- methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide
Figure imgf000111_0001
[0332] Step 1: Synthesis of 4-(4-methyl-1,3-thiazol-5-yl)benzonitrile (G)
[0333] To a stirred solution of 4-bromobenzonitrile (E, 20 g, 109.88 mmol) in DMA (250 mL) under a nitrogen atmosphere was added 4-methyl-1,3-thiazole (F, 21.88 g, 220.67 mmol), palladium (II) acetate (743 mg, 3.31 mmol) and potassium acetate (21.66 g, 220.71 mmol) at room temperature. The resulting solution was heated to 150 oC and stirred at this temperature for 5 hours, LC-MS indicated formation of the desired product. The reaction was cooled to room temperature, diluted with 1 L of water and extracted with ethyl acetate (300 mL x 3). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (200 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent: ethyl
acetate/petroleum ether, v: v = 1:5) to give the G (yield: 91%) as a white solid.
[0334] Step 2: Synthesis of [4-(4-methyl-1,3-thiazol-5-yl)phenyl]methanamine (H)
[0335] To a stirred solution of 4-(4-methyl-1,3-thiazol-5-yl)benzonitrile (G, 35.0 g, 174.8 mmol) in tetrahydrofuran (1000 mL) was added LiAlH4 (20.0 g, 526.3 mmol) in portions at 0oC in 10 min under a nitrogen atmosphere. The resulting solution was then stirred at 60 oC for 3h. LC-MS indicated formation of the desired product. The reaction was then cooled to 0oC, quenched by the addition water (20 mL, added slowly), aq. solution of NaOH(15%, 20 mL) and water (60 mL). The resulting mixture was then extracted with ethyl acetate (300 mL x 2). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (100 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent:
dichloromethane/methanol (v:v = 10:1)) to give H (yield: 56%) as a yellow oil.
[0336] Step 3: Synthesis of tert-butyl (2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol- 5-yl)phenyl]methyl}carbamoyl)pyrrolidine-1-carboxylate (J)
[0337] To a stirred solution of (2S,4R)-1-[(tert-butoxy)carbonyl]-4-hydroxypyrrolidine- 2-carboxylic acid (I, 2.7 g, 11.7 mmol) in N,N-dimethylformamide (20 mL) was added DIEA (2.52 g, 19.50 mmol), HATU (4.47 g, 11.76 mmol) and [4-(4-methyl-1,3-thiazol-5- yl)phenyl]methanamine (H, 2.0 g, 9.79 mmol) at room temperature. The resulting mixture was stirred at room temperature overnight, LC-MS indicated formation of the desired product. The reaction mixture was diluted with water (20 mL) and extracted with ethyl acetate (50 mL x 3). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (50 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent:
dichloromethane/methanol (v:v = 20:1)) to give J (yield: 56%) as a yellow solid.
[0338] Step 4: Synthesis of (2S,4R)-4-hydroxy-N-{[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}pyrrolidine-2-carboxamide hydrochloride (K)
[0339] To a stirred solution of tert-butyl (2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3- thiazol-5-yl)phenyl]methyl}carbamoyl)pyrrolidine-1-carboxylate (J, 45 g, 107.78 mmol), was added a solution of hydrogen chloride in dioxane (4N, 300 mL) . The resulting solution was stirred at 20 oC for 2 hours. The solids were collected by filtration to give K (yield: 98%) as a yellow solid, which was used for the next step without any further purification.
[0340] Step 5: Synthesis of tert-butyl N-[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl- 1,3-thiazol-5-yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamate (M)
[0341] To a stirred solution of (2S)-2-{[(tert-butoxy)carbonyl]amino}-3,3- dimethylbutanoic acid (L, 15.7 g, 68.0 mmol) in N,N-dimethylformamide (500 mL) was added DIEA (29.2 g, 225.9 mmol), HATU (25.9 g, 68.1 mmol) and (2S,4R)-4-hydroxy-N-{[4-(4- methyl-1,3-thiazol-5-yl)phenyl]methyl} pyrrolidine-2-carboxamide hydrochloride (K, 20.0 g, 56.5 mmol) at room temperature. The resulting solution was stirred at room temperature for 16 hours, LC-MS indicated formation of the desired product. The reaction mixture was diluted by water (200 mL) and extracted with ethyl acetate (200 mL x 3). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (50 mL x 2), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v = 2:1)) to give M (yield: 51%) as a yellow solid.
[0342] Step 6: Synthesis of (2S,4R)-1-[(2S)-2-amino-3,3-dimethylbutanoyl]-4-hydroxy- N-{[4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl}pyrrolidine-2-carboxamide hydrochloride (ULM-1)
[0343] To a stirred solution of tert-butyl N-[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4- methyl-1,3-thiazol-5-yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamate (M, 12 g, 22.61 mmol) in dioxane (20 mL) was added a solution of hydrogen chloride in dioxane (4N, 80 mL) at room temperature. The resulting solution was stirred at room temperature for 2 h, LC-MS indicated formation of the desired product. Precipitated solids were collected by filtration to give ULM-1 (yield: 48%) as a yellow solid. 1HNMR (400 MHz, CD3OD): ^ 9.84-9.82 (s, 1H), 7.58-7.54 (m, 4H), 4.71-4.41 (m, 4H), 4.13-4.08 (m, 1H), 3.86- 3.71 (m, 2H), 3.36 (s, 1H), 2.60-2.58 (s, 3H), 2.35-2.07 (m, 2H), 1.19-1.12(m, 9H). LC-MS (ES+): m/z 431.11 [MH+], tR = 0.73 min (2.0 minute run).
[0344] ULM-2: (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4- (thiazol-5-yl)benzyl)pyrrolidine-2-carboxamide:
Figure imgf000113_0001
.
[0345] ULM-2 was synthesized according to similar procedure described above for the synthesis of ULM-1, utilizing 4-bromobenzonitrile and 1,3-thiazole as starting materials. LC-MS (ES+): m/z 417.10 [MH+], tR = 0.51 min (2.0 minute run).
[0346] ULM-3: (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-((S)-1- (4-(4-methylthiazol-5-yl)phenyl)ethyl)pyrrolidine-2-carboxamide:
[0347]
Figure imgf000114_0001
.
[0348] Step 1: Synthesis of tert-butyl N-[(1S)-1-(4-bromophenyl)ethyl]carbamate (O)
[0349] To a stirred mixture of (1S)-1-(4-bromophenyl)ethan-1-amine (N, 10.0 g, 49.98 mmol) in dichloromethane (100 mL) was added Et3N (10.0 g, 99.01 mmol) and (Boc)2O (13.0 g, 59.63 mmol). The resulting mixture was stirred at room temperature for 2 hours. The bulk of solvent was then removed under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether, v: v = 1:10) to give O (yield: 99%) as a white solid.
[0350] Step 2: Synthesis of tert-butyl N-[(1S)-1-[4-(4-methyl-1,3-thiazol-5- yl)phenyl]ethyl]carbamate (P)
[0351] To a stirred solution of tert-butyl N-[(1S)-1-(4-bromophenyl)ethyl]carbamate (O, 15.0 g, 49.97 mmol) in DMA (100 mL), under an atmosphere of nitrogen, was added 4-methyl- 1,3-thiazole (9.9 g, 99.84 mmol), potassium acetate (9.8 g, 99.86 mmol) and Pd(OAc)2 (112.5 mg, 0.50 mmol) at room temperature. The resulting mixture was then stirred at 120oC for 2 hours. The reaction mixture was then cooled to room temperature, diluted by water (120mL), and extracted with ethyl acetate (200 mL x 3). The organic layers were combined, dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether, v: v = 1:5) to give P (yield: 47%) as a white solid. LC-MS (ES+): m/z 319.13 [MH+], tR = 0.97 min (2.0 minute run).
[0352] Step 3. Synthesis of (1S)-1-[4-(4-methyl-1,3-thiazol-5-yl)phenyl]ethan-1-amine hydrochloride (Q)
[0353] To a stirred solution of tert-butyl N-[(1S)-1-[4-(4-methyl-1,3-thiazol-5- yl)phenyl]ethyl]carbamate (P, 7.5 g, 23.55 mmol) in methanol (20 mL) was bubbled in hydrogen chloride (gas) at room temperature for 2 hours. Then the resulting mixture was concentrated under vacuum to give Q (yield: 86%) as a white solid, which was used in the next step without any further purifications.
[0354] Intermediate Q was converted to ULM-3 in a similar manner as described for the conversion of H to ULM-1.1H NMR (300MHz, DMSO): ^ 8.99 (s, 1 H), 8.57-8.55 (d, J = 7.8 Hz, 1 H), 8.01 (br. s, 3 H), 7.46-7.43 (d, J = 8.4 Hz, 2 H), 7.39-7.37 (d, J = 8.4 Hz, 2 H), 4.98- 4.90 (m, 1 H), 4.57-4.51 (m, 1 H), 4.34 (br. s, 1 H), 3.94-3.92 (m, 1 H), 3.69-3.66 (m, 1 H), 3.53- 3.49 (m, 1 H), 2.52 (s, 3 H), 2.10-2.07 (m, 1 H), 1.83-1.81 (m, 1 H), 1.40-1.30 (m, 3 H), 1.03 (s, 9 H). LC-MS (ES+): m/z 445.05 [MH+], tR = 0.53 min (2.0 minute run).
[0355] ULM-4: (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4- (oxazol-5-yl)benz l rrolidine-2-carboxamide h drochloride:
Figure imgf000115_0001
.
[0356] Step 1: 1. Synthesis of 4-(1,3-oxazol-5-yl)benzonitrile (S)
[0357] To a stirred solution of 4-formylbenzonitrile (R, 1.0 g, 7.63 mmol) in methanol (40 mL) was added [[(4-methylbenzene)sulfonyl]methyl](methyliumylidyne)azanuide (1.6 g, 8.40 mmol) and potassium carbonate (1.4 g, 9.91 mmol), the resulting mixture was stirred at room temperature for 1.5 hours. The bulk of solvent was then removed under reduced pressure. The residue was diluted with saturated aqueous sodium bicarbonate (20 mL) and was extracted with dichloromethane (30 mL x 3). The organic layers were combined, washed with brine (30 mL), dried over anhydrous sodium sulfate and concentrated under vacuum to give a crude product, which was purified by re-crystallization using dichloromethane and hexane to give S (1.0 g) as a white solid.1H NMR (400 MHz, DMSO) ^ 8.56 (s, 1H), 7.97-7.83 (m, 5H); LC-MS (ES+): m/z 170.95 [MH+], tR = 0.79 min (2.0 minute run).
[0358] Step 2. Synthesis of [4-(1,3-oxazol-5-yl)phenyl]methanamine (T)
[0359] To a stirred solution of 4-(1,3-oxazol-5-yl)benzonitrile (S, 900.0 mg, 5.29 mmol) in methanol (15 mL) was added Raney-Ni (900 mg) and aq. ammonium hydroxide (3.0 mL). Hydrogen gas was then introduced into the reaction mixture via a balloon. The resulting mixture was stirred at room temperature for 16 hours. The solids were then removed by filtration and the solution was concentrated under vacuum to give T (yield: 81%) as brown oil, which was used in the next step without any further purifications. LC-MS (ES+): m/z 175.90 [MH+], tR = 0.26 min (2.0 minute run).
[0360] Intermediate T was converted to ULM-4 in a similar manner as described for the conversion of H to ULM-1. LC-MS (ES+): m/z 400.96 [MH+], tR = 0.66 min (2.0 minute run).
[0361] ULM-5: (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4- methyloxazol-5-yl)benzyl)pyrrolidine-2-carboxamide:
Figure imgf000116_0001
.
[0362] [4-(4-methyl-1,3-oxazol-5-yl)phenyl]methanamine (V) was synthesized according to similar procedure described above for the synthesis of [4-(1,3-oxazol-5- yl)phenyl]methanamine (T).
[0363] Intermediate V was converted to ULM-5 in a similar manner as described for the conversion of H to ULM-1. LC-MS (ES+): m/z 415.10 [MH+], tR = 1.17 min (2.6 minute run). [0364] ULM-6: (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-N-(4-chlorobenzyl)-4- hydroxypyrrolidine-2-carboxamide hydrochloride:
Figure imgf000117_0001
.
[0365] ULM-6 was synthesized according to similar procedure described above for the synthesis of ULM-1, utilizing 4-chlorobenzonitrile as the starting material.
[0366] ULM-7: (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-N-(4-cyanobenzyl)-4-
Figure imgf000117_0002
.
[0367] ULM-7 was synthesized according to similar procedure described above for the synthesis of ULM-1, utilizing 4-cyanobenzonitrile as the starting material.
[0368] ULM-8: (2S,4R)-1-((S)-2-amino-3-methylbutanoyl)-4-hydroxy-N-(4-(4-
Figure imgf000117_0003
[0369] ULM-8 was synthesized according to similar procedure described above for the synthesis of ULM-1, utilizing (S)-2-(tert-butoxycarbonylamino)-3-methylbutanoic acid and 4- methyl-1,3-thiazole (F) as starting materials.
[0370] ULM-9: (2S,4R)-1-((S)-2-amino-3-methylbutanoyl)-4-hydroxy-N-(4-(thiazol- 5-yl)benzyl)pyrrolidine-2-carboxamide hydrochloride: .
[0371] ULM-9 was synthesized according to similar procedure described above for the synthesis of ULM-1, utilizing (S)-2-(tert-butoxycarbonylamino)-3-methylbutanoic acid and 1,3- thiazole as starting materials.
[0372] ULM-10: (2S,4R)-1-((S)-2-amino-3-methylbutanoyl)-4-hydroxy-N-(4-(4- methyloxazol-5-yl)benzyl)pyrrolidine-2-carboxamide h drochloride:
Figure imgf000118_0001
[0373] ULM-10 was synthesized according to similar procedure described above for the synthesis of ULM-5, utilizing (S)-2-(tert-butoxycarbonylamino)-3-methylbutanoic acid as starting material.
[0374] ULM-11: (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(1- methyl-1H-pyrazol-5-yl)benzyl)pyrrolidine-2-carboxamide hydrochloride:
Figure imgf000118_0002
.
[0375] ULM-11 was synthesized according to similar procedure described above for the synthesis of ULM-1, utilizing 1-methylpyrazole as the starting material.
[0376] ULM-12: (2S,4R)-4-tert-butoxy-N-(2-hydroxy-4-(4-methylthiazol-5- yl)benzyl)-1-((S)-3-methyl-2-(1-oxoisoindolin-2-yl)butanoyl)pyrrolidine-2-carboxamide:
Figure imgf000119_0001
.
[0377] Step 1: Synthesis of 2-hydroxy-4-(4-methyl-1,3-thiazol-5-yl)benzonitrile (BH)
[0378] To a stirred solution of 4-bromo-2-hydroxybenzonitrile (BG, 28 g, 141.40 mmol) in DMA (300 mL) was added 4-methyl-1,3-thiazole (28.1 g, 283.40 mmol), potassium acetate (28 g, 285.31 mmol) and palladium (II) acetate (940 mg, 4.19 mol) at room temperature under an atmosphere of nitrogen. The resulting mixture was then heated to 150 oC and stirred at this temperature for 2.5 h, LC-MS indicated formation of the desired product. The reaction was then cooled to room temperature, diluted by water (1000 mL) and then extracted with ethyl acetate (500 mL x 3). The organic layers were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a crude residue, which was purified by a flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v : v = 1 : 1) to give BH (yield: 78%) as a yellow solid. LC-MS (ES+): m/z 216.95 [MH+], tR = 1.25 min (2.6 minute run).
[0379] Step 1: 2-(aminomethyl)-5-(4-methyl-1,3-thiazol-5-yl)phenol (BI)
[0380] To a stirred solution of 2-hydroxy-4-(4-methyl-1,3-thiazol-5-yl)benzonitrile (BH, 15.6 g, 72.14 mmol) in tetrahydrofuran (400 mL) under an atmosphere of nitrogen was added LiAlH4 (11 g, 289.86 mmol) in several portions at 10 oC. The resulting mixture was then heated to reflux for 3 h, LC-MS indicated formation of the desired product. The reaction was then cooled to 0 oC, quenched by the water (10 mL, added slowly and drop wise), 15% NaOH (aq.) (30 mL) and water (10 mL). The solids precipitated were removed by filtration, the solution phase was concentrated under reduced pressure followed by high vacuum pump to give BI
(yield: 65%). LC-MS (ES+): m/z 220.85 [MH+], tR = 1.02 min (2.6 minute run).
[0381] Step 3. Synthesis of 9H-fluoren-9-ylmethyl (2S,4R)-4-(tert-butoxy)-2-({[2- hydroxy-4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl}carbamoyl)pyrrolidine-1-carboxylate (BJ)
[0382] To a stirred solution of (2S,4R)-4-(tert-butoxy)-1-[(9H-fluoren-9- ylmethoxy)carbonyl]pyrrolidine-2-carboxylic acid (BI, 18.6 g) in N,N-dimethylformamide (250 mL) was added DIEA (7.9 g, 61.24 mmol), HATU (17.3 g, 45.53 mmol) and 2-(aminomethyl)-5- (4-methyl-1,3-thiazol-5-yl)phenol (20 g, 90.79 mmol) at room temperature. The resulting mixture was stirred overnight at room temperature, and LC-MS indicated formation of the desired product. The reaction mixture was diluted by water (200 mL) and then extracted with ethyl acetate (300 mL x 3). The organic layers were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent: dichloromethane/methanol (v: v = 25:1)) to give BJ (yield: 31%) as a yellow oil. LC-MS (ES+): m/z 611.20 [MH+], tR = 1.12 min (2.0 minute run).
[0383] Step 4: Synthesis of (2S,4R)-4-(tert-butoxy)-N-{[2-hydroxy-4-(4-methyl-1,3- thiazol-5-yl)phenyl]methyl}pyrrolidine-2-carboxamide (BK)
[0384] To a stirred solution of 9H-fluoren-9-ylmethyl (2S,4R)-4-(tert-butoxy)-2-({[2- hydroxy-4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl}carbamoyl)pyrrolidine-1-carboxylate (BJ, 17.2 g, 28.12 mmol) in dichloromethane (270 mL) was added piperidine (30 mL, 280.00 mmol) at room temperature. The resulting solution was stirred at room temperature for 3 h, and LC- MS indicated formation of the desired product. The reaction mixture was concentrated under vacuum to give a crude residue, which was then diluted by dichloromethane (300 mL), washed with water (300 mL x 2), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent: dichloromethane/methanol (v: v = 20:1)) to give BK (yield: 71%) as a yellow oil. LC-MS (ES+): m/z 389.95 [MH+], tR = 0.88 min (2.0 minute run).
[0385] Step 5: Synthesis of (2S,4R)-4-(tert-butoxy)-N-{[2-hydroxy-4-(4-methyl-1,3- thiazol-5-yl)phenyl]methyl}-1-[(2S)-3-methyl-2-(1-oxo-2,3-dihydro-1H-isoindol-2- yl)butanoyl]pyrrolidine-2-carboxamide ULM-12) [0386] To a stirred solution of (2S)-3-methyl-2-(1-oxo-2,3-dihydro-1H-isoindol-2- yl)butanoic acid (3.6 g, 15.43 mmol) in N,N-dimethylformamide (50 mL) was added DIEA (2.7 g, 20.93 mmol), HATU (5.89 g, 15.49 mmol) and (2S,4R)-4-(tert-butoxy)-N-{[2-hydroxy-4-(4- methyl-1,3-thiazol-5-yl)phenyl]methyl}pyrrolidine-2-carboxamide (BK, 4.0 g, 10.27 mmol) at room temperature. The resulting solution was stirred overnight at room temperature, and LC- MS indicated formation of the desired product. The reaction was diluted by the water (100 mL) and extracted with dichloromethane (100 mL x 3). The organic layers were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a crude residue, which was purified by a flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v = 2:1)) to give ULM-12 (yield: 43%) as a yellow solid.1H NMR (400 MHz, CD3OD) ^ 8.88 (s, 1 H), 7.83-7.81 (d, J = 7.6 Hz, 1 H), 7.66-7.63 (m, 2 H), 7.61-7.59 (m, 1 H), 7.36-7.34 (d, J = 8.0 Hz, 1 H), 6.94-6.87 (d, J = 6.4 Hz, 1 H), 4.88 (s, 1 H), 4.56-4.39 (m, 6 H), 3.88-3.81 (m, 2 H), 2.51 (s, 3 H), 2.47-2.45 (m, 1 H), 2.15-2.13 (m, 2 H), 1.16-1.14 (d, J = 6.4 Hz, 3 H) 1.02 (s, 9 H), 0.89-0.86 (d, J = 6.4 Hz, 3 H); LC-MS (ES+): m/z 605.40 [MH+], tR = 1.91 min (3.6 minute run).
[0387] Unless otherwise noted, the following intermediates and their analogs (for examples, but not limited to, analogs with substitutions such as halogens) were synthesized according to similar procedures described above for the synthesis of ULM-12, by utilizing corresponding starting materials and reagents.
[0388] ULM-13: (2S,4R)-4-tert-butoxy-1-((S)-2-(6-fluoro-1-oxoisoindolin-2-yl)-3- methylbutanoyl)-N-(2-hydroxy-4-(4-meth lthiazol-5- l)benzyl)pyrrolidine-2-carboxamide:
Figure imgf000121_0001
.
[0389] ULM-14: (2S,4R)-4-tert-butoxy-1-((S)-2-(7-cyano-1-oxoisoindolin-2-yl)-3- methylbutanoyl)-N-(2-hydroxy-4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide:
Figure imgf000122_0001
.
[0390] ULM 15: (2S,4R)-1-((S)-2-Amino-3,3-dimethylbutanoyl)-4-hydroxy-N-((R)- 2-hydroxy-1-(4-(4-methylthiazol-5-yl)phenyl)ethyl)pyrrolidine-2-carboxamide
hydrochloride.
Figure imgf000122_0002
Figure imgf000122_0003
[0391] Step 1: Synthesis of 1-(4-Bromophenyl)-2-hydroxyethanone (B).
[0392] To a suspension of 2-bromo-1-(4-bromophenyl)ethanone (A) (60.0 g, 0.217 mmol) in EtOH (85%, 600 mL) was added NaOCHO (44.37 g, 0.652 mol) at room temperature. The mixture was heated to 110oC until all solid was dissolved and stirred for another 3 hours. Then it was reduced to one-third of its volume in vacuo. The residue was poured into ice water (200 mL). After stirring for 30 minutes, the resulting off-white precipitate was filtered, washed with cold water (100 mL), and dried to afford the desired product (B) (39.0 g, 84%) as a white solid.1H NMR (400 MHz, CDCl3): ^ 7.79 (d, J = 8.8 Hz, 2H), 7.66 (d, J = 8.8 Hz, 2H), 4.85(s, 2H), 3.43(t, J = 4.4 Hz, 1H).
[0393] Step 2: Synthesis of 1-(4-Bromophenyl)-2-(tert-butyldimethylsilyloxy)ethanone (C).
[0394] To a solution of (B) (39.0 g, 0.181 mol) and imidazole (37.0 g, 0.544 mol) in DCM (500 mL) was added TBSCl ( 32.75 g, 0.218 mmol) at 0oC. The reaction mixture was warmed to room temperature and stirred for another 3 hours. After it was quenched with H2O (200 mL), the organic phase was washed with brine (100 mL x 3), dried over Na2S2O4 and concentrated under vacuum to afford the desired product (C) (55.0 g, crude, 92%) as a white solid.1H NMR (400 MHz, CDCl3): ^ 7.70 (d, J = 8.4 Hz, 2H), 7.49 (d, J = 8.8 Hz, 2H), 4.73 (s, 2H), 1.46 (d, J = 6.4 Hz, 1H), 0.81 (s, 9H), 0.00 (s, 6H).
[0395] Step 3: Synthesis of (S,Z)-N-(1-(4-Bromophenyl)-2-(tert- butyldimethylsilyloxy)ethylidene)-2-methylpropane-2-sulfinamide (D).
[0396] To a solution of (C) (55.0 g, 0.167 mmol) and (S)-2-methylpropane-2-sulfinamide (30.36 g, 0.25 mmol) in THF (600 mL) was added Ti(OiPr)4 (142.4 g, 0.501 mol) at 25oC. The mixture was heated to 80oC overnight. After it was cooled to 0 oC, the reaction was quenched by addition of H2O (100 mL). The mixture was filtered through Celite, and the solid was washed with EtOAc (1000 mL). The combined organic layers were washed with brine (200 mL x 3), and concentrated under vacuum. The residue was purified by silica gel column chromatography with EtOAc/PE (1/100~10) to afford the desired product (D) (20.0 g, 41%) as a brown syrup.1H NMR (400 MHz, CDCl3): ^ 7.66 ( br, 2H), 7.47 (d, J = 8.4 Hz, 2H), 4.92-4.96 ( m, 1H), 3.63- 3.69 (m, 1H), 1.24 (s, 9H), 0.76 (s, 9H), 0.03 (d, J = 5.2 Hz, 6H.
[0397] Step 4: Synthesis of (S)-N-((R)-1-(4-Bromophenyl)-2-(tert- butyldimethylsilyloxy)ethyl)-2-methylpropane-2-sulfinamide. (E).
[0398] To a suspension of (D) (18.0 g, 41.61 mmol ) in THF (100 mL) was added BH3/THF(104 mL, 1.0 M, 104 mmol) at -78oC. The resulting solution was stirred -78 oC for 4 hours. It was quenched by addition of acetone at -78 oC (20 mL), and H2O (20 mL) subsequently. The organic phase was washed with brine, dried over Na2SO4, and concentrated under vacuum. The residue was purified by silica gel column chromatography with PE/EA (10~5 /1) to afford the desired product (E) (15.0 g, 83% ) as a light brown solid.1H NMR (400 MHz, CDCl3): ^ 7.52 (d, J = 8.4Hz, 2H), 7.431 (d, J = 8.4 Hz, 2H), 4.44-4.48 (m, 1H), 4.16 (m, 1H), 3.83-3.90 (m, 2H), 2.11 (t, J = 2.4 Hz, 1H), 1.27 (s, 9H), 0.90 (s, 9H), 0.01 (d, J = 5.2 Hz, 6H).
[0399] Step 5: Synthesis of (R)-Tert-butyl 1-(4-bromophenyl)-2-hydroxyethylcarbamate (F).
[0400] A solution of (E) (10.0 g, 23.0 mmol) in HCl(g)/CH3OH (50 mL) was stirred at room temperature for 3 hours. The solvent was removed under vacuum to afford the desired product of (R)-2-amino-2-(4-bromophenyl)ethanol hydrochloride (6.0 g, crude). To a solution of (R)-2-amino-2-(4-bromophenyl)ethanol hydrochloride in CH3OH (50 mL) were added TEA (11.6 g, 116 mmol) and Boc2O (7.5 g, 45.0 mmol) subsequently at 0oC. The resulting solution was stirred for 3 hours. The solvent was removed under vacuum. The residue was taken up with EtOAc (200 mL), and the mixture was washed with brine (100 mL x 2), dried over Na2SO4, and concentrated under vacuum. The residue was purified by silica gel column chromatography (ethyl acetate/petroleum ether = 1/4~1) to afford the desired product (F) (6.0 g, 82% yield) as a colorless oil.1H NMR (400 MHz, DMSO-d6): ^ 7.50 (d, J = 8.4 Hz, 2H), 7.25 (d, J = 8.4 Hz, 2H), 4.81 (s, 1H), 4.48-4.50 (m, 1H), 3.47-.349 (m, 2H), 1.40 (s, 9H).
[0401] Step 6: Synthesis of (R)-Tert-butyl 2-hydroxy-1-(4-(4-methylthiazol-5- yl)phenyl)ethylcarbamate (G).
[0402] To a solution of (F) (6.0 g, 18.97mmol) in DMA (50 mL) were added 4- methylthiazole (3.79 g, 38.2 mmol), KOAc (3.72 g, 38.5 mmol) and Pd(OAc)2 (426 mg, 1.90 mmol) subsequently. The solution was stirred at 130oC for 5 h under N2 atmosphere. After cooling to room temperature, the mixture was filtered through Celite, and the solid was washed with EtOAc (200 mL). The resulting solution was washed with brine (100 mL × 3). The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated under vacuum. The residue was purified by silica gel column chromatography (ethyl acetate/petroleum ether = 1/4~1) to afford (G) (2.0 g, 32% yield) as a light brown solid.
[0403] Step 7: Synthesis of (R)-2-Amino-2-(4-(4-methylthiazol-5-yl)phenyl)ethanol hydrochloride (H).
[0404] To a solution of (G) (2.0 g, 5.98 mmol) in DCM (20 mL) was added HCl (g) 4M dioxene (10 mL) at 25oC. The resulting solution was stirred at room temperature for 2 hours. The solvent was removed under vacuum to afford the desired product (H) (1.1 g, 68%) as a light yellow solid.1H NMR (400 MHz, CD3OD): ^ 9.20 (s, 1H), 7.58-7.63 (m, 4H), 4.42-4.45 (m, 1H), 3.93-3.97 (m, 1H), 3.82-3.87 (m, 1H), 2.53 (s 3H).
[0405] Step 8: Synthesis of tert-butyl (S)-1-((2S,4R)-2-(((R)-2-Hydroxy-1-(4-(4- methylthiazol-5-yl) phenyl)ethyl)carbamoyl)-4-hydroxypyrrolidin-1-yl)-3,3-dimethyl-1- oxobutan-2-ylcarbamate (I).
[0406] To a solution of (H) (1.1 g, 4.06 mmol), (2S,4R)-1-((S)-2-(tert-butoxycarbonyl)- 3,3-dimethylbutanoyl)-4-hydroxypyrrolidine-2-carboxylic acid (2.10 g, 6.09 mmol) and DIEA (1.60 g, 13.0 mmol) in DCM (20 mL) was added HATU (2.30 g, 6.08 mmol) at 25oC. The reaction mixture was stirred at 25oC for 2 hours. Then it was quenched by addition of H2O (20 mL). The mixture was washed with brine, dried over Na2SO4, and concentrated under vacuum. The residue was purified by column chromatography with DCM/CH3OH (30~20:1) as eluent to afford (I) (1.7 g, 75%) as a light yellow solid.1H NMR (400 MHz, CDCl3): ^ 8.68 (s, 1H), 7.28- 7.42 (m, 5H), 5.14-5.30 (m, 2H), 4.65 (t, J = 8.4 Hz, 1H), 4.52 (s, 1H), 4.21 (d, J = 9.2 Hz, 1H), 4.10-4.13 (m, 1H), 3.94 (m, 1H), 3.84 (m, 1H), 3.68-3.75 (m, 1H), 2.96 (s, 1H), 2.53 (s, 3H), 2.35 (m, 1H), 2.16-2.19 (m, 1H), 1.42 (s, 9H), 1.26 (s, 9H).
[0407] Step 9: Synthesis of (2S,4R)-1-((S)-2-Amino-3,3-dimethylbutanoyl)-4-hydroxy- N-((R)-2-hydroxy-1-(4-(4-methylthiazol-5-yl)phenyl)ethyl)pyrrolidine-2-carboxamide hydrochloride (ULM-15).
[0408] To a solution of (I) (1.7 g, 3.03 mmol) in CH3OH (20 mL) was added
AcCl/CH3OH (v:v=1:10, 10 mL, AcCl was added dropwise to the methanol solution at 0oC and the solution was stirred for 1 hour). The resulting solution was stirred at 25oC for 1 hour. Then the solvent was removed under vacuum to afford the desired product (ULM-15) (1.5 g, 94%) as a yellow solid.1H NMR (400 MHz, CD3OD): ^ 9.77 (s, 1H), 7.52-7.56 (m, 4H), 5.02 (t, J = 6.0 Hz, 1H), 4.73 (t, J = 8.0 Hz, 1H), 4.49 (s, 1H), 4.06 (s, 1H), 3.82 (d, J = 4.8 Hz, 3H), 3.65-3.69 (m, 1H), 2.58 (s, 3H), 2.28-2.34 (m, 1H), 1.94-2.01 (m, 1H), 1.14 (s, 9H). Chemical Formula: C23H32N4O4S / C23H32N4O4S HCl; Molecular Weight: 460.59 / 497.05. [0409] Synthesis of Linker Chemistry, L
[0410] L-1: 2-(3-(5-(tosyloxy)pentyloxy)propoxy)acetic acid
[0411]
Figure imgf000126_0001
.
[0412] Step 1: Synthesis of ({[5-(prop-2-en-1-yloxy)pentyl]oxy}methyl)benzene
[0413] To a stirred solution of 5-(benzyloxy)pentan-1-ol (W, 4.0 g, 20.59 mmol) in N,N- dimethylformamide (50 mL) was added sodium hydride (1.24 g, 51.67 mmol) in portions at 0 °C under an atmosphere of nitrogen. The resulting mixture was then stirred at room temperature for 1 hour. To this mixture was added 3-bromoprop-1-ene (3.71 g, 30.67 mmol), the reaction mixture was stirred overnight at 60 °C in an oil bath. LC-MS indicated formation of the desired product. The reaction mixture was cooled to 0 °C and then quenched by water (100 mL), the resulting mixture was extracted with ethyl acetate (200 mL x 2). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (60 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue. The residue was purified by a flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v = 1:40)) to give 4.57 g of X.1H NMR (300MHz, CDCl3): ^ 7.36(s, 4 H), 7.32 (m, 1 H), 5.98 (m, 1 H), 5.33 (m, 1H), 5.21 (m, 1H), 4.53 (s, 2H), 3.99 (m, 2H), 3.53 (m, 4H), 1.72 (m, 4H), 1.52 (m, 2H). LC-MS (ES+): m/z 235.00 [MH+], tR = 1.18 min (2.0 minute run).
[0414] Step 2: Synthesis of 3-{[5-(benzyloxy)pentyl]oxy}propan-1-ol (Y)
[0415] To a 250-mL round-bottom flask with 9-BBN (0.5 M in THF, 77 mL) was added a solution of ({[5-(prop-2-en-1-yloxy)pentyl]oxy}methyl)benzene (X, 3.0 g, 12.80 mmol) in anhydrous tetrahydrofuran (20 mL) with stirring at 0 °C under an atmosphere of nitrogen. The resulting solution was stirred overnight at room temperature. LC-MS indicated formation of the desired product. Methanol (15 mL, with 30% sodium hydroxide and 30% H2O2) was added to the reaction and the resulting mixture was stirred at room temperature for 2 hours. This mixture was then extracted with ethyl acetate (20 mL x 3). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (100 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue. The residue was purified by a flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v: v = 1:1)) to provide 1.96 g of Y as light yellow oil.1H NMR (300MHz, CDCl3): ^7.34 (m, 5H), 4.49 (s, 2H), 3.75 (m, 2H), 3.59 (m, 2H), 3.49 (m, 4H), 2.65 (bs, 1 H), 1.84 (m, 2H), 1.68 (m, 4H), 1.50 (m, 2H). LC-MS (ES+): m/z 253.17 [MH+], tR = 1.44 min (2.6 minute run).
[0416] Step 3: Synthesis of tert-butyl 2-(3-{[5-(benzyloxy)pentyl]oxy}propoxy)acetate (Z)
[0417] To a stirred solution of 3-{[5-(benzyloxy)pentyl]oxy}propan-1-ol (Y, 3.7 g, 14.66 mmol) in dichloromethane (30 mL) was added a solution of NaOH in water (37%, 30 mL) followed by tert-butyl 2-bromoacetate (11.39 g, 58.39 mmol) and TBACl (4.17 g). The resulting mixture was stirred at room temperature overnight. LC-MS indicated formation of the desired product. The reaction mixture was then extracted with ethyl acetate (50 mL x 3). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (60 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue. The residue was purified by a flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v = 1:2) to give 3.2g of Z as a yellow oil.1H NMR (400MHz, CDCl3): ^7.34(s, 4 H), 7.29 (m, 1 H), 4.50 (s, 4H), 4.3 (m, 2H), 3.51 (m, 4H), 3.42 (m, 2H), 1.98 (m, 2H), 1.67 (m, 4H), 1.48 (s, 9H), 1.46 (m, 2H). LC-MS (ES+): m/z 367.25 [MH+], tR = 1.28 min (2.0 minute run).
[0418] Step 4: Synthesis of tert-butyl 2-[3-[(5-hydroxypentyl)oxy]propoxy]acetate (AA)
[0419] To a stirred solution of tert-butyl 2-(3-{[5- (benzyloxy)pentyl]oxy}propoxy)acetate (Z, 3.2 g, 8.73 mmol) in methanol (30 mL) was added AcOH (1.5 mL), palladium on carbon (1.5 g) under an atmosphere of nitrogen. Hydrogen was then introduced to the reaction mixture via a hydrogen balloon, and the reaction was stirred at room temperature for 3h. The solid material was removed by filtration, the solution was concentrated under vacuum to provide 2.3 g of AA as light yellow oil, which was used for the next step without any further purifications. LC-MS (ES+): m/z 277.10 [MH+], tR = 0.86 min (2.0 minute run).
[0420] Step 5: Synthesis of tert-butyl 2-[3-({5-[(4- methylbenzenesulfonyl)oxy]pentyl}oxy)propoxy]acetate (AB) [0421] To a stirred solution of tert-butyl 2-[3-[(5-hydroxypentyl)oxy]propoxy]acetate (AA, 2.3 g, 8.32 mmol) in dichloromethane (30 mL) was added 4-methylbenzene-1-sulfonyl chloride (3.17 g, 16.63 mmol), triethylamine (2.52 g, 24.90 mmol) and 4-dimethylaminopyridine (203 mg, 1.66 mmol) at room temperature. The resulting mixture was stirred overnight at room temperature. The resulting mixture was concentrated under reduced pressure to give a crude residue, which was purified by a flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v = 1:2) to give 2.6 g of AB as a yellow oil. 1H NMR (300MHz, CDCl3): δ 7.77 (d, J = 8.1 Hz, 2 H), 7.36 (d, J = 8.1 Hz, 2 H), 4.51 (s, 2H), 4.31 (m, 2H), 4.13 (m, 2H), 3.52 (m, 4H), 2.05 (s, 3H), 1.97 (m, 2H), 1.69 (m, 4H), 1.48 (s, 9H), 1.46 (m, 2H).^LC-MS (ES+): m/z 431.20 [MH+], tR = 1.21 min (2.0 minute run).
[0422] Step 1: Synthesis of 2-[3-({5-[(4-methylbenzenesulfonyl)oxy]pentyl}oxy)propoxy]acetic acid (L-1)
[0423] To a stirred solution of tert-butyl 2-[3-({5-[(4- methylbenzenesulfonyl)oxy]pentyl}oxy)propoxy]acetate (AB, 1.3 g, 3.02 mmol) in
dichloromethane (10 mL) was added trifluoroacetic acid (10 mL) at room temperature. The resulting solution was stirred at room temperature for 3 hours. The reaction mixture was then concentrated under vacuum to give 1.5 g (crude) of L-1, which was used for next step without any further purification. LC-MS (ES+): m/z 375.34 [MH+], tR = 1.39 min (2.6 minute run).
[0424] The following Linkers (L) were prepared in a similar manner as for the preparation of L- 1.
[0425] L-2: 2-(3-(3,3-dimethyl-5-(tosyloxy)pentyloxy)propoxy)acetic acid
Figure imgf000128_0001
.
[0426] L-3: 2-(3-(3-hydroxy-5-(tosyloxy)pentyloxy)propoxy)acetic acid
Figure imgf000128_0002
.
[0427] L-4: 2-(2-(2-(2-(tosyloxy)ethoxy)ethoxy)ethoxy)acetic acid
Figure imgf000128_0003
. [0428] To a stirred solution of ethyl 2-[2-(2-{2-[(4- methylbenzenesulfonyl)oxy]ethoxy}ethoxy)ethoxy]acetate (AC, 2 g, 5.12 mmol, 1.00 equiv) in methanol (20 mL) was added a solution of NaOH (500 mg, 12.50 mmol) in water (4 mL), and the resulting mixture was stirred at room temperature for 2 hours. Aqueous hydrogen chloride (1 M) was then added to the reaction mixture to adjust pH to ~5. Solids precipitated were collected by filtration to give L-4 (yield: 98%). Mass (ES+): m/z 363, [MH+].
[0429] The following Linkers (L) were prepared in a similar manner as for the preparation of L- 4.
[0430] L-5: 2-(2-((2R,3R)-3-(2-(tosyloxy)ethoxy)butan-2-yloxy)ethoxy)acetic acid
Figure imgf000129_0001
.
[0431] L-6: 2-(2-((2S,3S)-3-(2-(tosyloxy)ethoxy)butan-2-yloxy)ethoxy)acetic acid
Figure imgf000129_0002
.
Figure imgf000129_0003
.
[0433] Step 1: Synthesis of 4-{4-[(4-methylbenzenesulfonyl)oxy]butoxy}butan-1-ol (AE)
[0434] To a stirred solution of 4-(4-hydroxybutoxy)butan-1-ol (AD, 2 g, 12.33 mmol) in dichloromethane (20 mL) was added Ag2O (4.25 g, 18.49 mmol), KI (409 mg, 2.46 mmol) and TsCl (2.345 g, 12.30 mmol). The resulting mixture was stirred at room temperature for 12 hours. The inorganic salt formed was removed by filtration and the organic solution was concentrated under reduced pressure to give a crude residue. The residue was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v = 1:1)) to give AE (yield: 28%) as a colorless oil. [0435] Step 2: Synthesis of ethyl 2-(4-{4-[(4-methylbenzenesulfonyl)oxy]butoxy}butoxy)acetate (AF)
[0436] To a stirred solution of 4-{4-[(4-methylbenzenesulfonyl)oxy]butoxy}butan-1-ol (AE, 1.1 g, 3.48 mmol) in dichloromethane (10 mL) was slowly added BF3.Et2O (49.4 mg, 0.35 mmol) followed by ethyl 2-diazoacetate (794 mg, 6.96 mmol) at 0 °C. The resulting mixture was stirred overnight at room temperature. The reaction was then quenched by water (2.0 mL). The resulting mixture was extracted with dichloromethane (50mL x 3), the organic layers were combined, dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue. The residue was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v: v = 1:4) to give AF (yield: 93 as light yellow oil. Mass (ES+): m/z 403.10 [MH+].
[0437] Step 3: Synthesis of 2-(4-{4-[(4-methylbenzenesulfonyl)oxy]butoxy}butoxy)acetic acid (L-7)
[0438] To a stirred solution of ethyl 2-(4-{4-[(4- methylbenzenesulfonyl)oxy]butoxy}butoxy)acetate (AF, 1.3 g, 3.23 mmol) in methanol (25mL) was added a solution of NaOH (388 mg, 9.70 mmol) in water (6 mL) at room temperature. The resulting solution was stirred at room temperature for 4 hours. The bulk of organic solvent was removed under reduced pressure, to the resulting mixture was added aqueous hydrogen chloride (1.0 M) to adjust the pH = ~5. The solution was then extracted with ethyl acetate (250 mL x 3), the organic layers were combined and dried over anhydrous sodium sulfate, concentrated under reduced pressure to give I-7 (yield: 93%) as light yellow oil. Mass (ES+): m/z 375.05 [MH+].
[0439] L-8: tert-but l 2- 3- 4- tos lox butox ro ox acetate
Figure imgf000130_0001
Figure imgf000130_0002
.
[0440] Step 1. Synthesis of 3-[4-(benzyloxy)butoxy]propan-1-ol (AH)
[0441] To a stirred solution of propane-1, 3-diol (1.52 g, 19.98 mmol) in N, N- dimethylformamide (20 mL) was added sodium hydride (840 mg, 35.00 mmol) at room temperature, the resulting mixture was stirred at room temperature for 30min. Then to the mixture was added 4-(benzyloxy) butyl 4-methylbenzene-1-sulfonate (AG, 6.68 g, 19.97 mmol) and the reaction was stirred overnight at 50 °C. TLC indicated formation of the desired product, at this time the reaction was allowed to cool down to room temperature. Water (10 mL) was added slowly to quench the reaction; the resulting mixture was then extracted with ethyl acetate (80 mL x 2). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (20 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v = 1:2)) to give AH (yield: 67%) as a light yellow oil. 1H NMR (300 MHz, CDCl3) ^ 7.38-7.29 (m, 5H), 4.52 (m, 2H), 3.80 (m, 2H), 3.61 (m, 2H), 3.49-3.46 (m, 4H), 2.04 (m, 2H), 1.82 (m, 2H), 1.68 (m, 2H))-Mass (ES+): m/z 239.05 [MH+].
[0442] Step 2. Synthesis of tert-butyl 2-[3-[4-(benzyloxy)butoxy]propoxy]acetate (AI).
[0443] To a stirred solution of 3-[4-(benzyloxy)butoxy]propan-1-ol (AH, 2.38 g, 9.99 mmol) in dichloromethane (15 mL) was added tert-butyl 2-bromoacetate (7.76 g, 39.78 mmol), TBAC (2.78 g, 10.00 mmol) followed by aqueous sodium hydroxide (37 %, 15 mL). The resulting mixture was stirred overnight at room temperature. The reaction mixture was then extracted with dichloromethane (100 mL x 3), the organic layers were combined, washed with saturated aqueous solution of sodium chloride (20 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue. The residue was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v = 1: 5)) to give AI (yield 57%) as a yellow oil. Mass (ES+): m/z 353.10 [MH+].
[0444] Step 3. Synthesis of tert-butyl 2-[3-(4-hydroxybutoxy)propoxy]acetate (AJ)
[0445] To a stirred mixture of tert-butyl 2-[3-[4-(benzyloxy)butoxy]propoxy]acetate (AI, 1 g, 2.84 mmol), palladium on carbon (10%, 200 mg) in methanol (20 mL) was added acetic acid (0.05 mL) under a nitrogen atmosphere. Hydrogen was then introduced to the reaction mixture via a balloon, the reaction was then stirred overnight at room temperature. The insoluble solids were removed by filtration and the solution phase was concentrated under reduced pressure to give the desired product (yield: 94%) as a yellow oil. Mass (ES+): m/z 263.05 [MH+].
[0446] Step 4. Synthesis of tert-butyl 2-(3-{4-[(4- methylbenzenesulfonyl)oxy]butoxy}propoxy)acetate (L-8) [0447] To a stirred solution of tert-butyl 2-[3-(4-hydroxybutoxy)propoxy]acetate (AJ, 700 mg, 2.67 mmol) in dichloromethane (10 mL) was added 4-methylbenzene-1-sulfonyl chloride (558.4 mg, 2.93 mmol), TEA (539.5 mg, 5.33 mmol) and 4-dimethylaminopyridine (32.6 mg, 0.27 mmol). The resulting mixture was stirred overnight at room temperature. The bulk of solvent was removed under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v= 1: 2)) to give titled product (yield: 52%) as a yellow oil.1H NMR (300 MHz, CDCl3) ^7.79 (d, J = 8.4 Hz, 2H), 7.35 (d, J = 8.0 Hz, 2H), 4.05 (m, 2H), 3.95 (s, 2H), 3.59 (m, 2H), 3.48 (m, 2H), 3.38 (m, 2H), 2.46 (s, 3H), 1.82 (m, 2H), 1.70 (m, 2H), 1.57 (m, 2H), 1.50 (s, 9H))-Mass (ES+): m/z 417.05 [MH+].
[0448]
Figure imgf000132_0001
.
[0450] L-9 was prepared in a similar manner as that used to prepare L-8, except that AK was used in place of AG. Mass (ES+): m/z 439.15 [MNa+].
[0451] L-10: tert-butyl 2-(6-(tosyloxy)hexa-2,4-diynyloxy)acetate
Figure imgf000132_0002
.
[0452] Step1: Synthesis of tert-butyl 2-[(6-hydroxyhexa-2,4-diyn-1-yl)oxy]acetate (AP)
[0453] To a stirred solution of hexa-2, 4-diyne-1, 6-diol (AO, 100 mg, 0.91 mmol) in N, N- dimethylformamide (5 mL) was added sodium hydride (32 mg, 1.33 mmol) at 0 oC. The resulting mixture was then warmed up to room temperature and stirred at room temperature for 30 min. The reaction mixture was cooled to 0 oC followed by addition of tert-butyl 2-bromoacetate (176 mg, 0.90 mmol), and the resulting mixture was stirred at 0 oC for 2h. LC-MS indicated formation of the desired product. The reaction was then quenched by water (10 mL, added slowly) at 0 oC, and was extracted with ethyl acetate (20 x 2 mL). The organic layers were combined, dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v = 1:2)) to give AP (yield: 49%) as a yellow oil.
[0454] Step 2. Synthesis of tert-butyl 2-({6-[(4-methylbenzenesulfonyl)oxy]hexa-2,4-diyn-1- yl}oxy)acetate (L-10)
[0455] To a stirred solution of tert-butyl 2-[(6-hydroxyhexa-2, 4-diyn-1-yl) oxy] acetate (AP, 50 mg, 0.22 mmol) in ether (2 mL) was added 4-toluenesulfonyl chloride (51 mg, 0.27 mmol) at 0 oC, followed by potassium hydroxide (125 mg, 2.23 mmol) in several batches at 0 oC. The resulting mixture was stirred at 0 oC for 4 hours. LC-MS indicated formation of the desired product. Water (10 mL) was added to the reaction, and the resulting mixture was extracted with ethyl acetate (20 mL x 2). The organic layers were combined, dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v = 1:2)) to give L- 10 (yield: 71%) as a yellow oil.1H NMR (300 MHz, CDCl3): ^ 7.83 (d, J = 6.0 Hz, 2H), 7.39 (d, J = 6.0 Hz, 2H), 4.79 (s, 2H), 4.37 (s, 2H), 4.05 (s, 2H), 2.48 (s, 3H), 1.51 (s, 9H); LC-MS (ES+): m/z 401.05 [MNa+], tR = 1.71 min (2.6 minute run).
[0456] The following Linkers (L) were prepared in a similar manner as for the preparation of L- 10.
[0457] L-11: tert-butyl 3-(6-(tos lox hexa-24-di n lox ropanoate
Figure imgf000133_0001
.
[0458] L-12: tert-butyl 4-(6-(tos lox hexa-24-di n lox butanoate
Figure imgf000133_0002
.
[0459] L-13: ethyl 2-(2-(2-aminoethoxy)ethoxy)acetate hydrochloride
Figure imgf000134_0001
Figure imgf000134_0002
.
[0460] Step 1: Synthesis of tert-butyl N-[2-(2-hydroxyethoxy)ethyl]carbamate (AR)
[0461] To a stirred solution of 2-(2-aminoethoxy)ethan-1-ol (AQ, 5.25 g, 49.94 mmol) in tetrahydrofuran (100 mL) was added aqueous solution of sodium bicarbonate (20% (w/w), 40 ml) and (Boc)2O (11.4 g, 52.23 mmol, added in several batches) at 0 °C. The resulting mixture was then warmed up slowly to room temperature and stirred at room temperature for 5h. The bulk of organic solvent was removed under reduced pressure and the resulting residue was diluted with water (300 mL), extracted with of ethyl acetate (100 mL x 3). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (20 mL x 2), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give AR (yield: 98%) as colorless oil.
[0462] Step 2: Synthesis of ethyl 2-[2-(2-{[(tert-butoxy)carbonyl]amino}ethoxy)ethoxy]acetate (AS)
[0463] To a stirred solution of tert-butyl N-[2-(2-hydroxyethoxy)ethyl]carbamate (AR, 4.0 g, 19.49 mmol) in dichloromethane (30 mL) was added 1-diazo-3-methoxypropan-2-one (3.34 g, 29.27 mmol) and BF3-Et2O (0.2 mL) at room temperature. The resulting solution was stirred at room temperature for 2 hours. Water (20 mL) was added to the reaction mixture, organic layer was separated and washed with brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a crude residue. The residue was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v: v = 1:2)) to give AS (yield: 18%) as yellow solid.1H NMR (400MHz, CDCl3): ^ 4.25-4.22 (q, J = 7.2 Hz, 2 H), 4.14 (s, 2 H), 3.74 (b, 2 H), 3.72 (b, 1 H), 3.67-3.32 (m, 4 H), 1.414 (s, 9 H), 1.31 (t, J = 7.2 Hz, 3 H).
[0464] Step 3: Synthesis of ethyl 2-[2-(2-aminoethoxy)ethoxy]acetate hydrochloride (L-13)
[0465] To a stirred solution of ethyl 2-[2-(2-{[(tert- butoxy)carbonyl]amino}ethoxy)ethoxy]acetate (AS, 500 mg, 1.72 mmol) in 1,4-dioxane (10 mL) was introduced hydrogen chloride (gas) via bubbling at room temperature for 2h. The solvent was then removed under vacuum to give L-13 (yield: 99%). LC-MS (ES+): m/z 192.00 [MH+], tR = 0.41 min (2.0 minute run).
[0466] L-14: ethyl 2-(5-aminopentyloxy)acetate
Figure imgf000135_0001
.
[0467] Step 1: Synthesis of tert-butyl 5-hydroxypentylcarbamate (AU)
[0468] To a stirred solution of 5-aminopentan-1-ol (AT, 3.1 g, 30.05 mmol) in dichloromethane (30 mL) was added di-tert-butyl dicarbonate (6.56 g, 30.06 mmol) at 0 °C. The resulting mixture was then stirred at room temperature for 4h. The solvent was removed under reduced pressure to give a crude residue which was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v= 1: 2)) to give AU (yield: 98%) as a colorless oil. LC-MS (ES+): m/z 204.00 [MH+], tR =1.29 min (2.6 minute run).
[0469] Step 2: Synthesis of ethyl 2-[(5-{[(tert-butoxy)carbonyl]amino}pentyl)oxy]acetate (AV)
[0470] To a stirred solution of tert-butyl N-(5-hydroxypentyl)carbamate (AU, 1.5 g, 7.38 mmol) in dichloromethane (10 mL) was added BF .
3Et2O (0.1 mL) at 0 °C. To this mixture was then added a solution of ethyl 2-diazoacetate (850 mg, 7.45 mmol) in dichloromethane (2 mL) at 0 °C. The resulting mixture was allowed to warm up to room temperature and stirred at room temperature for 2 hours. Saturated aqueous sodium bicarbonate (30 mL) was added to the reaction, the resulting mixture was extracted with ethyl acetate (150 mL x 3). The organic layers were combined, dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v= 1: 7)) to give AV (yield: 15%) as a colorless oil. LC-MS (ES+): m/z 290.05 [MH+], tR =1.55 min (2.6 minute run).
[0471] Step 3: Synthesis of ethyl 2-(5-aminopentyloxy)acetate (L-14)
[0472] To a stirred solution of ethyl ethyl 2-[(5-{[(tert- butoxy)carbonyl]amino}pentyl)oxy]acetate (AV, 400 mg, 1.38 mmol) in dichloromethane (5 mL) was added trifluoroacetic acid (5 mL) at room temperature. The resulting solution was stirred at room temperature for 2 hours. The reaction mixture was then concentrated under vacuum to give L-14 (yield: 84%) as a yellow oil. LC-MS (ES+): m/z 190.00 [MH+], tR =1.01 min (2.6 minute run).
[0473] L-15: meth l 2- 2- 2- meth lamino ethox ethox acetate
Figure imgf000136_0001
Figure imgf000136_0002
.
[0474] Step 1: Synthesis of 2-[2-(benzylamino)ethoxy]ethan-1-ol (AX)
[0475] To a stirred solution of 2-(2-aminoethoxy)ethan-1-ol (AW, 5.0 g) and benzaldehyde (5.0 g) in THF (50 mL) was added sodium triacetoxyborohydride (15.8 g, 74.5 mmol) at 0 °C. The resulting solution was then stirred at room temperature for 4 hours. Water (50 mL) was added to the reaction and the resulting mixture was extracted with ethyl acetate (50 mL x 2). The organic layers were combined, dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent: dichloromethane/methanol (v:v = 3:1) to give AX (yield: 85%) as a white solid. LC-MS (ES+): m/z 195.95[MH+], tR = 0.22 min (2.0 minute run).
[0476] Step 2: Synthesis of 2-{2-[benzyl(methyl)amino]ethoxy}ethan-1-ol (AY)
[0477] To a stirred solution of of 2-[2-(benzylamino)ethoxy]ethan-1-ol (AX, 10.0 g) in methanol (200 mL) was added formaldehyde (38% in water) (4.9 mL) and triacetoxyborohydride (17.0 g) at room temperature. The resulting solution was stirred at room temperature for 2 hours.
Saturated aq. sodium bicarbonate (100 mL) was added to the reaction, and bulk of organic solvent was then removed under reduced pressure. The resulting mixture was extracted with ethyl acetate (200 mL x 3). The organic layers were combined, dried over anhydrous sodium sulfate and then concentrated under reduced pressure followed by high vacuum pump to give AY (yield: 33%) as a yellow oil. LC-MS (ES+): m/z 210.00 [MH+], tR = 0.43 min (2.0 minute run).
[0478] Step 3: Synthesis of methyl 2-(2-{2-[benzyl(methyl)amino]ethoxy}ethoxy)acetate (AZ) [0479] To a stirred solution of 2-{2-[benzyl(methyl)amino]ethoxy}ethan-1-ol (AY, 2 g) in dichloromethane (20 mL) was added a solution of sodium hydroxide (37%) in water (20 mL) followed by tert-butyl 2-bromoacetate (7.76 g) and TBAC (2.78 g) at room temperature. The resulting mixture was stirred at room temperature for 15 hours. The aqueous layer was separated, and to which aq. hydrogen chloride (4N) was added to adjust the pH to ~3 before it was concentrated under reduced pressure to give a crude residue. Methanol (20 mL) was then added to this residue and insoluble salts were filtered out. The solution was concentrated under vacuum to give 2-(2-[2-[benzyl(methyl)amino]ethoxy]ethoxy)acetic acid (yield: 78%) as a yellow oil. To a stirred solution of 2-(2-{2-[benzyl(methyl)amino]ethoxy}ethoxy)acetic acid (2 g, 7.48 mmol, 1.00 equiv) prepare above in methanol (50 mL) was slowly added sulfuric acid (2 mL) at room temperature. The resulting solution was stirred at 70 oC in an oil bath for 3h. The bulk of solvent was removed under reduced pressure to give a residue, which was diluted with H2O (30 mL). Sodium carbonate was then added to the mixture to adjust the pH to ~8. The mixture was then extracted with ethyl acetate (50 mL x 2), the organic layers were combined, dried over anhydrous sodium sulfate and then concentrated under reduced pressure followed by high vacuum pump to give AZ (yield: 29%) as a yellow oil. LC-MS (ES+): m/z 281.95 [MH+], tR = 0.30 min (2.0 minute run).
[0480] Step 4: Synthesis of methyl 2-{2-[2-(methylamino)ethoxy]ethoxy}acetate (L-15)
[0481] To a stirred mixture of methyl 2-(2-{2-[benzyl(methyl)amino]ethoxy}ethoxy)acetate (AZ, 600 mg, 2.13 mmol) and palladium on carbon (300 mg) in methanol (30 mL) under a nitrogen atmosphere was charged with hydrogen gas via a balloon. The resulting mixture was stirred at room temperature for 15 hours. The solid material was removed by filtration and the solution was concentrated under vacuum to give L-15 (400 mg) as yellow oil, which was used for next step without any further purifications. LC-MS (ES+): m/z 191.95 [MH+], tR = 0.31 min (2.0 minute run).
[0482] L-16: ethyl 2-(5-(methylamino)pentyloxy)acetate
Figure imgf000138_0001
.
[0483] Step 1: Synthesis of ethyl 2-[(5-{[(tert- butoxy)carbonyl](methyl)amino}pentyl)oxy]acetate (BB)
[0484] To a stirred solution of ethyl 2-[(5-{[(tert-butoxy)carbonyl]amino}pentyl)oxy]acetate (BA, 1.1 g, 3.8 mmol) in N,N-dimethylformamide (10 mL) was added CH3I (0.71 mL, 11.4 mmol) at 0 °C, followed by sodium hydride (304 mg, 7.60 mmol, 60% in mineral oil) in several portions at 0 °C. The resulting mixture was stirred at room temperature for 16 hours. Water (1.0 mL) was added and the resulting mixture was extracted with ethyl acetate (50 mL x 2). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (100 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue which was purified by a flash silica gel chromatography (eluent: ethyl
acetate/petroleum ether (v: v = 1: 10)) to give BB (yield: 21%) as a yellow oil. LC-MS (ES+): m/z 326.20 [MNa+], tR = 1.55 min (2.6 minute run).
[0485] Step 2: Synthesis of ethyl 2-{[5-(methylamino)pentyl]oxy}acetate (L-16)
[0486] To a stirred solution of ethyl 2-[(5-{[(tert- butoxy)carbonyl](methyl)amino}pentyl)oxy]acetate (BB, 240 mg, 0.79 mmol) in
dichloromethane (5 mL) was added trifluoroacetic acid (0.5 mL). The resulting solution was stirred at room temperature for 16 hours. The solvents were removed under recued pressure followed by high vacuum pump to give L-16 (yield: 99%) as a yellow oil. LC-MS (ES+): m/z 204.20 [MH+], tR = 0.56 min (2.0 minute run).
[0487] L-17: 2-(3-(2-(tosyloxy)ethoxy)propoxy)acetic acid
Figure imgf000139_0001
.
[0488] Step 1: Synthesis of tert-butyl 2-{3-[2-(benzyloxy)ethoxy]propoxy}acetate (BD)
[0489] To a stirred solution of 3-[2-(benzyloxy)ethoxy]propan-1-ol (BC, 1.8 g, 8.56 mmol) and tert-butyl 2-bromoacetate (6.6 g, 33.84 mmol, 4.00 equiv) in dichloromethane (40 mL) was added TBAC (2.4 g) and aq. Solution of sodium hydroxide (37%, 40 mL). The resulting mixture was stirred at room temperature overnight. LC-MS indicated formation of the desired product. The reaction mixture was then extracted with ethyl acetate (150 x 3 mL), the organic layers combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a crude residue, which was purified by a flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v : v = 1 : 2) to give BD (yield: 90%) as a colorless oil.1H NMR (300 MHz, CDCl3): ^ 7.35-7.27 (m, 5H), 4.57 (s, 2H), 3.94 (s, 2H), 3.63-3.57 (m, 8H), 1.96-1.87 (m, 2H), 1.47 (s, 9H); LC-MS (ES+): m/z 347.10 [MNa+], tR = 1.72 min (2.6 minute run).
[0490] Step 2: Synthesis of tert-butyl 2-[3-(2-hydroxyethoxy)propoxy]acetate (BE)
[0491] To a stirred mixture of tert-butyl 2-{3-[2-(benzyloxy)ethoxy]propoxy}acetate (BD, 2.5 g, 7.71 mmol) and palladium on carbon (2.0 g) in methanol (20 mL) under a nitrogen atmosphere was introduced hydrogen gas via a balloon. The resulting mixture was stirred overnight at room temperature under hydrogen gas atmosphere. LC-MS indicated completion of the reaction. The solids were removed by filtration, the solution was concentrated under vacuum to give BE (yield: 99%) as a colorless oil. LC-MS (ES+): m/z 257.10 [MNa+], tR = 1.21 min (2.6 minute run).
[0492] Step 3: Synthesis of tert-butyl 2-(3-{2-[(4- methylbenzenesulfonyl)oxy]ethoxy}propoxy)acetate (BF)
[0493] To a stirred solution of tert-butyl 2-[3-(2-hydroxyethoxy)propoxy]acetate (BE, 1.8 g, 7.68 mmol) in dichloromethane (50 mL) was added 4-toluenesulfonyl chloride (2.2 g, 11.54 mmol), triethylamine (2.33 g, 23.03 mmol) and 4-dimethylaminopyridine (95 mg, 0.78 mmol). The resulting mixture was stirred overnight at room temperature. LC-MS indicated formation of the desired product. The reaction mixture was concentrated under reduced pressure to give a crude residue, which was purified by a flash silica gel chromatography (eluent: ethyl
acetate/petroleum ether (v : v = 1 : 2) to give BF (yield: 80%) as a yellow oil.1H NMR (400 MHz, CDCl3): ^ 7.80 (d, J = 8.0 Hz, 2H), 7.34 (d, J = 8.4 Hz, 2H), 4.15 (t, J = 3.6 Hz, 2H), 3.93 (s, 2H), 3.61 (t, J = 3.6 Hz, 2H), 3.55-3.49 (m, 4H), 2.45 (s, 3H), 1.85-1.78 (m, 2H), 1.48 (s, 9H); LC-MS (ES+): m/z 411.00 [MNa+], tR = 1.12 min (2.0 minute run).
[0494] Step 4: Synthesis of 2-(3-{2-[(4-methylbenzenesulfonyl)oxy]ethoxy}propoxy)acetic acid (L-17)
[0495] To a stirred solution of tert-butyl 2-(3-{2-[(4- methylbenzenesulfonyl)oxy]ethoxy}propoxy)acetate (BF, 400 mg, 1.03 mmol) in
dichloromethane (3 mL) was added trifluoroacetic acid (1 mL) at room temperature. The resulting solution was stirred at room temperature for 1 hour. LC-MS indicated completion of the reaction. The reaction mixture was concentrated under reduced pressure to give L-17 (350 mg) as a yellow oil, which was used for next step without further purifications. LC-MS (ES+): m/z 332.90 [MH+], tR = 0.81 min (2.0 minute run).
[0496] Unless otherwise noted, the following intermediates and their analogs (for examples, but not limited to, analogs with substitutions such as halogens) were synthesized according to similar procedures described above for the synthesis of L-17, by utilizing corresponding starting materials and reagents.
[0497] L-18: 2-(2-hydroxyethoxy)ethyl 4-methylbenzenesulfonate
Figure imgf000140_0001
.
[0498] L-19: ethyl 2-(2-(2-(tosyloxy)ethoxy)ethoxy)acetate
Figure imgf000140_0002
.
[0499] L-20: ethyl 3-(2-(2-(tosyloxy)ethoxy)ethoxy)propanoate
Figure imgf000140_0003
.
[0500] L-21: ethyl 5-(tosyloxy)pentanoate .
[0501] L-22: ethyl 3-(2-(tosyloxy)ethoxy)propanoate
Figure imgf000141_0001
.
[0502] L-23: ethyl 2-(5-(tosyloxy)pentyloxy)acetate
.
[0503] L-24: ethyl 3-(5-(tosyloxy)pentyloxy)propanoate .
[0504] L-25: 5-hydroxypentyl 4-methylbenzenesulfonate
.
[0505] L-26: ethyl 2-(5-(tosyloxy)pentyloxy)acetate
.
[0506] L-27: ethyl 2-(3-(tosyloxy)propoxy)acetate
.
[0507] L-28: ethyl 2-(2-(tosyloxy)ethoxy)acetate .
[0508] L-29: ethyl 2-(4-(2-(tosyloxy)ethoxy)butoxy)acetate .
[0509] L-30: 2-(2-(2-hydroxyethoxy)ethoxy)ethyl 4-methylbenzenesulfonate .
[0510] L-31: 2-((2R,3R)-3-(2-hydroxyethoxy)butan-2-yloxy)ethyl 4-methylbenzenesulfonate
Figure imgf000142_0001
.
[0511] L-32: 2-(2-piperazin-1-yl)-ethoxy-acetic acid
Figure imgf000142_0002
[0512] L-33: methyl 6-(4-(2-(2-(tert-butoxy)-2-oxoethoxy)ethyl)piperazin-1-yl)nicotinate
Figure imgf000142_0003
[0513] Step 1: Synthesis of tert-butyl 4-(5-(methoxycarbonyl)pyridin-2-yl)piperazine-1- carboxylate:
Figure imgf000142_0004
[0514] A mixture of methyl 6-fluoronicotinate (2.0 g, 13.2 mmol), tert-butyl piperazine-1- carboxylate (2.4 g, 13.2 mmol) and N-ethyl-N-isopropylpropan-2-amine (3.3 g, 26.4 mmol) in anhydrous 1-methylpyrrolidin-2-one (10 ml) was stirred at 90°C for 12 hours. TLC showed the reaction was complete. The cooled reaction mixture was partitioned between water (10 ml) and ethyl acetate (50 ml). The organic layer was collected, washed with brine (50 ml x 2), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by column (eluted with 20% ethyl acetate in hexane) to afford tert-butyl 4-(5- (methoxycarbonyl)pyridin-2-yl)piperazine-1-carboxylate (4.0 g, yield 95%) as yellow solid. 1HNMR (400 MHz, CDCl3): ^ 1.48 (s, 9H), 3.53-3.56 (m, 4H), 3.67-3.69 (m, 4H), 3.87 (s, 3H), 6.58 (d, J = 8.8 Hz, 2H), 8.02-8.05 (m, 1H), 8.79-8.80 (m, 1H). Chemical Formula: C16H23N3O4, Molecular Weight: 321.37.
[0515] Step 2: Synthesis of methyl 6-(piperazin-1-yl)nicotinate
Figure imgf000143_0001
[0516] A mixture of tert-butyl 4-(5-(methoxycarbonyl)pyridin-2-yl)piperazine-1-carboxylate (4.0 g, 12.4 mol) and 2,2,2-trifluoroacetic acid (10 ml) in dichloromethane (10 ml) was stirred at room temperature for 2 hours. TLC showed the reaction was complete. The volatiles were evaporated under reduced pressure. The residue was taken up with dichloromethane (50 ml) and washed with aqueous sodium bicarbonate solution (1N, 15 ml), dried over sodium sulfate to give methyl 6-(piperazin-1-yl)nicotinate (3.8 g, crude) as yellow oil which was used in next step without further purification. 1HNMR (400 MHz, DMSO-d): ^ 3.13-3.16 (m, 4H), 3.80 (s, 3H), 3.82-3.85 (m, 4H), 6.96 (d, J = 9.2 Hz, 1H), 8.00-8.03 (m, 1H), 8.67-8.68 (m, 1H). Chemical Formula: C11H15N3O2, Molecular Weight: 221.26.
[0517] Step 3: Synthesis of methyl 6-(4-(2-(2-(tert-butoxy)-2-oxoethoxy)ethyl)piperazin-1- yl)nicotinate.
Figure imgf000143_0002
A mixture of methyl 6-(piperazin-1-yl)nicotinate (500 mg, 2.3 mmol), tert-butyl 2-(2- (tosyloxy)ethoxy)acetate (745 mg, 2.3 mmol) and potassium carbonate (1.2 g, 9.0 mmol) in anhydrous N,N-dimethylformamide (10 ml) was stirred at 40°C for 12 hours. TLC showed the reaction was complete. The cooled reaction mixture was partitioned between water (20 ml) and ethyl acetate (20 ml). The organic layer was collected, washed with brine (100 ml x 2), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluted with 20% ethyl acetate in hexane) to afford methyl 6-(4-(2-(2-(tert-butoxy)-2-oxoethoxy)ethyl)piperazin-1-yl)nicotinate (L-33) (400 mg, yield 46%) as yellow solid.
[0518] Synthesis of Examples
[0519] Example 1: (2S,4R)-1-((S)-2-(2-(3-(5-(4-(3-(4-cyano-3-(trifluoromethyl)phenyl)-5,5- dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)phenoxy)pentyloxy)propoxy)acetamido)-3,3- dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2- carboxamide:
Figure imgf000144_0001
.
[0520] Step 1: Synthesis of tert-butyl 2-(3-{[5-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenoxy)pentyl]oxy}propoxy)acetate (BG)
[0521] To a stirred solution of tert-butyl 2-[3-[(5-[[(4- methylbenzene)sulfonyl]oxy]pentyl)oxy]propoxy]acetate (AB, 150 mg, 0.35 mmol) in acetonitrile (10 mL) was added 4-[3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- sulfanylideneimidazolidin-1-yl]-2-(trifluoromethyl)benzonitrile (ABM-3, 141 mg, 0.35 mmol) and potassium carbonate (144 mg, 1.04 mmol). The resulting mixture was stirred overnight at 80 °C in an oil bath. LC-MS indicated formation of the desired product. The reaction mixture was then extracted with ethyl acetate (20 mL x 2). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (20 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether, v: v = 1:1) to give 0.22 g of BG as a yellow oil.1H NMR (400 MHz, CDCl3): ^ 7.96 (s, 2H), 7.86 (d, J = 8.6 Hz, 1H), 7.19 (d, J = 8.8 Hz, 2H), 7.02 (d, J = 8.6 Hz, 2H), 4.50 (s, 2H), 4.30 (t, J = 6.4 Hz, 2H), 4.02 (t, J = 6.4 Hz, 2H), 3.53 (m, 2H), 3.44 (m, 2H), 1.96-1.80 (m, 4H), 1.69-1.53 (m, 2H), 1.49 (s, 6H), 1.48 (s, 9H), 1.44-1.22 (m, 2H); Mass (ES+): m/z 686.35 [MNa+].
[0522] Step 2: Synthesis of 2-(3-[[5-(4-[3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5-dimethyl-4- oxo-2-sulfanylideneimidazolidin-1-yl]phenoxy)pentyl]oxy]propoxy)acetic acid (BH)
[0523] To a stirred solution of tert-butyl 2-(3-{[5-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenoxy)pentyl]oxy}propoxy)acetate (BG, 220 mg, 0.33 mmol) in dioxane (4.0 mL) was added hydrogen chloride (2N in water, 1.0 mL). The resulting mixture was stirred at 80 °C for 2h. LC-MS indicated formation of the desired product. The resulting mixture was concentrated under reduced pressure to provide 200 mg of BH as light yellow oil. Mass (ES+): m/z 608.25 [MH+].
[0524] Step 3: Synthesis of Example 1:
[0525] To a stirred solution of 2-(3-[[5-(4-[3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5-dimethyl- 4-oxo-2-sulfanylideneimidazolidin-1-yl]phenoxy)pentyl]oxy]propoxy)acetic acid (BH, 160 mg, 0.26 mmol) in N,N-dimethylformamide (5 mL) was added
Figure imgf000145_0001
dimethylbutanoyl]-4-hydroxy-N-{[4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl}pyrrolidine-2- carboxamide hydrochloride (ULM-1, 182 mg, 0.39 mmol), DIPEA (151 mg, 1.17 mmol), EDCI (101 mg, 0.53 mmol) and HOBt (70 mg, 0.52 mmol). The resulting mixture was stirred at room temperature for 5 h and LC-MS indicated formation of the desired product. Water (20 mL) was added to the reaction, the resulting mixture was extracted with ethyl acetate (20 mL x 2). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (20 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue. The residue was purified by Prep-HPLC to give 60 mg of Example 1 as a white solid.1H NMR (400 MHz, CD3OD): ^ 8.88 (s, 1H), 8.16 (d, J = 8.0 Hz, 2H), 8.00 (s, 1H), 7.49- 7.42 (m, 4H), 7.28 (d, J = 8.8 Hz, 2H), 7.06 (m, 2H), 4.87 (s, 1H), 4.59 (m, 3H), 4.37 (m, 1H), 4.05 (m, 4H), 3.88 (m, 2H), 3.65 (m, 2H), 3.58 (m, 2H), 3.50 (m, 2H), 2.48 (s, 3H), 2.25 (m, 1H), 2.10 (m, 1H), 1.90 (m, 2H), 1.80 (m, 2H), 1.66 (m, 2H), 1.56 (s, 8H), 1.04 (s, 9H); LC-MS (ES+): m/z 1020.20 [MH+], tR = 2.28 min (3.6 minute run).
[0526] Example 2: (2S,4R)-1-((S)-2-(2-(3-(5-(4-(3-(6-cyano-5-(trifluoromethyl)pyridin-3-yl)- 5,5-dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)phenoxy)pentyloxy)propoxy)acetamido)-3,3- dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2- carboxamide:
Figure imgf000146_0001
.
[0527] Step 1: Synthesis of 2-[3-({5-[(4-methylbenzenesulfonyl)oxy]pentyl}oxy)propoxy]acetic acid (L-1)
[0528] To a stirred solution of tert-butyl 2-[3-({5-[(4- methylbenzenesulfonyl)oxy]pentyl}oxy)propoxy]acetate (AB, 1.3 g, 3.02 mmol) in
dichloromethane (10 mL) was added trifluoroacetic acid (10 mL) at room temperature. The resulting solution was stirred at room temperature for 3 hours. The reaction mixture was then concentrated under vacuum to give 1.5 g (crude) of L-1, which was used for next step without any further purification. LC-MS (ES+): m/z 375.34 [MH+], tR = 1.39 min (2.6 minute run).
[0529] Step 2: Synthesis of (2S,4R)-1-[(2S)-3,3-dimethyl-2-{2-[3-({5-[(4- methylbenzenesulfonyl)oxy]pentyl}oxy)propoxy]acetamido}butanoyl]-4-hydroxy-N-{[4-(4- methyl-1,3-thiazol-5-yl)phenyl]methyl}pyrrolidine-2-carboxamide (BI)
[0530] To a stirred solution 2-[3-({5-[(4- methylbenzenesulfonyl)oxy]pentyl}oxy)propoxy]acetic acid (L-1, 1.5 g, 4.01 mmol) in N,N- dimethylformamide (20 mL) was added HATU (1.36 g, 3.58 mmol), DIEA (0.7 mL) and (2S,4R)- 1-[(2S)-2-amino-3,3-dimethylbutanoyl]-4-hydroxy-N-{[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}pyrrolidine-2-carboxamide (ULM-1, 1.3 g, 3.02 mmol) at room temperature. The resulting mixture was stirred for 2h at room temperature. It was then diluted with water (100 mL) and extracted with ethyl acetate (100 mL x 3). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (60 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent: dichloromethane/methanol (v: v = 10:1)) to give 0.5 g of BI. LC-MS (ES+): m/z 787.34 [MH+], tR = 1.87 min (3.0 minute run).
[0531] Step 3: Synthesis of (2S,4R)-1-[(2S)-2-[2-(3-{[5-(4-{3-[6-cyano-5- (trifluoromethyl)pyridin-3-yl]-5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1- yl}phenoxy)pentyl]oxy}propoxy)acetamido]-3,3-dimethylbutanoyl]-4-hydroxy-N-{[4-(4-methyl- 1,3-thiazol-5-yl)phenyl]methyl}pyrrolidine-2-carboxamide (Example 2)
[0532] To a stirred solution of 5-[3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- sulfanylideneimidazolidin-1-yl]-3-(trifluoromethyl)pyridine-2-carbonitrile (ABM-4, 52 mg, 0.13 mmol), (2S,4R)-1-[(2S)-3,3-dimethyl-2-{2-[3-({5-[(4- methylbenzenesulfonyl)oxy]pentyl}oxy)propoxy]acetamido}butanoyl]-4-hydroxy-N-{[4-(4- methyl-1,3-thiazol-5-yl)phenyl]methyl}pyrrolidine-2^carboxamide (BI, 100 mg, 0.13 mmol) in N,N-dimethylformamide (10 mL) was added potassium carbonate (34 mg, 0.25 mmol) under an atmosphere of nitrogen. The resulting solution was stirred for 2 h at 80oC. The resulting mixture was concentrated under vacuum to give a crude residue, which was purified by Prep-HPLC to give 38.1 mg of Example 2 as a white solid.1H NMR (300 MHz, CD3OD): ^ 9.12 (s, 1H), 8.83(s, 1H), 8.63 (s, 1H), 7.44-7.39 (m, 4H), 7.00 (d, J = 9.0 Hz, 2H), 7.20 (d, J = 9.0 Hz, 2H), 4.80-4.26 (m, 5H), 4.06-3.65 (m, 6H), 3.62-3.35 (m, 6H), 2.43 (s, 3H) , 2.21-2.01 (m, 2H), 1.85- 1.65 (m, 4H), 1.60-1.42 (m, 10H), 1.00 (s, 9H): LC-MS (ES+): m/z 1021.12 [MH+], tR = 2.36 min (3.6 minute run).
[0533] Unless otherwise noted, the following examples were synthesized according to analogous procedures described above for synthesis of examples 1 and 2, utilizing corresponding reagents, intermediates, and starting materials. [0534] When referring to the specific exemplary compounds presented herein, the specification uses the terms“example #.” For example, compound 1 (Table 2) is also referred to as Example 1.
[0535] Table 2. Exemplary Compounds.
Figure imgf000148_0001
Figure imgf000149_0001
. ,R =. mn . mnue run.
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0001
Figure imgf000165_0001
. , R . . .
[0536] Example 54: (2S,4R)-1-((S)-2-(2-(6-(4-(3-(4-cyano-3-(trifluoromethyl)phenyl)-5,5- dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)phenoxy)hexa-2,4-diynyloxy)acetamido)-3,3- dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2- carboxamide:
Figure imgf000166_0001
.
[0537] Step 1: Synthesis of tert-butyl 2-{[6-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenoxy)hexa-2,4-diyn-1-yl]oxy}acetate (BJ)
[0538] This material was synthesized according to a similar procedure described in reaction step 1 for the synthesis of Example 1. LC-MS (ES+): m/z 634.05 [MNa+], tR = 1.26 min (2.0 minute run).
[0539] Step 2: Synthesis of 2-{[6-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5-dimethyl-4- oxo-2-sulfanylideneimidazolidin-1-yl}phenoxy)hexa-2,4-diyn-1-yl]oxy}acetic acid (BK)
[0540] This material was synthesized according to a similar procedure described in reaction step 2 for the synthesis of example 1. LC-MS (ES+): m/z 556.10 [MH+], tR = 1.54 min (2.6 minute run).
[0541] Step 3: Synthesis of (2S,4R)-1-[(2S)-2-(2-{[6-(4-{3-[4-cyano-3- (trifluoromethyl)phenyl]-5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenoxy)hexa- 2,4-diyn-1-yl]oxy}acetamido)-3,3-dimethylbutanoyl]-4-hydroxy-N-{[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}pyrrolidine-2^carboxamide (Example 54) [0542] This material was synthesized according to a similar procedure described in reaction step 3 for the synthesis of Example 1.1H NMR (400 MHz, CD3OD): ^ 8.88 (s, 1H), 8.15 (d, J = 8.4 Hz, 2H), 8.00 (d, J = 1.6 Hz, 1H), 7.49-7.43 (m, 4H), 7.34 (d, J = 8.8 Hz, 2H), 7.14 (d, J = 8.8 Hz, 2H), 4.93 (s, 2H), 4.71 (s, 1H), 4.60-4.34 (m, 6H), 4.08 (s, 2H), 3.90-3.80 (m, 2H), 2.49 (s, 3H), 2.25-2.22 (m, 1H), 2.13-2.05 (m, 1H), 1.56 (s, 6H), 1.03 (s, 9H); LC-MS (ES+): m/z 968.45
[MH+], tR = 1.67 min (3.0 minute run).
[0543] Table 3. Exemplary Compounds.
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000169_0001
[0544] Example 62: (2S,4R)-1-((S)-2-tert-butyl-16-(4-(3-(4-cyano-3- (trifluoromethyl)phenyl)-5,5-dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)phenyl)-4,13-dioxo- 6,9-dioxa-3,12-diazahexadecane)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine- 2-carboxamide:
Figure imgf000170_0001
Figure imgf000170_0002
Figure imgf000170_0003
.
[0545] Step 1: Synthesis of ethyl 2-(2-{2-[4-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)butanamido]ethoxy}ethoxy)acetate (BL)
[0546] To a stirred solution of 4-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5-dimethyl-4-oxo- 2-sulfanylideneimidazolidin-1-yl}phenyl)butanoic acid (ABM-12, 417 mg, 0.88 mmol) in N,N- dimethylformamide (10 mL) was added HATU (669 mg, 1.76 mmol), DIEA (454 mg, 3.51 mmol) and ethyl 2-[2-(2-aminoethoxy)ethoxy]acetate hydrochloride (L-13, 400 mg, 1.76 mmol) at 0 °C . The resulting solution was stirred at 0 °C for 30 minutes, and then it was warmed up to room temperature and stirred at room temperature for 15 hours. A mixture of water/ice (1: 1, 50 mL) was added to the reaction, the resulting mixture was extracted with ethyl acetate (100 mL x 3). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (20 mL x 2), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v = 1:1) to give BL (yield: 35%) as a yellow solid. LC-MS (ES+): m/z 649.15[MH+], tR = 1.05 min (2.0 minute run). [0547] Step 2: Synthesis of 2-(2-{2-[4-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5-dimethyl- 4-oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)butanamido]ethoxy}ethoxy)acetic acid (BM)
[0548] To a stirred solution of ethyl 2-(2-{2-[4-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)butanamido]ethoxy}ethoxy)acetate (BL, 200 mg, 0.31 mmol) in methanol (10 mL) was added a solution of NaOH (123 mg, 3.08 mmol) in water (10 mL) at room temperature. The resulting solution was then heated to 50 °C and stirred at this temperature for 2 hours. The bulk of organic solvent was removed under reduced pressure. To the remaining residue was added aqueous hydrogen chloride (1 M) to adjust the pH to ~3. The resulting mixture was extracted with ethyl acetate (50 mL x 2), the organic layers were combined, washed with saturated aqueous solution of sodium chloride (20 mL x 2), dried over anhydrous sodium sulfate and then concentrated under reduced pressure followed by high vacuum pump to give BM (yield: 78%) as a yellow solid. LC-MS (ES+): m/z 621.20 [MH+], tR = 0.96 min (2.0 minute run).
[0549] Step 3: Synthesis of (2S,4R)-1-[(2S)-2-[2-(2-{2-[4-(4-{3-[4-cyano-3- (trifluoromethyl)phenyl]-5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1- yl}phenyl)butanamido]ethoxy}ethoxy)acetamido]-3,3-dimethylbutanoyl]-4-hydroxy-N-{[4-(4- methyl-1,3-thiazol-5-yl)phenyl]methyl}pyrrolidine-2-carboxamide (Example 62)
[0550] To a stirred solution of 2-(2-{2-[4-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)butanamido]ethoxy}ethoxy)acetic acid (BM, 200 mg, 0.32 mmol) in N,N-dimethylformamide (20 mL) was added HATU (245 mg, 0.64 mmol), DIEA (166 mg, 1.28 mmol) and (2S,4R)-1-[(2S)-2-amino-3,3-dimethylbutanoyl]-4- hydroxy-N-{[4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl}pyrrolidine-2^carboxamide
hydrochloride (ULM-1, 226 mg, 0.48 mmol) at 0 °C. The resulting solution was stirred at 0 °C for 30 min, and then it was warmed up to room temperature and stirred at room temperature for 15 hours. A mixture of water/ice (1: 1, 50 mL) was added to the reaction, the resulting mixture was extracted with ethyl acetate (100 mL x 3). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (50 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by Prep- HPLC to give Example 62 (yield: 6%) as a yellow solid.1H NMR (400MHz, CD3OD): ^ 8.89 (s, 1H), 8.18-8.16 (d, J = 8.4 Hz, 2H), 8.01-7.99 (d, J = 8.0 Hz, 1H), 7.47-7.41(m, 4H), 7.38-7.36 (d, J = 8.4 Hz, 2H), 7.30-7.28 (d, J = 8.4 Hz, 2H), 4.87 (s, 1H), 4.78-4.60 (m, 3H), 4.39-4.35 (d, J = 15.2 Hz, 1H), 4.04-3.98 (m, 2H), 3.98-3.85 (m, 2H), 3.72-3.60 (m, 7H), 3.50-3.49(m, 1H), 2.71-2.69 (m, 2H), 2.49 (s, 3H), 2.45-2.28 (m, 3H), 2.25-2.10 (m, 1H), 2.10-1.95 (m, 2H), 1.58 (s, 6H), 1.09 (s, 9H); LC-MS (ES+): m/z 1033.50 [MH+], tR = 3.06 min (5.6 minute run).
[0551] Examples 63-65 were synthesized according to similar procedure described for synthesis of example 62, by using corresponding starting materials and intermediates.
[0552] Table 4. Exemplary Compounds.
Figure imgf000172_0001
Figure imgf000173_0001
[0553] Example 66: 2-(2-(4'-(3-(4-cyano-3-(trifluoromethyl)phenyl)-5,5-dimethyl-4-oxo-2- thioxoimidazolidin-1-yl)biphenyl-4-yloxy)ethoxy)ethyl (S)-1-((2S,4R)-4-hydroxy-2-(4-(4- methylthiazol-5-yl)benzylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2- ylcarbamate:
Figure imgf000173_0002
Figure imgf000173_0003
.
[0554] Step 1: Synthesis of 4-[3-(4-{4-[2-(2-hydroxyethoxy)ethoxy]phenyl}phenyl)-4,4- dimethyl-5-oxo-2--ulfanylideneimidazolidin-1-yl]-2-(trifluoromethyl)benzonitrile (BN)
[0555] To a stirred solution of 4-{3-[4-(4-hydroxyphenyl)phenyl]-4,4-dimethyl-5-oxo-2- sulfanylideneimidazolidin-1-yl}-2-(trifluoromethyl)benzonitrile (ABM-14, 610.5 mg, 1.27 mmol) in N,N-dimethylformamide (10 mL) was added K2CO3 (318.46 mg, 2.29 mmol) and 2-{2-[(4- methylbenzenesulfonyl)oxy]ethoxy}ethan-1-ol (L-18, 300 mg, 1.15 mmol) at room temperature. The resulting mixture was then stirred at 80 °C for 2 hours in an oil bath, LC-MS indicated formation of the desired product. The reaction mixture was cooled down to room temperature, water (20mL) was added and the resulting mixture was extracted with ethyl acetate (100 mL x 2). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (20 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by a flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v = 7:3) to give BN (yield: 66%) as a light yellow oil. LC-MS (ES+): m/z 570, [MH+], tR = 1.60 min (2.0 minute run).
[0556] Step 2: synthesis of 2-{2-[4-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5-dimethyl-4- oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)phenoxy]ethoxy}ethyl N-[(2S)-1-[(2S,4R)-4- hydroxy-2-({[4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3- dimethyl-1-oxobutan-2-yl]carbamate (Example 66)
[0557] To a stirred solution of 4-[3-(4-{4-[2-(2-hydroxyethoxy)ethoxy]phenyl}phenyl)-4,4- dimethyl-5-oxo-2--ulfanylideneimidazolidin-1-yl]-2-(trifluoromethyl)benzonitrile (200 mg, 0.35 mmol) in dichloromethane (10 mL) was added triethylamine (106.5 mg, 1.05 mmol), followed by triphosgene (36.5 mg, 0.12 mmol) which was added slowly in 30 minutes at 0 °C. To this mixture was then added (2S,4R)-1-[(2S)-2-amino-3,3-dimethylbutanoyl]-4-hydroxy-N-{[4-(4-methyl-1,3- thiazol-5-yl)phenyl]methyl}pyrrolidine-2-carboxamide hydrochloride (ULM-1, 196.9 mg, 0.42 mmol) at 0 °C. The resulting mixture was then warmed up to room temperature and stirred at room temperature for 2 hours. Water (20mL) was added to the reaction and the resulting mixture was extracted with dichloromethane (50 mL x 3). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (20 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by Prep-HPLC to give Example 66 (yield: 6%) as a white solid.1H-NMR (400MHz, CD3OD): ^ 8.88 (s, 1 H), 8.20-8.17 (m, 2 H), 8.04-8.02 (d, J = 8.0 Hz, 1 H), 7.77-7.72 (m, 2 H), 7.65-7.59 (m, 2 H), 7.48-7.42 (m, 6 H), 7.08-7.06 (d, J = 8.4 Hz, 2 H), 4.61-4.53 (m, 1 H), 4.47-4.44 (s, 1 H), 4.38-4.34 (m, 2 H), 4.25-4.20 (m, 4 H), 3.92-3.90 (m, 3 H), 3.82-3.79 (m, 3 H), 2.48 (s, 3 H), 2.26-2.21 (m, 1 H), 2.13-1.09 (m, 1 H), 1.61 (s, 6 H), 1.30 (s, 1 H), 1.04 (s, 9 H); LC-MS (ES+): m/z 1026.40 [MH+], tR = 2.23 min (3.0 minute run). [0558] Example 67: (2S,4R)-1-((S)-2-(2-(2-(2-(4'-(3-(4-cyano-3-(trifluoromethyl)phenyl)- 5,5-dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)biphenyl-4-yloxy)ethoxy)ethoxy)acetamido)- 3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2- carboxamide:
Figure imgf000175_0001
Figure imgf000175_0002
Figure imgf000175_0003
.
[0559] Step 1: Synthesis of ethyl 2-(2-{2-[4-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)phenoxy]ethoxy}ethoxy)acetate (BO)
[0560] To a stirred solution of 4-{3-[4-(4-hydroxyphenyl)phenyl]-4,4-dimethyl-5-oxo-2- sulfanylideneimidazolidin-1-yl}-2-(trifluoromethyl)benzonitrile (ABM-14, 300 mg, 0.62 mmol) in N,N-dimethylformamide (10 mL) was added K2CO3 (172 mg, 1.24 mmol) and ethyl 2-(2-{2- [(4-methylbenzenesulfonyl)oxy]ethoxy}ethoxy)acetate (L-19, 237.4 mg, 0.69 mmol). The resulting mixture was stirred at 80 °C in an oil bath for 2 hours. The reaction was cooled down to room temperature, water (50mL) was added and the resulting mixture was extracted with ethyl acetate (100 mL x 2). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (30 mL x 3), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by a flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v = 3:7)) to give BO (yield: 48%) as light yellow oil. LC- MS (ES+): m/z 656, [MH+], tR = 1.19 min (2.0 minute run).
[0561] Step 2: Synthesis of 2-(2-{2-[4-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5-dimethyl- 4-oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)phenoxy]ethoxy}ethoxy)acetic acid (BP)
[0562] To a stirred solution of ethyl 2-(2-{2-[4-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)phenoxy]ethoxy}ethoxy)acetate (BO, 198 mg, 0.30 mmol) in ethanol (5 mL) was added a solution of sodium hydroxide (36.3 mg, 0.91 mmol) in water (2 mL) at room temperature. The resulting solution was stirred overnight at room temperature, the bulk of organic solvent was then removed under reduced pressure. To the remaining aqueous residue was added hydrogen chloride in water (1N) to adjust the pH to ~5.0, and the resulting mixture was extracted with ethyl acetate (250 mL x 2). The organic layers were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure followed by high vacuum pump to give BP (yield: 99%) as a light yellow oil. LC-MS (ES+): m/z 628, [MH+], tR = 1.08 min (2.0 minute run).
[0563] Step 3: Synthesis of (2S,4R)-1-[(2S)-2-[2-(2-{2-[4-(4-{3-[4-cyano-3- (trifluoromethyl)phenyl]-5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1- yl}phenyl)phenoxy]ethoxy}ethoxy)acetamido]-3,3-dimethylbutanoyl]-4-hydroxy-N-{[4-(4- methyl-1,3-thiazol-5-yl)phenyl]methyl}pyrrolidine-2-carboxamide (Example 67)
[0564] To a stirred solution of 2-(2-{2-[4-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)phenoxy]ethoxy}ethoxy)acetic acid (BP, 190 mg, 0.30 mmol) in N,N-dimethylformamide (10 mL) was added HATU (149.7 mg, 0.39 mmol), DIEA (156.4 mg, 1.21 mmol) and (2S,4R)-1-[(2S)-2-amino-3,3-dimethylbutanoyl]-4- hydroxy-N-{[4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl}pyrrolidine-2^carboxamide
hydrochloride (ULM-1, 183.9 mg, 0.39 mmol). The resulting solution was stirred at room temperature for 2 hours. Water (50mL) was added and the resulting mixture was extracted with ethyl acetate (100 mL x 2). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (25 mL x 3), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified Prep-HPLC to give Example 67 (yield: 17%) as a white solid.1H-NMR (400MHz, CD3OD): ^ 8.82 (s, 1 H), 8.19-8.16 (d, J = 9.0 Hz, 2 H), 8.02-8.00 (d, J = 8.1 Hz, 1 H), 7.72-7.69 (d, J = 8.1 Hz, 2 H), 7.61- 7.55 (m, 2 H), 7.46-7.37 (m, 6 H),7.08-7.01 (m, 2 H), 4.71(s, 1 H), 4.61-4.51 (m, 1 H), 4.47 (s, 2 H), 4.38-4.31 (m, 1 H), 4.23-4.20 (m, 2 H), 4.01(s, 2 H), 3.96-3.78 (m, 4 H), 3.63 (s, 4 H), 2.43 (s, 3H), 2.27-2.20 (m,1 H), 2.13-2.04 (m, 1 H), 1.61 (s, 6 H), 1.04 (s, 9 H); LC-MS (ES+): m/z 1040.10 [MH+], tR = 2.26 min (3.0 minute run).
[0565] Examples 74 and 76 were synthesized according to similar procedure described for synthesis of Example 66, by using corresponding starting materials and intermediates. Examples 68-73, 75, 77-79 were synthesized according to similar procedure described for synthesis of Example 67, by using corresponding starting materials and intermediates.
[0566] Table 5. Exemplary Compounds.
Figure imgf000177_0001
Figure imgf000178_0001
, . , , . , . , .
(3.0 minute run).
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
[0567] Example 80: (2S,4R)-1-((S)-2-(2-(3-(2-(4-(4-(3-(4-cyano-3-(trifluoromethyl)phenyl)- 5,5-dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)phenyl)piperidin-1- yl)ethoxy)propoxy)acetamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5- yl)benzyl)pyrrolidine-2-carboxamide:
Figure imgf000181_0002
. [0568] Step 1: synthesis of (2S,4R)-1-[(2S)-3,3-dimethyl-2-[2-(3-{2-[(4-methylbenzenesulfonyl) oxy]ethoxy}propoxy)acetamido]butanoyl]-4-hydroxy-N-{[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}pyrrolidine-2-carboxamide (BQ)
[0569] To a stirred solution of 2-(3-{2-[(4-methylbenzenesulfonyl)oxy]ethoxy}propoxy)acetic acid (L-17, 300 mg, 0.90 mmol) in N,N-dimethylformamide (5 mL) was added EDCI (350 mg, 1.83 mmol), HOBt (240 mg, 1.78 mmol) and DIEA (350 mg, 2.71 mmol) at room temperature. The resulting solution was stirred at room temperature for 10 minutes. Then to the solution was added (2S,4R)-1-[(2S)-2-amino-3,3-dimethylbutanoyl]-4-hydroxy-N-{[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}pyrrolidine-2-carboxamide (ULM-1, 390 mg, 0.91 mmol), and the resulting solution was stirred at room temperature for 1 hour. Water (30mL) was added and the resulting mixture was extracted with ethyl acetate (30 mL x 3). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (30 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by a flash silica gel chromatography (eluent: dichloromethane/methanol (v:v = 10:1) to give BQ
(yield: 64%) as a yellow solid. LC-MS (ES+): m/z 745.35 [MH+], tR = 0.96 min (2.0 minute run).
[0570] Step 2: Synthesis of (2S,4R)-1-[(2S)-2-[2-(3-{2-[4-(4-{3-[4-cyano-3- (trifluoromethyl)phenyl]-5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)piperidin- 1-yl]ethoxy}propoxy)acetamido]-3,3-dimethylbutanoyl]-4-hydroxy-N-{[4-(4-methyl-1,3-thiazol- 5-yl)phenyl]methyl}pyrrolidine-2-carboxamide (Exampl 80)
[0571] To a stirred solution of 4-{4,4-dimethyl-5-oxo-3-[4-(piperidin-4-yl)phenyl]-2- sulfanylideneimidazolidin-1-yl}-2-(trifluoromethyl)benzonitrile (ABM-25, 150 mg, 0.32 mmol), (2S,4R)-1-[(2S)-3,3-dimethyl-2-[2-(3-{2-[(4-methylbenzenesulfonyl)
oxy]ethoxy}propoxy)acetamido]butanoyl]-4-hydroxy-N-{[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}pyrrolidine-2-carboxamide (BQ, 236 mg, 0.32 mmol) in N,N- dimethylformamide (5 mL) was added potassium carbonate (131 mg, 0.95 mmol). The resulting mixture was stirred at 60 oC overnight. The reaction mixture was cooled to room temperature, water (20mL) was added and the resulting mixture was extracted with ethyl acetate (30 mL x 3). The organic layers were combined, washed with saturated aqueous solution of sodium chloride (20 mL), dried over anhydrous sodium sulfate and then concentrated under reduced pressure to give a crude residue, which was purified by Prep-HPLC to give Example 80 (yield: 7%) as a white solid.1H NMR (300 MHz, CD3OD): ^ 8.91 (s, 1H), 8.15 (d, J = 4.5 Hz, 2H), 8.02 (d, J = 4.5 Hz, 1H), 7.40 (m, 7H), 4.45 (d, J = 12.0 Hz, 1H), 4.45 (m, 4H), 4.02 (d, J = 3.9 Hz, 2H), 3.70 (m, 10H), 3.38 (m, 2H), 3.11 (m, 3H), 2.48 (s, 3H), 2.26 (m, 8H), 1.54 (s, 6H), 1.03 (s, 9H); LC- MS (ES+): m/z 1045.35 [MH+], tR = 2.74 min (5.6 minute run).
[0572] Example 81 was synthesized according to similar procedure described for synthesis of Example 80, by using corresponding starting materials and intermediates.
[0573] Example 81: (2S,4R)-1-((S)-2-(2-(4-(2-(4-(4-(3-(4-cyano-3-(trifluoromethyl)phenyl)- 5,5-dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)phenyl)piperidin-1- yl)ethoxy)butoxy)acetamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5- yl)benzyl)pyrrolidine-2-carboxamide
Figure imgf000183_0001
;
[0575] 1H NMR (300 MHz, DMSO): ^ 8.98 (s, 1H), 8.63-8.61 (m, 1H), 8.40-8.37 (m, 1H), 8.37-8.34 (m, 1H), 8.11-8.01(m, 1H), 7.44-7.40 (m, 3H), 7.37-7.32 (m, 6H), 4.57-4.54 (d, J = 9.6 Hz, 1H), 4.47-4.45 (m, 2H), 4.45-4.44 (m, 2H), 4.39-4.37 (m, 1H), 3.92 (s, 2H), 3.71-3.65 (m, 2H), 3.58-3.47 (m, 5H), 3.45-3.40 (m, 4H), 2.99-2.95 (m, 2H), 2.51 (s, 3H), 2.12-2.02 (m, 3H), 1.93-1.90 (m, 1H), 1.90-1.79 (m, 3H), 1.77-1.71 (m, 5H), 1.67-1.61 (m, 6H), 0.94 (s, 9H); Mass (ES+): m/z 1059.44 [MH+].
[0576] Example 82: (2S,4R)-N-(2-(2-(2-(2-(4-(3-(4-cyano-3-(trifluoromethyl)phenyl)-5,5- dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)phenoxy)ethoxy)ethoxy)ethoxy)-4-(4- methylthiazol-5-yl)benzyl)-4-hydroxy-1-((S)-3-methyl-2-(1-oxoisoindolin-2- yl)butanoyl)pyrrolidine-2-carboxamide:
Figure imgf000184_0001
.
[0577] Step 1: Synthesis of 4-[3-(4-{2-[2-(2-hydroxyethoxy)ethoxy]ethoxy}phenyl)-4,4- dimethyl-5-oxo-2--ulfanylideneimidazolidin-1-yl]-2-(trifluoromethyl)benzonitrile (BR)
[0578] To a stirred solution of 4-[3-(4-hydroxyphenyl)-4,4-dimethyl-5-oxo-2- sulfanylideneimidazolidin-1-yl]-2-(trifluoromethyl)benzonitrile (ABM-3, 405 mg, 1.00 mmol) in CH3CN (20 mL) was added potassium carbonate (276 mg, 1.98 mmol) and 2-(2-{2-[(4- methylbenzenesulfonyl)oxy]ethoxy}ethoxy)ethan-1-ol (L-30, 456 mg, 1.50 mmol) at room temperature. The resulting mixture was then heated to 80 oC and stirred at this temperature overnight. LC-MS indicated formation of the desired product. The reaction mixture was cooled to room temperature, concentrated under vacuum to give a crude residue, which was purified by a flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v = 1:1)) to give BR (yield: 91%) of as a brown oil.
[0579] Step 2: Synthesis of 2-{2-[2-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5-dimethyl-4- oxo-2-sulfanylideneimidazolidin-1-yl}phenoxy)ethoxy]ethoxy}ethyl 4-methylbenzene-1- sulfonate (BS)
[0580] To a stirred solution of 4-[3-(4-{2-[2-(2-hydroxyethoxy)ethoxy]ethoxy}phenyl)-4,4- dimethyl-5-oxo-2--ulfanylideneimidazolidin-1-yl]-2-(trifluoromethyl)benzonitrile (BR, 490 mg, 0.91 mmol) in dichloromethane (10 mL) was added tosyl chloride (190 mg, 1.00 mmol), potassium iodide (30.2 mg) and silver oxide (314 mg) at room temperature. The resulting mixture was then stirred at 30oC for 6h, LC-MS indicated formation of the desired product. The inorganic salts were removed from the reaction by filtration, the solution phase was concentrated under vacuum to give a crude residue, which was purified by a flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v = 1:3)) to give BS (yield: 60%) of as a light yellow solid.
[0581] Step 3: Synthesis of (2S,4R)-4-(tert-butoxy)-N-{[2-(2-{2-[2-(4-{3-[4-cyano-3- (trifluoromethyl)phenyl]-5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1- yl}phenoxy)ethoxy]ethoxy}ethoxy)-4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl}-1-[(2S)-3- methyl-2-(1-oxo-2,3-dihydro-1H-isoindol-2-yl)butanoyl]pyrrolidine-2-carboxamide (BT)
[0582] To a stirred solution of 2-{2-[2-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5-dimethyl- 4-oxo-2-sulfanylideneimidazolidin-1-yl}phenoxy)ethoxy]ethoxy}ethyl 4-methylbenzene-1- sulfonate (BS, 207 mg, 0.30 mmol) and (2S,4R)-4-(tert-butoxy)-N-{[2-hydroxy-4-(4-methyl-1,3- thiazol-5-yl)phenyl]methyl}-1-[(2S)-3-methyl-2-(1-oxo-2,3-dihydro-1H-isoindol-2- yl)butanoyl]pyrrolidine-2-carboxamide (ULM-12, 181 mg, 0.30 mmol) in N,N- dimethylformamide (2 mL) was added potassium carbonate (83 mg, 0.60 mmol) at room temperature. The resulting mixture was then heated to 80 oC and stirred at the same temperature overnight, and LC-MS indicated formation of the desired product. The reaction was then cooled to room temperature, diluted by water (10 mL) and then extracted with ethyl acetate (20 mL x 3). The organic layers were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a crude residue, which was purified by a flash silica gel chromatography (eluent: ethyl acetate/petroleum ether (v:v = 1 : 1) to give BT (yield: 54%) as a white solid.
[0583] Step 4: Synthesis of (2S,4R)-N-{[2-(2-{2-[2-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]- 5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenoxy)ethoxy]ethoxy}ethoxy)-4-(4- methyl-1,3-thiazol-5-yl)phenyl]methyl}-4-hydroxy-1-[(2S)-3-methyl-2-(1-oxo-2,3-dihydro-1H- isoindol-2-yl)butanoyl]pyrrolidine-2-carboxamide (Example 82)
[0584] To a stirred solution of (2S,4R)-4-(tert-butoxy)-N-{[2-(2-{2-[2-(4-{3-[4-cyano-3- -trifluoromethyl)phenyl]-5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1- yl}phenoxy)ethoxy]ethoxy}ethoxy)-4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl}-1-[(2S)-3^ methyl-2-(1-oxo-2,3-dihydro-1H-isoindol-2-yl)butanoyl]pyrrolidine-2^carboxamide (BT, 180 mg, 0.16 mmol) in dichloromethane (5 mL) was added trifluoroacetic acid (0.5 mL) at room temperature. The resulting solution was stirred room temperature for 6 hours, LC-MS indicated formation of the desired product. Saturated aq. solution of sodium bicarbonate was added to the reaction to neutralize the trifluoroacetic acid. Organic layer was separated, the aqueous layer was extracted with of dichloromethane (10 mL x 2). The organic layers combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a crude residue, which was purified by Pre-HPLC to give Example 82 (yield: 31%) as a white solid.1H NMR
(400MHz, CD3OD): ^ 8.90 (s, 1 H), 8.40-8.38 (d, J = 8.0Hz, 2 H), 8.29 (s, 1 H), 8.09-8.07 (d, J = 8.4 Hz, 1 H), 7.72-7.70 (d, J = 7.6Hz, 1 H), 7.62-7.61 (d, J = 4.0Hz, 2 H), 7.50-7.40(m, 1H), 7.35-7.33 (d, J = 7.6Hz, 1H), 7.27-7.25 (d, J =8.8Hz, 2 H), 7.10-7.06 (m, 3H), 7.05-7.00 (m, 1H), 5.09 (s, 1H), 4.72-4.69 (d, J =10.8 Hz, 1H), 4.61 -4.41 (m, 2H), 4.41 -4.31 (m, 2H), 4.31 - 4.21 (m, 2H), 4.21 -4.11 (m, 2H), 4.11 -4.01 (m, 2H), 3.82-3.71 (m, 5H), 3.69-3.61 (m, 5H), 2.51 (m, 3H), 2.47-2.25 (m, 1 H), 2.10-2.00 (m, 1H), 2.00-1.95 (m, 1H), 1.48 (s, 6 H), 0.97- 0.96 (d, J = 6.4Hz, 3H), 0.74-0.72 (d, J = 6.4Hz, 3H); LC-MS (ES+): m/z 1068.20 [MH+], tR = 1.59 min (3.0 minute run).
[0585] Examples 83-85 were synthesized according to similar procedure described for synthesis of Example 82, by using corresponding starting materials and intermediates.
[0586] Table 6. Exemplary Compounds.
Figure imgf000186_0001
Figure imgf000187_0001
[0587] Synthesis of example 86.
Figure imgf000188_0001
[0588] Step 1: Synthesis of tert-butyl 3-{2-[(5-bromopyridin-2-yl)oxy]ethoxy}propanoate:
Figure imgf000188_0002
.
[0589] To a stirred solution of 5-bromopyridin-2-ol (3.0 g, 17.24 mmol), tert-butyl 3-(2- hydroxyethoxy)propanoate (3.3 g, 17.19 mmol) and triphenylphosphine (6.8 g, 25.81 mmol) in tetrahydrofuran (120.0 mL) was added diethyl diazene-1,2-dicarboxylate (4.49 g, 25.78 mmol) dropwise at 0 oC under an atmosphere of nitrogen. The resulting solution was stirred overnight at room temperature. The reaction mixture was concentrated under reduced pressure to give a crude residue, which was purified by silica gel flash chromatography (eluent: ethyl acetate/petroleum ether, v/v =1/3) to provide the titled product (yield: 50%) as colorless oil.
[0590] Step 2: Synthesis of tert-butyl 3-(2-{[5-(4-nitrophenyl)pyridin-2- yl]oxy}ethoxy)propanoate:
Figure imgf000188_0003
[0591] To a stirred mixture of tert-butyl 3-{2-[(5-bromopyridin-2-yl)oxy]ethoxy}propanoate (3.0 g, 8.67 mmol) and (4-nitrophenyl)boronic acid (1.5 g, 8.87 mmol) in a mixed solvent of dioxane (90.0 mL) and water (9.0 mL) was added potassium carbonate (2.4 g, 17.36 mmol) and Pd(PPh3)4 (450.0 mg, 0.39 mmol) under an atmosphere of nitrogen. The resulting mixture was stirred for 12 hours at 100 oC. The bulk of solvent was removed under reduced pressure, and the resulting aqueous residue was extracted with ethyl acetate (100 mL x 2). The organic layers were combined, washed with brine (70 mL x 2), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash
chromatography (eluent: ethyl acetate/petroleum ether, v/v =1/5) to provide the titled product (yield: 83%) a yellow solid. Mass (ES+): m/z 389.00 [MH+].
[0592] Step 3: Synthesis of tert-butyl 3-(2-{[5-(4-aminophenyl)pyridin-2- yl]oxy}ethoxy)propanoate:
Figure imgf000189_0001
.
[0593] To a stirred solution of tert-butyl 3-(2-{[5-(4-nitrophenyl)pyridin-2- yl]oxy}ethoxy)propanoate (2.8 g, 7.21 mmol) in ethanol (200.0 mL) under an atmosphere of nitrogen was added palladium on carbon (1.5 g) at room temperature. The reaction mixture was then charge with hydrogen gas and stirred at room temperature for 12 hours. The solids were removed by filtration and the solution phase was concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluent: ethyl
acetate/petroleum ether, v/v=1/3) to provide the titled product (yield: 89%) a yellow oil. LC-MS (ES+): m/z 358.97 [MH+].
[0594] Example 86 was synthesized from tert-butyl 3-(2-{[5-(4-aminophenyl)pyridin-2^ yl]oxy}ethoxy)propanoate, according to chemistry highlighted above (steps 4-8), utilizing similar procedures described for the similar chemistry carried out for the synthesis of examples 67, 75, 103, by using corresponding starting materials and intermediates.
[0595] Example 90 was synthesized according to similar procedures described for the synthesis of examples 86, by using corresponding starting materials and intermediates. [0596] Examples 88, 91-92 were synthesized according to similar procedures described for the synthesis of examples 80, 75, 103, by using corresponding starting materials and intermediates.
[0597] Examples 87, 89, 93-102, 104-134, 136-142, 146-149 were synthesized according to similar procedures described for the synthesis of examples 75, by using corresponding starting materials and intermediates.
[0598] Table 7. Exemplary Compounds.
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
- - . ,- - .
Figure imgf000197_0001
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000200_0001
Figure imgf000201_0001
Figure imgf000202_0001
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000206_0001
. , , . , . , , , , .
[MH+]
Figure imgf000207_0001
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0001
[0599] Examples 135, 143-145 were synthesized according to similar procedures described for the synthesis of examples 103, by using corresponding starting materials and intermediates.
[0600] Synthesis of example 103:
Figure imgf000211_0001
Figure imgf000211_0002
Figure imgf000211_0003
.
[0601] Step 1: Synthesis of tert-butyl N-[4-(4-aminophenyl)phenyl]carbamate: .
[0602] To a stirred solution of 4-(4-aminophenyl)aniline (15.0 g, 81.42 mmol) in a mixed solvent of N,N-dimethylformamide / tetrahydrofuran /water (v/v/v = 100/300/50 mL) was added potassium carbonate (9.5 g, 68.74 mmol) and di-tert-butyl dicarbonate (13.67 g, 62.63 mmol) at room temperature. The resulting mixtire was stirred for 5h at room temperature. The reaction was then diluted by water (500 mL) and extracted with ethyl acetate (200 mL x 3). The organic layers were combined, washed with brine (50 mL x 2), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluent: ethyl acetate/petroleum ether, v:v = 1:2) to provide the titled product (yield: 97%) as a yellow solid.
[0603] Step 2: Synthesis of tert-butyl N-(4-{4-[(1-cyano-1- methylethyl)amino]phenyl}phenyl)carbamate:
. [0604] To a stirred solution of tert-butyl N-[4-(4-aminophenyl)phenyl]carbamate (7.0 g, 24.62 mmol) in acetone (100 mL) under an atmosphere of nitrogen was added
trimethylsilanecarbonitrile (4.9 g, 49.49 mmol) drop wise at 0 °C, followed by addition of iodine (630.0 mg, 2.48 mmol) in several batches at 0 °C. The resulting mixture was stirred for 15h at room temperature. The reaction was then quenched by the addition of water (100 mL), and the resulting solution was extracted with ethyl acetate (100 mL x 2). The organic layers were combined, washed with brine (70 mL x 2), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash
chromatography (eluent: ethyl acetate/petroleum ether, v:v = 1:3) to provide the titled product (yield: 87%) as a yellow solid. Mass (ES+): m/z 352.20 [MH+].
[0605] Step 3: Synthesis of tert-butyl N-[4-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-4-imino- 5,5-dimethyl-2-sulfanylideneimidazolidin-1-yl}phenyl)phenyl]carbamate:
Figure imgf000212_0001
.
[0606] To a stirred solution of tert-butyl N-(4-{4-[(1-cyano-1- methylethyl)amino]phenyl}phenyl)carbamate (3.1 g, 8.82 mmol) in toluene (40.0 mL) was added 4-dimethylaminopyridine (1.6 g, 13.10 mmol) and 4-isothiocyanato-2- (trifluoromethyl)benzonitrile (2.0 g, 8.76 mmol) at room temperature under an atmosphere of nitrogen. The resulting solution was stirred for 12h at 100°C in an oil bath. The reaction mixture was then concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluent: ethyl acetate/petroleum ether, v:v = 1:1) to provide the titled product (yield: 36%) as a yellow solid. Mass (ES+): m/z 580.30 [MH+].
[0607] Step 4: Synthesis of 4-{3-[4-(4-aminophenyl)phenyl]-4,4-dimethyl-5-oxo-2- sulfanylideneimidazolidin-1-yl}-2-(trifluoromethyl)benzonitrile:
Figure imgf000212_0002
. [0608] To a stirred solution of tert-butyl N-[4-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-4- imino-5,5-dimethyl-2-sulfanylideneimidazolidin-1-yl}phenyl)phenyl]carbamate (2.0 g) in methanol (20 mL) was added hydrogen chloride (3 N solution in water, 5 mL) at room
temperature. The resulting solution was stirred for 2 h at 70°C in an oil bath. The reaction mixture was then concentrated under reduced pressure to remove the bulk of methanol. To the resulting aqueous mixture was added sodium bicarbonate (sat. aqueous solution) to adjust the pH to ~ 8, and the resulting mixture was extracted with ethyl acetate (80 mL x 3). The organic layers were combined, washed with brine (30 mL x 2), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash
chromatography (eluent: ethyl acetate/petroleum ether, v:v = 1:2) to provide the titled product (yield: 45%) as a yellow solid. Mass (ES+): m/z 481.15 [MH+].
[0609] Step 5: Synthesis of tert-butyl 2-(4-{[4-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)phenyl]amino}butoxy)acetate:
Figure imgf000213_0001
.
[0610] To a stirred solution of 4-{3-[4-(4-aminophenyl)phenyl]-4,4-dimethyl-5-oxo-2- sulfanylideneimidazolidin-1-yl}-2-(trifluoromethyl)benzonitrile (200.0 mg, 0.42 mmol) in dichloromethane (10 mL) was added acetic acid (0.01 mL) and tert-butyl 2-(4-oxobutoxy)acetate (93.0 mg, 0.46 mmol) at room temperature. The resulting mixture was stirred for 10 min at room temperature, then to the mixture was added sodium triacetoxyborohydride (124.0 mg, 0.59 mmol). The resulting mixture was stirred overnight at room temperature. The reaction mixture was diluted by water (30 mL), extracted with dichloromethane (20 mL x 3). The organic layers were combined, washed with brine (20 mL x 2), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluent: ethyl acetate/petroleum ether, v:v = 1:2) to provide the titled product (yield: 36%). Mass (ES+): m/z 667.20[MH+].
[0611] Step 6: Synthesis of 2-(4-{[4-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5-dimethyl-4- oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)phenyl]amino}butoxy)acetic acid:
Figure imgf000214_0001
.
[0612] To a stirred solution of tert-butyl 2-(4-{[4-(4-{3-[4-cyano-3-(trifluoromethyl)phenyl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)phenyl]amino}butoxy)acetate (100.0 mg, 0.15 mmol) in dichloromethane (10 mL) was added trifluoroacetic acid (2.0 mL) at room temperature. The resulting solution was stirred for 2h at room temperature. The reaction mixture was then concentrated under reduced pressure to give a crude material (yield: 99% based on crude) which was used for next step reaction without any further purification. Mass (ES+): m/z 611.10[MH+]
[0613] Step 7: Synthesis of example 103.
[0614] This compound was synthesized from 2-(4-{[4-(4-{3-[4-cyano-3- (trifluoromethyl)phenyl]-5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1- yl}phenyl)phenyl]amino}butoxy)acetic acid and (2S,4R)-1-[(2S)-2-amino-3,3-dimethylbutanoyl]- 4-hydroxy-N-{[4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl}pyrrolidine-2^carboxamide hydrochloride, according to similar procedures in the last step (amide coupling) described for the synthesis of example 75.
[0615] Synthesis of tert-butyl 2-(4-oxobutoxy) acetate:
Figure imgf000214_0002
.
[0616] To a stirred solution of tert-butyl 2-(4-hydroxybutoxy)acetate (1.0 g, 4.90 mmol) in dichloromethane (10 mL) was added (1,1,1-triacetoxy)-1,1-dihydro-1,2-benziodoxol-3(1H)-one (2.7 g, 6.37 mmol) at room temperature. The resulting mixture was stirred for 12h at room temperature. The reaction mixture was then diluted with water (20 mL), extracted with ethyl acetate (20 mL x 3). The organic layers were combined, washed with brine (20 mL x 2), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluent: ethyl acetate/petroleum ether, v/v = 1:2) to provide the titled product (yield: 50%) as colorless oil.1H NMR (300 MHz, CD3OD) ^ 9.68 (s, 1H), 3.95 (s, 2H), 3.48-3.45 (m, 2H), 2.51-2.50 (m, 2H), 1.81-1.63 (m, 2H), 1.42 (s, 9H). [0617] Table 8. Exemplary Compounds.
Figure imgf000215_0001
Figure imgf000216_0001
Figure imgf000217_0001
Figure imgf000218_0001
[0618] Synthesis of example 150:
Figure imgf000218_0002
.
[0619] Step 1: Synthesis of methyl 4-{[5-({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(1,3-thiazol-5- yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)pentyl]oxy}benzoate:
Figure imgf000219_0001
[0620] To a stirred solution of 2-({5-[4-(methoxycarbonyl)phenoxy]pentyl}oxy)acetic acid (200 mg), (2S,4R)-1-[(2S)-2-amino-3,3-dimethylbutanoyl]-4-hydroxy-N-{[4-(1,3-thiazol-5- yl)phenyl]methyl}pyrrolidine-2-carboxamide hydrogen chloride salt (149 mg, 0.32 mmol), N- ethyl-N-isopropylpropan-2-amine (185 mg, 1.44 mmol) in anhydrous N,N-dimethylformamide (5 mL) was added HATU (2-(7-aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium
hexafluorophosphate ) (203 mg, 0.54 mmol) at 0 °C. The resulting mixture was allowed to warm up to room temperature and stirred at room temperature for 20 min. TLC and LC-MS showed formation of the desired product. The mixture was partitioned between ethyl acetate (100 mL) and water (50 mL). The organic layer was collected, washed with brine (50 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluent 2% methanol in methylene dichloride) to afford the titled product (yield 25%, 2 steps) as a white solid. Mass: (ES+): m/z 695.30 [M+H+].
[0621] Step 2: Synthesis of 4-{[5-({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(1,3-thiazol-5- yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)pentyl]oxy}benzoic acid:
[0622] To a stirred solution of methyl 4-{[5-({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(1,3-thiazol-5- yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)pentyl]oxy}benzoate (150 mg, 0.22 mmol) in a mixed solvents of tetrahydrofuran (4 mL)-water (2 mL)-methanol (1 ml) was added lithium hydroxide monohydrate (36 mg, 0.86 mmol) at room temperature. The resulting mixture was stirred at 35°C overnight. TLC and LC-MS showed formation of the desired product. The reaction mixture was acidified with aqueous HCl (3N) to pH =3-4 and extracted with methylene dichloride (50 mL × 2). The organic layers were combined, washed with brine, dried over Na2SO4 and concentrated to afford the titled product (110 mg, crude) as a white solid which was used for next step without further purification. Mass: (ES+): m/z 681.20 [M+H+].
[0623] Step 3: Synthesis of exmaple 150:
Figure imgf000220_0001
.
[0624] To a stirred mixture of 4-{[5-({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(1,3-thiazol-5- yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)pentyl]oxy}benzoic acid (110 mg, 0.16 mmol), 2-chloro-4-[trans-3- amino-2,2,4,4-tetramethylcyclobutoxy]benzonitrile hydrogen chloride salt (50 mg, 0.16 mmol), N-ethyl-N-isopropylpropan-2-amine (77 mg, 0.64 mmol) in anhydrous N,N-dimethylformamide (4 mL) was added HATU ((2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate )) (68 mg, 0.18 mmol) at 0 °C. The resulting mixture was allowed to warm up to room temperature and stirred at room temperature for 20 min. TLC and LC-MS showed formation of the desired product. The reaction mixture was partitioned between ethyl acetate (100 mL) and water (40 mL). The organic phase was separated, washed with brine (50mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by preparative TLC (eluent: 5% methanol in methylene dichloride) to afford the titled product (yield 25%, 2 steps) as a white solid.
[0625] Synthesis of 2-({5-[4-(methoxycarbonyl)phenoxy]pentyl}oxy)acetic acid
Figure imgf000220_0002
.
[0626] Step 1: Synthesis of tert-butyl 2-{[5-(benzyloxy)pentyl]oxy}acetate:
Figure imgf000220_0003
. [0627] To a stirred mixture of 5-(benzyloxy)pentan-1-ol (10 g, 51.5 mmol), tert-butyl 2- bromoacetate (40.2 g, 206 mmol) and tetrabutyl ammonium chloride (14.2 g, 51.5 mmol) in methylene dichloride (60 mL) was added sodium hydroxide (40 ml, 35% in water) at room temperature, and the resulting mixture was stirred at room temperature for 16 hours. The reaction mixture was then partitioned between methylene dichloride (200 mL) and water (100 mL). The organic layer was collected and washed with brine (50 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluent: 5% ethyl acetate in hexane) to afford tert-butyl 2-{[5- (benzyloxy)pentyl]oxy}acetate (yield 31.6%) as light yellow oil. LC-MS: (ES+): m/z 331.10 [M+Na+], 1H NMR (400 MHz, CDCl3): ^ 1.48 (s, 9H), 1.63-1.67 (m, 6H), 3.46-3.53 (m, 4H), 4.10 (s, 2H), 4.50 (s, 2H), 7.28-7.34 (m, 5H).
[0628] Step 2: Synthesis of tert-butyl 2-[(5-hydroxypentyl)oxy]acetate:
Figure imgf000221_0001
.
[0629] To a stirred solution of tert-butyl 2-{[5-(benzyloxy)pentyl]oxy}acetate (5 g, 16.2 mmol) in ethanol (100 ml) under a nitrogen atmosphere was added palladium on carbon (10%, 600 mg) at room temperature. The resulting mixture was stirred at 50°C overnight under hydrogen atmosphere (hydrogen balloon). TLC showed formation of desired product. Palladium on carbon was removed through filtration and washed with ethyl acetate (50 mL). The filtrate was concentrated under reduced pressure to afford tert-butyl 2-[(5-hydroxypentyl)oxy]acetate (2.5 g, crude) as colorless oil which was used in next step without further purification.
[0630] Step 3: Synthesis of tert-butyl 2-({5-[(4-methylbenzenesulfonyl)oxy]pentyl}oxy)acetate:
Figure imgf000221_0002
.
[0631] To a stirred solution of tert-butyl 2-[(5-hydroxypentyl)oxy]acetate (2.5 g, crude) and triethylamine (3.5 g, 34.5 mmol) in anhydrous methylene dichloride (50 mL) was added a solution of 4-toluenesulfonyl chloride (2.7 g, 13.8 mmol) in anhydrous methylene dichloride (8 mL) drop wise at 0 °C. The reaction mixture was allowed to warm up to room temperature and stirred at room temperature overnight. TLC showed formation of desired product. The mixture was quenched with aqueous solution of potassium carbonate (1N, 50 mL) at room temperature and extracted with ethyl acetate (50 mL × 3). The organic layers were combined, washed with brine (50 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluent: 1% methanol in methylene dichloride) to afford tert-butyl 2-({5-[(4- methylbenzenesulfonyl)oxy]pentyl}oxy)acetate (yield 35.1%) as colorless oil. Mass: (ES+): m/z 395.10 [MNa+].
[0632] Step 4: Synthesis of methyl 4-({5-[2-(tert-butoxy)-2-oxoethoxy]pentyl}oxy)benzoate:
Figure imgf000222_0001
[0633] To a stirred mixture of tert-butyl 2-({5-[(4- methylbenzenesulfonyl)oxy]pentyl}oxy)acetate (1.0g, 2.7 mmol) and potassium carbonate (266 mg, 1.6 mmol) in acetonitrile (15 mL) was added methyl 4-hydroxybenzoate (500 mg, 3.29 mmol) at room temperature. The resulting mixture was refluxed overnight. TLC showed formation of desired product. The reaction mixture was cooled to room temperature, and partitioned between ethyl acetate (150 mL) and water (50 mL). The organic layer was washed with washed with brine (50 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluent 10% ethyl acetate in hexane) to afford methyl 4-({5-[2-(tert-butoxy)-2- oxoethoxy]pentyl}oxy)benzoate (yield 33%) as colorless oil. Mass (ES+): m/z 353.10 [M+Na+]; 1H NMR (400 MHz, CDCl3): ^ 1.48 (s, 9H), 1.55-1.61 (m, 2H), 1.68-1.72 (m, 2H), 1.80-1.87 (m, 2H), 3.55 (t, J = 6.4 Hz, 2H), 3.88 (s, 3H), 3.96 (s, 2H), 4.02 (t, J = 6.4 Hz, 2H), 6.89 (d, J = 9.2 Hz, 2H), 7.97 (d, J = 9.2 Hz, 2H).
[0634] Step 5: Synthesis of 2-({5-[4-(methoxycarbonyl)phenoxy]pentyl}oxy)acetic acid:
Figure imgf000222_0002
.
[0635] To a stirred solution of methyl 4-({5-[2-(tert-butoxy)-2-oxoethoxy]pentyl}oxy)benzoate (300 mg, 0.85 mmol) in DCM (4 mL) was added and TFA (2 ml) at room temperature, the resulting solution was stirred at room temperature for 1 hour. TLC showed formation of the desired product. The solvent was evaporated to afford 2-({5-[4- (methoxycarbonyl)phenoxy]pentyl}oxy)acetic acid (200 mg, crude) as yellow oil which was used in next step without further purification.
[0636] Examples 151-157 were synthesized according to similar procedure described for synthesis of example 150, by using corresponding starting materials and intermediates.
[0637] Table 9. Exemplary Compounds.
Figure imgf000223_0001
Figure imgf000224_0001
Figure imgf000225_0001
Figure imgf000226_0001
ass : m z .
[0638] Synthesis of example 163:
Figure imgf000226_0002
.
[0639] Step 1: synthesis of methyl 4-[4-({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)butoxy]benzoate
Figure imgf000227_0001
[0640] To a stirred solution of 2-{4-[4-(methoxycarbonyl)phenoxy]butoxy}acetic acid (22.0 mg, 77.9 µmol) and (2S,4R)-1-[(2S)-2-amino-3,3-dimethylbutanoyl]-4-hydroxy-N-[(1S)-1-[4-(4-methyl-1,3-thiazol- 5-yl)phenyl]ethyl]pyrrolidine-2-carboxamide hydrochloride (36.3 mg, 77.9 µmol) in methylene chloride (2.0 mL) was added O-(benzotriazol-1-yl)-N,N,N4,N4-tetramethyluronium tetrafluoroborate (25.0 mg, 77.9 µmol) and diisopropylethylamine (40.5 µL, 233 µmol) at room temperature. The reaction mixture was stirred at room temperature for 30 minutes, LC-MS indicated formation of the desired product. The reaction mixture was concentrated under reduced pressure. The crude material was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO (gradient eluent: Heptane/Acetone (v:v = 100:0 to 0:100)) to give the titled product (yield: 78%) as a white solid. LC-MS (ES+): m/z 695.3138 [MH+].
[0641] Step 2: Synthesis of 4-[4-({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)butoxy]benzoic acid:
Figure imgf000227_0002
.
[0642] To a stirred solution of methyl 4-[4-({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol- 5-yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)butoxy]benzoate (42.4 mg, 61.0 µmol) in methanol (2.0 mL) was added 1 M NaOH in water (0.5 mL, 12.5 mmol) at room temperature. The reaction mixture was stirred at room temperature for 16 hours. LC-MS indicated formation of the desired product. The reaction mixture was quenched with 1.0 M aqueous HCl and then concentrated under reduced pressure to remove the methanol. The aqueous residue was extracted with EtOAc (15 mL x 2). The organic layer was washed with brine (5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give the titled product (yield: 82%) as a white solid. The material was used in next step without any further purification. Mass (ES+): m/z 681.2986 [MH+].
[0643] Step 3: Synthesis of example 163: [0644] To a stirred solution of 2-chloro-4-[trans-3-amino-2,2,4,4- tetramethylcyclobutoxy]benzonitrile (13.9 mg, 50.2 µmol) and 4-[4-({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4- (4-methyl-1,3-thiazol-5-yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)butoxy]benzoic acid (34.2 mg, 50.2 µmol) in methylene chloride (2.0 mL) was added O-(benzotriazol-1-yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate (16.1 mg, 50.2 µmol) and diisopropylethylamine (26.0 µL, 150 µmol) at room temperature. The reaction mixture was stirred at room temperature for 1.5 hours. LC-MS indicated formation of the desired product. The reaction mixture was quenched with water (5 mL) and extracted with DCM (15 mL x 3). The organic layers were combined, washed with aqueous NaHCO3 (5 mL), brine (5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO (eluent: DCM/MeOH (v:v = 90:10)) to give the titled product (yield: 39%) as an off white solid.
[0645] Synthesis of 2-{4-[4-(methoxycarbonyl)phenoxy]butoxy}acetic acid:
Figure imgf000228_0001
.
[0646] Step 1: synthesis of tert-butyl 2-{4-[(4-methylbenzenesulfonyl)oxy]butoxy}acetate:
Figure imgf000228_0002
.
[0647] This material was synthesized from tert-butyl 2-(4^hydroxybutoxy)acetate and 4-toluenesulfonyl chloride according to similar procedures described above for the synthesis of tert-butyl 2-({5-[(4- methylbenzenesulfonyl)oxy]pentyl}oxy)acetate.
[0648] Step 2: synthesis of methyl 4-{4-[2-(tert-butoxy)-2-oxoethoxy]butoxy}benzoate.
[0649] To a stirred mixture of methyl 4-hydroxybenzoate (27.99 mg, 184.0 µmol) and tert-butyl 2-{4- [(4-methylbenzenesulfonyl)oxy]butoxy}acetate in acetonitrile (2.0 mL) was added potassium
carbonate (34.67 mg, 250.9 µmol) at room temperature. The reaction mixture was then stirred at 80 ºC for 16 hours. LC-MS indicated formation of the desired product. The reaction mixture was concentrated under reduced pressure to give a crude residue, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO (gradient eluent: heptane/acetone (v:v = 100:0 to 50:50)) to give the titled product (yield: 94%) as a clear oil. Mass (ES+): m/z 361.16 [M+Na].
[0650] Step 3: Synthesis of Synthesis of 2-{4-[4-(methoxycarbonyl)phenoxy]butoxy}acetic acid:
[0651] To a stirred solution of methyl 4-{4-[2-(tert-butoxy)-2-oxoethoxy]butoxy}benzoate (53.1 mg, 156 µmol) in dichloromethane (1.0 mL) was added trifluoroacetic acid (1.0 mL, 12.9 mmol) at room temperature. The reaction mixture was then stirred at room temperature for 30 minutes. LC-MS indicated formation of the desired product. The reaction mixture was concentrated under reduced pressure to give the titled product (yield: 99% based on crude material) as an off white solid. The crude material was then used in next step without any further purification. Mass (ES+): m/z 305.10.
[0652] Examples 162, 164-171 were synthesized according to similar procedure described for synthesis of example 163, by using corresponding starting materials and intermediates.
[0653] Table 10. Exemplary Compounds.
Figure imgf000229_0001
Ex
Structure Compound name and Analytical data #
(2S,4R)^1^[(2S)^3,3^dimethyl^2^[2^({5^[(4^{[trans^3^(3^chloro^4^cyanophenoxy)^ 2,2,4,4^
tetramethylcyclobutyl]carbamoyl}phenyl)amino]pentyl}oxy)acetamido]butanoyl]^4^ hydroxy^N^[(1S)^1^[4^(4^methyl^1,3^thiazol^5^yl)phenyl]ethyl]pyrrolidine^2^ carboxamide
1H NMR (300 MHz, CD3OD):^8.88 (s, 1 H), 7.80-7.65 (m, 3 H), 7.50-7.33 (m, 4 H), 7.16 (s, 1H), 7.03-6.93 (m, 1 H), 6.54-6.43 (m, 2 H), 5.02– 4.95 (m, 1 H), 4.67 (s, 1 H), 4.65-4.50 (m, 1 H), 4.46-4.40 (m, 1 H), 4.29-4.25 (m, 1 H), 4.20-4.15 (m, 1 H), 4.04- 3.90 (m, 2 H), 3.89-3.85 (m, 1 H), 3.80-3.73 (m, 1 H), 3.66-3.52 (m, 2H), 3.20-3.10 (m, 2 H), 2.40 (s, 3 H), 2.25-1.95 (m, 1 H), 2.02– 1.90 (m, 1 H), 1.80-1.68 (m, 4 H), 1.65-1.50 (m, 2 H), 1.49-1.43 (m, 2H), 1.30-1.23 (m, 6 H), 1.22-1.15 (m, 6 H), 1.01 (s, 174 9 H); LC-MS (ES+): m/z, 968.40 [MH+]
(2S,4R)^1^[(2S)^3,3^dimethyl^2^(2^{4^[(4^{[trans^3^(3^chloro^4^cyanophenoxy)^2,2,4,4^ tetramethylcyclobutyl]carbamoyl}phenyl)amino]butoxy}acetamido)butanoyl]^4^ hydroxy^N^{[4^(4^methyl^1,3^thiazol^5^yl)phenyl]methyl}pyrrolidine^2^carboxamide 1H NMR (300 MHz, CD3OD):^8.88 (s, 1 H), 7.750-7.65 (m, 3 H), 7.50-7.33 (m, 4 H), 7.10-7.05 (m, 1H), 6.99-6.90 (m, 1 H), 6.54-6.43 (m, 2 H), 4.67 (s, 1 H), 4.60-4.50 (m, 3 H), 4.48-4.45 (m, 1 H), 4.21 (s, 1 H), 4.13-4.05 (m, 1 H), 3.98-3.90 (m, 2 H), 3.88- 3.70 (m, 2 H), 3.66-3.48 (m, 2 H), 3.20-3.03 (m, 2 H), 2.40 (s, 3 H), 2.25-2.12 (m, 1 H), 2.09– 1.99 (m, 1 H), 1.80-1.68 (m, 4 H), 1.30-1.10 (m, 12 H), 1.01 (s, 9 H); LC-MS 175 (ES+): m/z, 940.15 [MH+]
(2S,4R)^1^[(2S)^2^[2^({5^[(2^fluoro^4^{[trans^3^(3^chloro^4^cyanophenoxy)^2,2,4,4^ tetramethylcyclobutyl]carbamoyl}phenyl)amino]pentyl}oxy)acetamido]^3,3^ dimethylbutanoyl]^4^hydroxy^N^{[4^(4^methyl^1,3^thiazol^5^
yl)phenyl]methyl}pyrrolidine^2^carboxamide
1H NMR (300 MHz, CD3OD):^8.86 (s, 1 H), 7.80-7.70 (m, 1 H), 7.60-7.55 (m, 1 H), 7.50-7.37 (m, 4H), 7.14 (s, 1 H), 7.00-6.93 (m, 1 H), 6.80-6.65 (m, 1 H), 4.70 (s, 1 H), 4.65-4.50 (m, 3 H), 4.40-4.30 (m, 1 H), 4.29-4.25 (m, 1 H), 4.20-4.15 (m, 1 H), 4.04- 3.90 (m, 2 H), 3.89-3.85 (m, 1 H), 3.80-3.73 (m, 1 H), 3.70-3.65 (m, 1 H), 3.60-3.52 (m, 2H), 3.30-3.15 (m, 2 H), 2.40 (s, 3 H), 2.25-1.95 (m, 1 H), 2.02– 1.90 (m, 1 H), 1.80-1.68 (m, 4 H), 1.65-1.50 (m, 2 H), 1.30-1.23 (m, 6 H), 1.22-1.15 (m, 6 H), 1.01 (s, 176 9 H); LC-MS (ES+): m/z, 972.10 [MH+]
^^ (2S,4R)^1^[(2S)^3,3^dimethyl^2^(2^{4^[(4^{[trans^3^(3^chloro^4^cyanophenoxy)^2,2,4,4^ ^
^
^ tetramethylcyclobutyl]carbamoyl}phenyl)amino]butoxy}acetamido)butanoyl]^4^ ^
^
^ ^^ hydroxy^N^[(1S)^1^[4^(4^methyl^1,3^thiazol^5^yl)phenyl]ethyl]pyrrolidine^2^ ^ ^
^ carboxamide
1
^^ H NMR (400 MHz, CD3OD) ^ 8.86 (s, 1H), 7.72-7.64 (m, 3H), 7.44 (s, 4H), 7.12 (s, ^ 1H), 6.98 (d, J = 2.4 Hz, 1H), 6.64 (d, J = 8.8 Hz, 2H), 5.00 (d, J = 6.8 Hz, 1H), 4.69 (s, ^^ 1H), 4.62-4.58 (m, 1H), 4.44 (s, 1H), 4.28 (s, 1H), 4.12 (s, 1H), 4.00-3.93 (m, 2H), ^
3.87-3.75 (m, 2H), 3.65-3.59 (m, 2H),3.21 (s, 2H), 2.47 (s, 3H), 2.27-2.15 (m, 1H), 1.95 ^^ (m, 1H), 1.76 (s, 4H), 1.58-1.49 (m, 3H), 1.26 (d, J *^,^+^-'#^^^-"#^^^.^^^#^,-")^Mass ^
177 (ES+): m/z 955.20 [MH+]
Figure imgf000231_0001
. -. , , . , , . , . , , . , , . , , 178 .^,^^^^#^,-")^Mass (ES+): m/z 954.15 [MH+].
[0654] Synthesis of example 172:
Figure imgf000231_0002
.
[0655] Step 7: Synthesis of example 172:
[0656] TBTU (21.5 mg, 0.067 mmol) was added to a solution of 4-{[5-({[(2S)-1-[(2S,4R)-4-hydroxy-2- ({[4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)pentyl]amino}benzoic acid (31 mg, 0.044 mmol), 2-chloro-4-[trans-3-amino- 2,2,4,4-tetramethylcyclobutoxy]benzonitrile (12.4 mg, 0.044 mmol) in DMF (3.0 mL) and DIPEA (15.4 µL, 0.089 mmol) at room temperature. The resulting reaction mixture was stirred at room temperature for 1hr. LC-MS indicated formation of the desired product. The reaction mixture was diluted with EtOAc (30 mL), washed with water (15 mL x 2), brine (15 mL x 1), filtered through a Biotage universal phase separator and then concentrated under reduced pressure to give a crude residue, which was purified by silica gel chromatography on a Teledyne Combiflash ISCO system eluting with MeOH/DCM (v/v = 0:100 to 10:90) to yield the desired title product (yield: 41%).
[0657] Step 6: Synthesis of 4-{[5-({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)pentyl]amino}benzoic acid:
Figure imgf000232_0001
[0658] Lithium hydroxide (9.0 mg, 0.38 mmol) was added to a solution of methyl 4-{[5-({[(2S)-1- [(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3- dimethyl-1-oxobutan-2-yl]carbamoyl}methoxy)pentyl]amino}benzoate (96 mg, 0.14 mmol) in a mixed solvent of THF/water/methanol (v/v/v = 1/1/1, 2.00 mL) at room temperature. The resulting mixture was stirred at room temperature overnight. Aqueous HCl (1 N) was added to the reaction mixture to adjust pH to ~3. The resulting mixture was diluted with EtOAc (30 mL), washed with brine (15 mL x 2), dried over sodium sulfate, filtered through a Biotage Universal Phase Separator and then concentrated under reduced pressure to give a crude product, which was used for next step without any further purification. LC-MS (ES+): m/z 694.33[MH+].
[0659] Step 5: Synthesis of methyl 4-{[5-({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)pentyl]amino}benzoate.
Figure imgf000232_0002
.
[0660] TBTU (81.5 mg, 0.25 mmol) was added to a solution of 2-[(5-{[4- (methoxycarbonyl)phenyl]amino}pentyl)oxy]acetic acid (50.0 mg, 0.17 mmol), (2S,4R)-1-[(2S)-2-amino- 3,3-dimethylbutanoyl]-4-hydroxy-N-{[4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl}pyrrolidine-2- carboxamide (72.8 mg, 0.17 mmol) in DMF (3.0 mL) and DIPEA (59 µL, 0.34mmol) at room temperature. The resulting mixture was stirred at room temperature for 1h. The reaction mixture was diluted with EtOAc (30 mL), washed with water (15 mL x 2), brine (15 mL x 1), dried over sodium sulfate, filtered through a Biotage universal phase separator and then concentrated under reduced pressure to give a crude residue, which was purified by silica gel chromatography on a Teledyne Combiflash ISCO system eluting with MeOH/DCM (v/v = 0:100 to 10:90) to yield the titled product (yield: 51%, 2 steps). LC-MS (ES+): m/z 708.35 [MH+].
[0661] Step 4: Synthesis of 2-[(5-{[4-(methoxycarbonyl)phenyl]amino}pentyl)oxy]acetic acid:
Figure imgf000233_0001
.
[0662] Trifluoroacetic acid (2.63 mL, 34.5 mmol) was added to a solution of methyl 4-{[5-(2-methoxy- 2-oxoethoxy)pentyl]amino}benzoate (270 mg, 0.7682 mmol) in DCM (3.00 ml) at room temperature. The resulting mixture was stirred at 45 oC for 2h. The reaction mixture was then concentrated under reduced pressure to give a crude product, which was used for next step without any further purification. LC-MS (ES+): m/z 296.15 [MH+].
[0663] Step 3: Synthesis of methyl 4-({5-[2-(tert-butoxy)-2-oxoethoxy]pentyl}amino)benzoate:
Figure imgf000233_0002
.
[0664] To a solution of tert-butyl 2-[(5-oxopentyl)oxy]acetate (269 mg, 1.24 mmol) and methyl 4- aminobenzoate (187 mg, 1.24 mmol) in dichloroethane (5.00 mL) was added acetic acid (199 µL, 2.48 mmol) and sodium triacetoxyborohydride (394 mg, 1.86 mmol) at room temperature. The reaction mixture was stirred at room temperature for 18h. NaOH (1N solution in water) was then added to neutralize the acetic acid, the resulting reaction mixture was extracted with DCM (100 mL x 3). The organic layers were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a crude residue, which was purified by silica gel chromatography on a Teledyne Combiflash ISCO system eluting with MeOH/DCM (v/v = 0:100 to 15:85) to yield the desired title product (yield: 62 %).LC-MS (ES+): m/z 352.21 [MH+]
[0665] Step 2: Synthesis of tert-butyl 2-[(5-oxopentyl)oxy]acetate:
Figure imgf000234_0001
.
[0666] To a solution of tert-butyl 2-(hex-5-en-1-yloxy)acetate (300.0 mg, 1.40 mmol) in acetone (15.00 ml) were added potassium osmate(VI) dihydrate (15.5 mg, 0.042 mmol), follow by NMO (491.9 mg, 4.20 mmol) in water (4.5 ml) at room temperature. The resulting reaction mixture was stirred for 18h at room temperature. The reaction was monitored by TLC (EtOAc/Heptane, v/v = 25/75). Sodium periodate (898.2 mg, 4.20 mmol) was then added to the reaction mixture, the reaction was stirred at room temperature for another 3h. The reaction mixture was diluted with water (10 mL) and DCM (100 mL). The organic layer was separated and the aqueous layer was extracted with DCM (100 mL x 3). The combined organic layers were washed with brine (10 mL x 2) and then passed through a Universal Biotage Phase Separator and concentrated under reduced pressure to give a crude residue, which was purified by silica gel
chromatography on a Teledyne Combiflash ISCO system eluting with EtOAc/Heptane (v/v = 0:100 to 50:50) to yield the titled product (yield 90%).1H NMR (400 MHz, CDCl3) δ 9.75 (t, J = 1.8 Hz, 1H), 3.89- 3.93 (m, 2H), 3.51 (t, J = 6.1 Hz, 2H), 2.47 (dt, J = 1.6, 7.2 Hz, 2H), 1.69-1.78 (m, 2H), 1.64 (d, J = 8.2 Hz, 2H), 1.46 (s, 9H).
[0667] Step 1: Synthesis of tert-butyl 2-(hex-5-en-1-yloxy)acetate:
Figure imgf000234_0002
[0668] Tetrabutylammonium hydrogen sulfate (677.7 mg, 2.0 mmol) was added to a mixture of sodium hydroxide (23.9 g, 599 mmol) in water (20.0 mL) and toluene (20.00 ml) at 20 oC. To this mixture was added hex-5-en-1-ol (2.00 g, 20.0 mmol), the resulting mixture was stirred at 20 oC for 1h. The reaction was then cooled to 5 oC and tert-butyl 2-bromoacetate (20.0 mmol, 3.89 g) was added slowly while maintaining the internal temperature below 15 oC. The reaction mixture was then stirred at room temperature for additional 16h. The mixture was diluted with heptane (30 mL) and washed with water (20 mL). The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a crude residue, which was purified by silica gel chromatography on a Teledyne Combiflash ISCO system (gradient eluent: EtOAc/Heptane, v/v = 0/100 to 25/75) to afford the desired product (33%).1H NMR (400 MHz, CDCl3) δ 5.75-5.87 (m, 1H), 4.82-5.10 (m, 2H), 3.95 (s, 2H), 3.52 (t, J = 6.7 Hz, 2H), 2.08 (d, J = 7.0 Hz, 2H), 1.57-1.69 (m, 2H), 1.45-1.53 (m, 11H). LC-MS (ES+): m/z 237.14 [MNa+] [0669] Examples 173-178 were synthesized according to similar procedure described for synthesis of example 172, by using corresponding starting materials and intermediates.
[0670] Alternatively, steps 5-7 of example 174 is synthesized as following:
[0671] Step 7: synthesis of (2S,4R)-1-((S)-2-(2-((5-((4-((trans-3-(3-chloro-4-cyanophenoxy)- 2,2,4,4-tetramethylcyclobutyl)carbamoyl)phenyl)amino)pentyl)oxy)acetamido)-3,3- dimethylbutanoyl)-4-hydroxy-N-((S)-1-(4-(4-methylthiazol-5-yl)phenyl)ethyl)pyrrolidine-2- carboxamide:
Figure imgf000235_0001
[0672] A solution of 4-((5-(2-(((S)-1-((2S,4R)-4-hydroxy-2-(((S)-1-(4-(4-methylthiazol-5- yl)phenyl) ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)amino)-2- oxoethoxy)pentyl) amino)benzoic acid (1.17 g, 1.65 mmol) in methylene chloride (10 mL) was charged with HATU (688 mg, 1.81 mmol) and diisopropylethylamine (859 µL, 4.94 mmol). The reaction mixture was allowed to stir at room temperature for 10 minutes, then 4-(trans-3-amino- 2,2,4,4-tetramethylcyclobutoxy)-2-chlorobenzonitrile hydrochloride (545 mg, 1.73 mmol) was added. The reaction mixture was stirred at room temperature for 30 minutes. The reaction mixture was diluted with DCM (30 mL), then washed with water (10 mL), brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The crude material was purified by silica gel chromatography on a Teledyne Combiflash ISCO eluting with DCM/MeOH (100:0 to 90:10 to yield the desired product as a white solid (0.86 g, 54%). LC-MS (ES+): m/z 968.42 [MH+].
[0673] Step 6: synthesis of 4-((5-(2-(((S)-1-((2S,4R)-4-hydroxy-2-(((S)-1-(4-(4-methylthiazol-5- yl)phenyl) ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)amino)-2- oxoethoxy)pentyl) amino)benzoic acid
Figure imgf000236_0001
[0674] A solution of methyl 4-((5-(2-(((S)-1-((2S,4R)-4-hydroxy-2-(((S)-1-(4-(4-methylthiazol- 5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)amino)-2- oxoethoxy)pentyl)amino)benzoate (1.2 g, 1.66 mmol) in methanol (5 mL) was charged with 3 M NaOH (2.0 mL, 50.0 mmol). The reaction mixture was allowed to stir at room temperature for 72 hours. The reaction mixture was quenched with 1.0 M HCl and then concentrated under reduced pressure to remove the methanol. The aqueous was extracted with EtOAc (25 mL x 3). The combined organic layers were washed with brine (15 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The crude material was purified by silica gel
chromatography on a Teledyne Combiflash ISCO eluting with DCM/MeOH (100:0 to 90:10) to yield the desired product as a white solid (1.17 g, 100 %). LC-MS (ES+): m/z 708.32 [MH+].
[0675] Step 5: Synthesis of methyl 4-((5-(2-(((S)-1-((2S,4R)-4-hydroxy-2-(((S)-1-(4-(4- methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2- yl)amino)-2-oxoethoxy)pentyl)amino)benzoate.
Figure imgf000236_0002
[0676] A solution of 2-((5-((4-(methoxycarbonyl)phenyl)amino)pentyl)oxy)acetic acid (1.68 g, 5.68 mmol) and (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-((S)-1-(4-(4- methylthiazol-5-yl)phenyl)ethyl)pyrrolidine-2-carboxamide hydrochloride (2.73 g, 5.68 mmol) in methylene chloride (15 mL) was charged with O-(benzotriazol-1-yl)-N,N,N^,N^- tetramethyluronium tetrafluoroborate (1.82 g, 5.68 mmol) and diisopropylethylamine (2.95 mL, 17.0 mmol). The reaction mixture was allowed to stir at room temperature for 30 minutes. The reaction mixture was quenched with water (15 mL) and then extracted with DCM (15 mL). The organic layer was washed with brine (15 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The crude material was purified by silica gel chromatography on a Teledyne Combiflash ISCO eluting with DCM/MeOH (100:0 to 90:10) to yield the desired product as a white solid (1.2 g, 29%). LC-MS (ES+): m/z 722.34 [MH+].
[0677] Table 11. Exemplary Compounds.
Figure imgf000237_0001
Figure imgf000238_0001
[0678] Examples 179-181 were synthesized according to similar procedure described for synthesis of example 182, by using corresponding starting materials and intermediates.
[0679] Synthesis of example 182:
Figure imgf000239_0001
Figure imgf000239_0002
Figure imgf000239_0003
.
[0680] Step 8: Synthesis of example 182
[0681] To a stirred solution of 4-{4-[2-({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)ethoxy]phenyl}benzoic acid (89.0 mg, 122 µmol) in methylene chloride (2.0 mL) was added HATU (55.5 mg, 146 µmol) and diisopropylethylamine (63.7 µL, 366 µmol). The reaction mixture was stirred at room temperature for 10 minutes. The reaction mixture was then charged with 2- chloro-4-[trans-3-amino-2,2,4,4-tetramethylcyclobutoxy]benzonitrile (34.0 mg, 122 µmol). The reaction was stirred at room temperature for 30 minutes. The reaction was monitored by LC-MS, which indicated completion of reaction. The reaction mixture was quenched with water (5 mL) and then extracted with DCM (25 mL). The organic layer was washed with brine (5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO (eluent: DCM/MeOH (v:v = 90:10)) to give titled product (yield: 37%) as a white solid.
[0682] Step 7: Synthesis of 4-{4-[2-({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)ethoxy]phenyl}benzoic acid:
Figure imgf000240_0001
[0683] To a stirred solution of ethyl 4-{4-[2-({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol- 5-yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)ethoxy]phenyl}benzoate (188.4 mg, 248 µmol) in methanol (2.0 mL) was added 1 M NaOH in water (0.5 mL, 12.5 mmol). The reaction mixture was stirred at room temperature for 16 hours. The reaction was monitored by LC-MS, which indicated completion of reaction. The reaction mixture was quenched with 1.0 M HCl in water and then concentrated under reduced pressure to remove the methanol. The aqueous was extracted with EtOAc (25 mL). The organic layer was washed with brine (5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO (eluent:
DCM/MeOH (v:v = 90:10)) to give titled product (yield: 50%) as a white solid. LC-MS (ES+): m/z 729.18 [MH+]
[0684] Step 6: Synthesis of ethyl 4-{4-[2-({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)ethoxy]phenyl}benzoate:
Figure imgf000240_0002
.
[0685] To a stirred solution of 2-(2-{4-[4-(ethoxycarbonyl)phenyl]phenoxy}ethoxy)acetic acid (100 mg, 290.3 µmol) and (2S,4R)-1-[(2S)-2-amino-3,3-dimethylbutanoyl]-4-hydroxy-N-[(1S)-1-[4-(4-methyl- 1,3-thiazol-5-yl)phenyl]ethyl]pyrrolidine-2-carboxamide hydrochloride (135.5 mg, 290.3
µmol) in Dichloromethane (2.0 mL) was added O-(benzotriazol-1-yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate (93.20 mg, 290.3 µmol) and diisopropylethylamine (151.6 µL,870.9 µmol). The reaction mixture was stirred at room temperature for 30 minutes. The reaction was monitored by LC-MS, which indicated completion of reaction. The reaction mixture was concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO (gradient eluent: Heptane/Acetone (v:v = 100:0 to 0:100)) to give titled product (yield: 86%) as a white solid. LC-MS (ES+): m/z 757.3283 [MH+].
[0686] Step 5: Synthesis of 2-(2-{4-[4-(ethoxycarbonyl)phenyl]phenoxy}ethoxy)acetic acid:
Figure imgf000241_0001
.
[0687] To a stirred solution of ethyl 4-(4-{2-[2-(tert-butoxy)-2-oxoethoxy]ethoxy}phenyl)benzoate (245 mg, 611 µmol) in methylene chloride (1.0 mL) was added trifluoroacetic acid (1.0 mL, 12.9 mmol). The reaction mixture was stirred at room temperature for 30 minutes. The reaction was monitored by LC-MS, which indicated completion of reaction. The reaction mixture was concentrated under reduced pressure to give titled product (yield: 100% based on crude) as an off white solid. The material was used in next step without any further purification. LC-MS (ES+): m/z 345.1330 [MH+].
[0688] Step 4: Synthesis of ethyl 4-(4-{2-[2-(tert-butoxy)-2-oxoethoxy]ethoxy}phenyl)benzoate:
Figure imgf000241_0002
[0689] To a stirred mixture of ethyl 4'-hydroxy-[1,1'-biphenyl]-4-carboxylate (146.6 mg, 605.3 µmol) and tert-butyl 2-{2-[(4-methylbenzenesulfonyl)oxy]ethoxy}acetate (200.0 mg, 605.3 µmol) in acetonitrile (2.0 mL) was added potassium carbonate (125.4 mg, 907.9 µmol) at room temperature. The reaction mixture was then stirred at 80 ºC for 16 hours. The reaction was monitored by LC-MS, which indicated completion of reaction. The reaction mixture was concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO (gradient eluent: Heptane/EtOAc (v:v = 100:0 to 50:50)) to give titled product (yield: 99%) as a clear oil. LC-MS (ES+): m/z 423.18 [MNa+].
[0690] Step 3: synthesis of tert-butyl 2-{2-[(4-methylbenzenesulfonyl)oxy]ethoxy}acetate:
Figure imgf000241_0003
[0691] To a stirred solution of tert-butyl 2-(2-hydroxyethoxy)acetate (1.44 g, 0.19 mmol) in methylene chloride (10.0 mL) was added 4-methylbenzene-1-sulfonyl chloride (1.713 g, 0.21 mmol) and triethylamine (1.707 mL, 12.25 mmol) at room temperature. The reaction mixture was stirred at room temperature for 16 hours. The reaction was monitored by LC-MS, which indicated completion of reaction. The reaction mixture was concentrated under reduced pressure give a crude material, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO (gradient eluent:
Heptane/Acetone (v:v = 100:0 to 0:100)) to give titled product (yield: 69%) as a clear oil.1H NMR (400 MHz, CD3OD) ^ ppm 7.77 - 7.83 (m, 2 H), 7.44 (d, J = 7.83 Hz, 2 H), 4.14 - 4.19 (m, 2 H), 3.93 (s, 2 H), 3.68 - 3.74 (m, 2 H), 2.46 (s, 3 H), 1.46 (s, 9 H); LC-MS (ES+): m/z 353.1053 [MNa+], tR = 2.56 min.
[0692] Step 2: Synthesis of tert-butyl 2-(2-hydroxyethoxy)acetate:
Figure imgf000242_0001
.
[0693] To a stirred solution of tert-butyl 2-[2-(benzyloxy)ethoxy]acetate in Ethanol (10.0 mL) was added palladium on carbon (10% wt.) (1.99 g, 1.87 mmol). The reaction mixture was evacuated and purged with H2 gas (3 x). The reaction mixture was stirred at room temperature under an atmosphere of H2 for 16 hours. The reaction was monitored by TLC analysis, which indicated completion of reaction. The reaction mixture was filtered through a pad of celite and the filtrate was concentrated under reduced pressure to give titled product (yield: 87% based on crude) as a clear oil. The crude material was used in next step reaction without any further purification.
[0694] Step 1: Synthesis of tert-butyl 2-[2-(benzyloxy)ethoxy]acetate:
Figure imgf000242_0002
[0695] To a stirred solution of 2-(benzyloxy)ethanol (5.0 g, 32.8 mmol) and tert-butyl 2- bromoacetate (7.02 g, 36.0 mmol) in acetonitrile (10.0 mL) was added potassium carbonate (6.78 g, 49.1 mmol) at room temperature. The reaction mixture then stirred at 80 ºC for 16 hours. The reaction was monitored by TLC analysis, which indicated completion of reaction. The reaction mixture was diluted with water (10.0 mL) and extracted with EtOAc (20.0 mL). The organic layer was washed with water (5.0 mL), brine (5.0 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give titled product (yield: 100% based on crude) as a yellow oil. This crude material was used in next step reaction without any further purification.
[0696] Table 12. Exemplary Compounds. Ex# Structure Compound name and Analytical data
Figure imgf000243_0001
Figure imgf000244_0001
[0697] Examples 184-187 were synthesized according to similar procedure described for synthesis of example 183, by using corresponding starting materials and intermediates.
[0698] Synthesis of Example 183:
Figure imgf000244_0002
.
[0699] Step 7: Synthesis of Example 183 [0700] To a stirred solution of 2-chloro-4-[trans-3-amino-2,2,4,4- tetramethylcyclobutoxy]benzonitrile (25.3 mg, 90.9 µmol) and 4-{4-[({[(2S)-1-[(2S,4R)-4-hydroxy-2- ({[4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)methyl]phenoxy}benzoic acid (65 mg, 90.9 µmol) in methylene chloride (2.0 mL) was added O-(benzotriazol-1-yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate (29.1 mg, 90.9 µmol) and diisopropylethylamine (47.3 µL, 272 µmol) at room temperature. The reaction mixture was stirred at room temperature for 30 minutes. The reaction was monitored by LC-MS, which indicated completion of reaction. The reaction mixture was diluted with water (5 mL) and extracted with DCM (25 mL). The organic layer was separated and washed with brine (5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO (eluent: DCM/MeOH (v:v = 90:10)) to give titled product (yield: 22%) as a white solid.
[0701] Step 6: Synthesis of 4-{4-[({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)methyl]phenoxy}benzoic acid:
Figure imgf000245_0001
.
[0702] To a stirred solution of methyl 4-{4-[({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol- 5-yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)methyl]phenoxy}benzoate (68 mg, 93.2 µmol) in methanol (2.0 mL) was added 1 M NaOH solution in water (0.5 mL, 12.5 mmol) at room temperature. The reaction mixture was allowed to stir at room temperature for 16 hours. The reaction was monitored by LC-MS, which indicated completion of reaction. The reaction mixture was quenched with 1.0 M HCl solution in water (0.5 mL) and then concentrated under reduced pressure to remove the methanol. The aqueous was extracted with EtOAc (25 mL). The organic layer was separated, washed with brine (5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give titled product (yield: 98% based on crude) as a white solid. This material was used in next step reaction without any further purification. LC-MS (ES+): m/z
715.28[MH+]. [0703] Step 5: Synthesis of methyl 4-{4-[({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methoxy)methyl]phenoxy}benzoate:
Figure imgf000246_0001
[0704] To a stirred solution of 2-({4-[4-(methoxycarbonyl)phenoxy]phenyl}methoxy)acetic acid (30.0 mg, 94.8 µmol) and (2S,4R)-1-[(2S)-2-amino-3,3-dimethylbutanoyl]-4-hydroxy-N-[(1S)-1-[4-(4-methyl- 1,3-thiazol-5-yl)phenyl]ethyl]pyrrolidine-2-carboxamide hydrochloride (44.2 mg, 94.8 µmol) in methylene chloride (2.0 mL) was added O-(benzotriazol-1-yl)-N,N,N4,N4-tetramethyluronium tetrafluoroborate (30.4 mg, 94.8 µmol) and diisopropylethylamine (49.4 µL, 284 µmol) at room temperature. The reaction mixture was allowed to stir at room temperature for 30 minutes. The reaction was monitored by LC-MS, which indicated completion of reaction. The reaction mixture was concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO (gradient eluent: Heptane/Acetone (v:v = 100:0 to 0:100)) to give titled product (yield: 99%) as a white solid. LC-MS (ES+): m/z 729.30 [MH+].
[0705] Step 4: 2-({4-[4-(methoxycarbonyl)phenoxy]phenyl}methoxy)acetic acid:
Figure imgf000246_0002
.
[0706] A solution of methyl 4-(4-{[2-(tert-butoxy)-2-oxoethoxy]methyl}phenoxy)benzoate (200.0 mg, 537 µmol) in hydrogen chloride solution (4 M in dioxane, 2.0 mL) was stirred at room temperature for 2 hours. The reaction was monitored by LC-MS, which indicated completion of reaction. The reaction mixture was concentrated under reduced pressure to give titled product (yield: 95% based on crude) as an off white solid. This material used in next step reaction without any further purification. LC-MS (ES+): m/z 339.0858 [MNa+].
[0707] Step 3: Synthesis of methyl 4-(4-{[2-(tert-butoxy)-2-oxoethoxy]methyl}phenoxy)benzoate:
Figure imgf000246_0003
. [0708] To a stirred mixture of sodium hydroxide (1.16 g, 29 mmol) in water (2.0 mL) and toluene (2.0 mL) at 20 ºC was charged with tetrabutylammonium hydrogen sulfate (32.86 mg, 96.79 µmol), followed by methyl 4-[4-(hydroxymethyl)phenoxy]benzoate (250.0 mg, 967.9 µmol), the resulting mixture was stirred at 20 ºC for 1h. The mixture was then cooled to 5 ºC, tert-butyl 2-bromoacetate (207.5 mg, 1.064 mmol) was added slowly and the internal temperature was maintained below 15 ºC. Upon the completion of this addition, the reaction mixture was allowed to warm up to room temperature and stirred for 16h at room temperature. The reaction was monitored by LC-MS, which indicated completion of reaction. The mixture was diluted with water (5 mL) and extracted with EtOAc (30 mL). The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to a give a crude material, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO (eluent (gradient): Heptane/EtOAc (v:v = 100:0 to 70:30)) to give titled product (yield: 56%) as a white solid. LC-MS (ES+): m/z 395.15 [MNa+].
[0709] Step 2: Synthesis of methyl 4-[4-(hydroxymethyl)phenoxy]benzoate:
Figure imgf000247_0001
.
[0710] To a stirred solution of methyl 4-(4-formylphenoxy)benzoate (750.0 mg, 2.92
mmol) in methanol (2.0 mL) was added sodium borohydride (121 mg, 3.21 mmol) at room temperature. The reaction mixture was allowed to stir at room temperature for 30 min. The reaction was monitored by LC-MS, which indicated completion of reaction. The reaction mixture was slowly quenched with 1N HCl (solution in water), concentrated under reduced pressure to remove the bulk of methanol, and then extracted with DCM (30 mL). The organic layer was separated, washed with brine (5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO (eluent (gradient): Heptane/EtOAc (v:v = 100:0 to 50:50)) to give titled product (yield: 94%) as a white solid. LC-MS (ES+): m/z 259.10 [MH+].
[0711] Step 1: Synthesis of methyl 4-(4-formylphenoxy)benzoate:
Figure imgf000247_0002
.
[0712] To a stirred mixture of methyl 4-hydroxybenzoate (1.0 g, 6.57 mmol) and potassium
carbonate (1.36 g, 9.85 mmol) in dimethylformamide (2.0 mL) was added 4-fluorobenzaldehyde (815 mg, 6.57 mmol) at room temperature. The reaction mixture was then stirred at 80 ºC for 16h. The reaction was monitored by LC-MS, which indicated completion of reaction. The reaction mixture was cooled to room temperature, diluted with water (10 mL) and extracted with EtOAc (50 mL x 2). The organic layer was separated, washed with brine (10 mL x 2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO (eluent (gradient): Heptane/EtOAc (v:v = 100:0 to 50:50)) to give titled product (yield: 90%) as a white solid. LC-MS (ES+): m/z 257.08 [MH+].
[0713] Table 13. Exemplary Compounds.
Figure imgf000248_0001
[0714] Example 189 was synthesized according to similar procedure described for synthesis of example 188, by using corresponding starting materials and intermediates.
[0715] Synthesis of example 188:
Figure imgf000249_0001
.
[0716] Step 7: Synthesis of example 188
[0717] Trifluoroacetic acid (1.12 mL, 14.7 mmol) was added to a stirred solution of tert-butyl N-({[(2S)- 1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-oxazol-5-yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3- dimethyl-1-oxobutan-2-yl]carbamoyl}methyl)-N-[5-(4-{[trans-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamoyl}phenoxy)pentyl]carbamate (34 mg, 0.0327 mmol) in DCM (3.00 ml) at room temperature. The resulting mixture was stirred at 45 oC for 48h. The reaction mixture was then concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO system, eluting with MeOH/DCM (gradient: v:v = 0:100 to 10:90) to yield the desired title product (yield: 62 %).
[0718] Step 6: Synthesis of tert-butyl N-({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2-yl]carbamoyl}methyl)-N-[5-(4- {[trans-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamoyl}phenoxy)pentyl]carbamate:
Figure imgf000249_0002
.
[0719] TBTU (23.0 mg, 0.072 mmol) was added to a stirred solution of 4-[(5-{[(tert- butoxy)carbonyl]({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methyl)amino}pentyl)oxy]benzoic acid (38 mg, 0.04786 mmol) and 2-chloro-4-[trans-3- amino-2,2,4,4-tetramethylcyclobutoxy]benzonitrile (13.3 mg, 0.04786 mmol) in DMF (3.0 mL) and DIPEA (16.5 µL, 0.095 mmol) at room temperature. The resulting mixture was stirred at room temperature for 1h. The reaction was then diluted with EtOAc (30 mL), washed with brine (5 mL x 2), filtered through a Biotage Universal Phase Separator and then concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO system, eluting with MeOH/DCM (gradient: v:v = 0:100 to 10:90) to yield the desired title product (yield: 60 %).
[0720] Step 5: Synthesis of 4-[(5-{[(tert-butoxy)carbonyl]({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4- methyl-1,3-thiazol-5-yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methyl)amino}pentyl)oxy]benzoic acid:
Figure imgf000250_0001
[0721] Lithium hydroxide (3.0 mg, 0.128 mmol) was added to a stirred solution of methyl 4-[(5-{[(tert- butoxy)carbonyl]({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4-(4-methyl-1,3-thiazol-5- yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methyl)amino}pentyl)oxy]benzoate (37 mg, 0.046 mmol) in a mixed solvent of THF/water (v:v = 1:1, 2.00 mL) at room temperature. The resulting reaction mixture was stirred at room temperature overnight. To the reaction mixture was added 1N HCl (aqueous solution) to adjust pH = ~3. The resulting mixture was extracted with EtOAc (20 mL x 2), washed with brine (5 mL x 2), filtered through a Biotage Universal Phase Separator and then concentrated under reduced pressure to give a crude material (yield: 100 % based on crude). This crude product was used for the next step reaction without any further purification.
[0722] Step 4: Synthesis of methyl 4-[(5-{[(tert-butoxy)carbonyl]({[(2S)-1-[(2S,4R)-4-hydroxy-2-({[4- (4-methyl-1,3-thiazol-5-yl)phenyl]methyl}carbamoyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl}methyl)amino}pentyl)oxy]benzoate:
Figure imgf000251_0001
[0723] TBTU (36.6 mg, 0.1142 mmol) was added to a stirred solution of 2-{[(tert-butoxy)carbonyl]({5- [4-(methoxycarbonyl)phenoxy]pentyl})amino}acetic acid (37 mg, 0.076 mmol) and (2S,4R)-1-[(2S)-2- amino-3,3-dimethylbutanoyl]-4-hydroxy-N-{[4-(4-methyl-1,3-thiazol-5-yl)phenyl]methyl}pyrrolidine-2- carboxamide (32.8 mg, 0.076 mmol) in DMF (3.0 mL) and DIPEA (26.4 µL, 0.15 mmol) at room temperature. The resulting reaction mixture was stirred at room temperature for 1hr. The reaction was then diluted with EtOAc (30 mL), washed with brine (10 mL), filtered through a Biotage Universal Phase Separator and then concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO system, eluting with MeOH/DCM (gradient: v/v = 0/100 to 10/90) to yield the desired title product (yield: 64%).
[0724] Step 3: Synthesis of 2-{[(tert-butoxy)carbonyl]({5-[4- (methoxycarbonyl)phenoxy]pentyl})amino}acetic acid:
Figure imgf000251_0002
.
[0725] Palladium on carbon (96.8 mg, 0.91 mmol) was added to a stirred solution of methyl 4-[(5-{[2- (benzyloxy)-2-oxoethyl][(tert-butoxy)carbonyl]amino}pentyl)oxy]benzoate (83.0 mg, 0.171 mmol) in ethanol (20 ml) at room temperature. The reaction mixture was degassed and charged with H(g) and then stirred at room temperature for 16h under a hydrogen atmosphere. Solids were then removed by filtration and the solvent was concentrated under reduced pressure to give a crude material (yield: 98% based on crude). This crude product was used for the next step reaction without any further purification.
[0726] Step 2: Synthesis of methyl 4-[(5-{[2-(benzyloxy)-2-oxoethyl][(tert- butoxy)carbonyl]amino}pentyl)oxy]benzoate:
Figure imgf000252_0001
[0727] Di-tert-butyl dicarbonate (47.7 µL, 0.21 mmol) was added to a stirred solution of methyl 4-[(5- {[2-(benzyloxy)-2-oxoethyl]amino}pentyl)oxy]benzoate (73.0 mg, 0.19 mmol) in THF (5.0 ml) at room temperature. The reaction mixture was heated to reflux at 80 oC and stirred at 80 oC for 14 hours. The reaction was then cooled to room temperature, diluted with ethyl acetate (20 mL), washed with saturated aq. NaHCO3 (10 mL). The organic layer was separated and filtered using a Biotage Universal Phase Separator and then concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO system, eluting with EtOAc/Heptane (gradient v:v = 0:100 to 40:60) to yield the desired title product (yield: 95%).
[0728] Step 1: Synthesis of methyl 4-[(5-{[2-(benzyloxy)-2-oxoethyl]amino}pentyl)oxy]benzoate:
Figure imgf000252_0002
[0729] To a stirred mixture of methyl 4-[(5-oxopentyl)oxy]benzoate (269 mg, 1.13 mmol) and benzyl 2- aminoacetate hydrochloride (186 mg, 1.13 mmol) in DCE (5.00 mL) was added acetic acid (181 µL, 2.26 mmol) and sodium triacetoxyborohydride (358 mg, 1.69 mmol) at room temperature. The reaction mixture was stirred at room temperature for 18h. To the reaction mixture was added 1N NaOH aqueous solution to adjust pH = ~10, the resulting mixture was then extracted with DCM (30 mL x 3). The organic layer was separated, washed with brine (10 mL x 2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography on a Teledyne Combiflash ISCO, eluting with MeOH/DCM (gradient v:v = 0:100 to 15:85) to yield the titled product (17 %).
[0730] Table 14. Exemplary Compounds. Ex# Structure Compound name and Analytical data
Figure imgf000253_0001
Figure imgf000254_0001
Figure imgf000255_0001
[0731] Examples 191-199 was synthesized according to similar procedure described for synthesis of example 190, by using corresponding starting materials and intermediates.
[0732] Synthesis of example 190:
Figure imgf000255_0002
. [0733] Step 1: Synthesis of 5-{3-[4-(4-hydroxyphenyl)phenyl]-5-imino-4,4-dimethyl-2- sulfanylideneimidazolidin-1-yl}-3-(trifluoromethyl)pyridine-2-carbonitrile:
Figure imgf000256_0001
.
[0734] To a stirred solution of 5-isothiocyanato-3-(trifluoromethyl)pyridine-2-carbonitrile (440.0 mg, 1.92 mmol) in N,N-dimethylpyridin-4-amine (322.0 mg, 2.64 mmol) and toluene (10.0 mL) was added 2-[[4-(4-hydroxyphenyl)phenyl]amino]-2-methylpropanenitrile (400.0 mg, 1.59 mmol) under a nitrogen atmosphere at room temperature. The resulting solution was stirred at 100 oC for 12h. The reaction mixture was then concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether, v/v = 1/1) to give the titiled product (yield: 17%). Mass (ES+): m/z 482.20[MH+] .
[0735] Step 2: Synthesis of 5-{3-[4-(4-hydroxyphenyl)phenyl]-4,4-dimethyl-5-oxo-2- sulfanylideneimidazolidin-1-yl}-3-(trifluoromethyl)pyridine-2-carbonitrile:
Figure imgf000256_0002
.
[0736] To a stirred solution of 5-{3-[4-(4-hydroxyphenyl)phenyl]-5-imino-4,4-dimethyl-2- sulfanylideneimidazolidin-1-yl}-3-(trifluoromethyl)pyridine-2-carbonitrile (160.0 mg, 0.33 mmol) in methanol (5.0 mL) was added hydrogen chloride aqueous solution (2N, 2.0 mL) at room temperature. The resulting solution was then refluxed for 2 hours. The reaction was cooled to room temperature, concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether, v/v = 1/1) to give the titled product (yield: 69%) as a yellow solid. LC-MS (ES+): m/z 481.15[MH+].
[0737] Step 3. Synthesis of tert-butyl 2-{4-[4-(4-{3-[6-cyano-5-(trifluoromethyl)pyridin-3-yl]- 5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)phenoxy]butoxy}acetate:
Figure imgf000257_0001
.
[0738] To a stirred solution of 5-{3-[4-(4-hydroxyphenyl)phenyl]-4,4-dimethyl-5-oxo-2- sulfanylideneimidazolidin-1-yl}-3-(trifluoromethyl)pyridine-2-carbonitrile (110.0 mg, 0.23 mmol) and tert-butyl 2-{4-[(4-methylbenzenesulfonyl)oxy]butoxy}acetate (163.0 mg, 0.45 mmol) in N,N-dimethylformamide (3.0 mL) was added potassium carbonate (62.9 mg, 0.46 mmol) at room temperature. The resulting mixture was stirred at 60oC for 3 hours. The reaction was then cooled to room temperature, diluted with water (10 mL) and extracted with ethyl acetate (30 mL x 3). The organic layers were combined, dried over anhydrous sodium sulfate, concentrated under reduced pressure to give a crude material, which was purified by flash silica gel chromatography (eluent: ethyl acetate/petroleum ether, v/v =1/1) to give the titled product (yield: 98%) as a yellow solid.
[0739] Step 4. Synthesis of 2-{4-[4-(4-{3-[6-cyano-5-(trifluoromethyl)pyridin-3-yl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)phenoxy]butoxy}acetic acid:
Figure imgf000257_0002
.
[0740] To a stirred solution of tert-butyl 2-{4-[4-(4-{3-[6-cyano-5-(trifluoromethyl)pyridin-3- yl]-5,5-dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)phenoxy]butoxy}acetate (150.0 mg, 0.22 mmol) in dichloromethane (2.0 mL) was added trifluoroacetic acid (2.0 mL) at room temperature. The resulting solution was stirred for 2h at room temperature. The resulting mixture was concentrated under reduced pressure to give a crude material, which was used for next step reaction without any further purifications. Mass (ES+): m/z 613.00 [MH+].
[0741] Step 5. Synthesis of example 190:
Figure imgf000257_0003
. [0742] To a stirred solution of 2-{4-[4-(4-{3-[6-cyano-5-(trifluoromethyl)pyridin-3-yl]-5,5- dimethyl-4-oxo-2-sulfanylideneimidazolidin-1-yl}phenyl)phenoxy]butoxy}acetic acid (80.0 mg, 0.13 mmol) and (2S,4R)-1-[(2S)-2-amino-3,3-dimethylbutanoyl]-4-hydroxy-N-{[4-(1,3-oxazol-5- yl)phenyl]methyl}pyrrolidine-2-carboxamide (53.8 mg, 0.13 mmol) in N,N-dimethylformamide (2.0 mL) was added O-(7-azabenzotriazol-1-yl)-N,N,N,N-tetramethyluronium
hexafluorophosphate (51.0 mg, 0.13 mmol) and N-ethyl-N-isopropylpropan-2-amine (43.0 mg, 0.33 mmol) at room temperature. The resulting solution was stirred for 2h at room temperature. LC-MS indicated formation of the desired product. The reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (50 mL x 3). The organic layers were combined, washed with brine (10 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a crude material, which was purified by a silica gel flash chromatography (eluent: ethyl acetate/petroleum ether, v/v = 1/1) to give the titled product as a white solid (yield: 45%).
[0743] Table 15. Exemplary Compounds.
Figure imgf000258_0001
Figure imgf000259_0001
[0744] Example 201was synthesized according chemistry shown below, utilizing similar procedures used for the synthesis of example 75.
Figure imgf000259_0002
.
[0745] Examples 200, 202-203 was synthesized according to similar procedure described for synthesis of example 201, by using corresponding starting materials and intermediates.
[0746] Table 16. Exemplary Compounds.
Figure imgf000260_0001
[0747] Table 17. Additional Exemplary Compounds.
Figure imgf000260_0002
Figure imgf000261_0001
Figure imgf000262_0001
Figure imgf000263_0001
Figure imgf000264_0001
Figure imgf000265_0001
Figure imgf000266_0001
Figure imgf000267_0001
Figure imgf000268_0001
Figure imgf000269_0001
Figure imgf000270_0001
Figure imgf000271_0001
Figure imgf000272_0001
Figure imgf000273_0001
Figure imgf000274_0001
Figure imgf000275_0001
Figure imgf000276_0001
Figure imgf000277_0001
Figure imgf000278_0001
Figure imgf000279_0001
Figure imgf000280_0001
Figure imgf000281_0001
Figure imgf000282_0001
Figure imgf000283_0001
Figure imgf000284_0001
Figure imgf000285_0001
Figure imgf000286_0001
Figure imgf000287_0001
Figure imgf000288_0001
Figure imgf000289_0001
Figure imgf000290_0001
Figure imgf000291_0001
Figure imgf000292_0001
Figure imgf000293_0001
Figure imgf000294_0001
Figure imgf000295_0001
Figure imgf000296_0001
Figure imgf000297_0001
Figure imgf000298_0001
Figure imgf000299_0001
Figure imgf000300_0001
Figure imgf000301_0001
Figure imgf000302_0001
Figure imgf000303_0001
Figure imgf000304_0001
Figure imgf000305_0001
Figure imgf000306_0001
Figure imgf000307_0001
Figure imgf000308_0001
Figure imgf000309_0001
Figure imgf000310_0001
Figure imgf000311_0001
Figure imgf000312_0001
Figure imgf000313_0001
Figure imgf000314_0001
Figure imgf000315_0001
Figure imgf000316_0001
Figure imgf000317_0001
Figure imgf000318_0001
Figure imgf000319_0001
Figure imgf000320_0001
Figure imgf000321_0001
Figure imgf000322_0001
Figure imgf000323_0001
Figure imgf000324_0001
Figure imgf000325_0001
Figure imgf000326_0001
Figure imgf000327_0001
Figure imgf000328_0001
Figure imgf000329_0001
Figure imgf000330_0001
Figure imgf000331_0001
Measured Mass Ion Ex# Structure Data
MH+ 1 MH+ 2 592
593
[0748] Synthesis of Example 608
Figure imgf000332_0001
[0749] Step 1: Synthesis of tert-butyl 4-(5-(methoxycarbonyl)pyridin-2-yl)piperazine-1- carboxylate:
Figure imgf000332_0002
[0750] A mixture of methyl 6-fluoronicotinate (2.0 g, 13.2 mmol), tert-butyl piperazine-1- carboxylate (2.4 g, 13.2 mmol) and N-ethyl-N-isopropylpropan-2-amine (3.3 g, 26.4 mmol) in anhydrous 1-methylpyrrolidin-2-one (10 ml) was stirred at 90°C for 12 hours. TLC showed the reaction was complete. The cooled reaction mixture was partitioned between water (10 ml) and ethyl acetate (50 ml). The organic layer was collected, washed with brine (50 ml x 2), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by column (eluted with 20% ethyl acetate in hexane) to afford tert-butyl 4-(5- (methoxycarbonyl)pyridin-2-yl)piperazine-1-carboxylate (4.0 g, yield 95%) as yellow solid. 1HNMR (400 MHz, CDCl3): ^ 1.48 (s, 9H), 3.53-3.56 (m, 4H), 3.67-3.69 (m, 4H), 3.87 (s, 3H), 6.58 (d, J = 8.8 Hz, 2H), 8.02-8.05 (m, 1H), 8.79-8.80 (m, 1H). Chemical Formula: C16H23N3O4, Molecular Weight: 321.37.
[0751] Step 2: Synthesis of methyl 6-(piperazin-1-yl)nicotinate
Figure imgf000333_0001
[0752] A mixture of tert-butyl 4-(5-(methoxycarbonyl)pyridin-2-yl)piperazine-1-carboxylate (4.0 g, 12.4 mol) and 2,2,2-trifluoroacetic acid (10 ml) in dichloromethane (10 ml) was stirred at room temperature for 2 hours. TLC showed the reaction was complete. The volatiles were evaporated under reduced pressure. The residue was taken up with dichloromethane (50 ml) and washed with aqueous sodium bicarbonate solution (1N, 15 ml), dried over sodium sulfate to give methyl 6-(piperazin-1-yl)nicotinate (3.8 g, crude) as yellow oil which was used in next step without further purification. 1HNMR (400 MHz, DMSO-d): ^ 3.13-3.16 (m, 4H), 3.80 (s, 3H), 3.82-3.85 (m, 4H), 6.96 (d, J = 9.2 Hz, 1H), 8.00-8.03 (m, 1H), 8.67-8.68 (m, 1H). Chemical Formula: C11H15N3O2, Molecular Weight: 221.26. [0753] Step 3: Synthesis of methyl 6-(4-(2-(2-(tert-butoxy)-2-oxoethoxy)ethyl)piperazin-1- yl)nicotinate.
Figure imgf000334_0001
[0754] A mixture of methyl 6-(piperazin-1-yl)nicotinate (500 mg, 2.3 mmol), tert-butyl 2-(2- (tosyloxy)ethoxy)acetate (745 mg, 2.3 mmol) and potassium carbonate (1.2 g, 9.0 mmol) in anhydrous N,N-dimethylformamide (10 ml) was stirred at 40°C for 12 hours. TLC showed the reaction was complete. The cooled reaction mixture was partitioned between water (20 ml) and ethyl acetate (20 ml). The organic layer was collected, washed with brine (100 ml x 2), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluted with 20% ethyl acetate in hexane) to afford methyl 6-(4-(2-(2-(tert-butoxy)-2-oxoethoxy)ethyl)piperazin-1-yl)nicotinate (400 mg, yield 46%) as yellow solid.
[0755] Step 4: Synthesis of 2-(2-(4-(5-(methoxycarbonyl)pyridin-2-yl)piperazin-1- yl)ethoxy)acetic acid.
Figure imgf000334_0002
[0756] A mixture of methyl 6-(4-(2-(2-(tert-butoxy)-2-oxoethoxy)ethyl)piperazin-1-yl)nicotinate (150 mg, 0.39 mmol) and 2,2,2-trifluoroacetic acid (1 ml) in dichloromethane (1 ml) was stirred at room temperature for 2 hours. TLC showed the reaction was complete. The volatiles were evaporated under reduced pressure to afford 2-(2-(4-(5-(methoxycarbonyl)pyridin-2-yl)piperazin- 1-yl)ethoxy)acetic acid (150 mg, crude) as yellow oil which was used in next step without further purification. LC_MS: (ES+): m/z 324.1 [M+H]+. tR = 1.306 min. Chemical Formula:
C15H21N3O5; Molecular Weight: 323.34.
[0757] Step 5: Synthesis of methyl 6-(4-(2-(2-(((S)-1-((2S,4R)-4-hydroxy-2-(((S)-1-(4-(4- methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2- yl)amino)-2-oxoethoxy)ethyl)piperazin-1-yl)nicotinate.
Figure imgf000335_0001
[0758] To a stirred solution of 2-(2-(4-(5-(methoxycarbonyl)pyridin-2-yl)piperazin-1- yl)ethoxy)acetic acid (150 mg, crude), (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy- N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide hydrochloride (230 mg, 0.48 mmol), and N-ethyl-N-isopropylpropan-2-amine (260 mg, 1.92mmol) in anhydrous N,N- dimethylformamide (3 ml) was added HATU (2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3- tetramethyluronium hexafluorophosphate ) (360 mg, 0.96 mmol) at 0°C, the resulting mixture was allowed to warm to room temperature and stirred for 20 minutes. TLC showed the reaction was complete. The mixture was partitioned between ethyl acetate (10 ml) and water (10 ml). The organic layer were collected, washed with brine (10 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluted with 10% methanol in dichloromethane) to afford ethyl 6-(4-(2-(2- (((S)-1-((2S,4R)-4-hydroxy-2-(((S)-1-(4-(4-methylthiazol-5- yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)amino)-2- oxoethoxy)ethyl)piperazin-1-yl)nicotinate (130 mg, yield 44%) as white solid. LC_MS: (ES+): m/z 750.3 [M+H]+. tR = 2.025min.
[0759] Step 6: Synthesis of 6-(4-(2-(2-(((S)-1-((2S,4R)-4-hydroxy-2-(((S)-1-(4-(4-methylthiazol- 5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)amino)-2- oxoethoxy)ethyl)piperazin-1-yl)nicotinic acid:
Figure imgf000335_0002
[0760] A mixture of ethyl 6-(4-(2-(2-(((S)-1-((2S,4R)-4-hydroxy-2-(((S)-1-(4-(4-methylthiazol- 5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)amino)-2- oxoethoxy)ethyl)piperazin-1-yl)nicotinate (130 mg, 0.173 mmol) and lithium hydroxide monohydrate (14.2 mg, 0.35 mmol) in tetrahydrofuran (5 ml)-water (2 ml)-methanol (2 ml) was stirred at 40°C overnight. TLC showed the reaction was completed. The mixture solution was cooled to room temperature, acidified with diluted hydrochloride acid (3N) till pH 3-4, and extracted with dichloromethane (10 ml × 2). The combined organic layers were washed with brine (10 ml), dried over sodium sulfate and concentrated under reduced pressure to afford 6-(4-(2-(2- (((S)-1-((2S,4R)-4-hydroxy-2-(((S)-1-(4-(4-methylthiazol-5- yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)amino)-2- oxoethoxy)ethyl)piperazin-1-yl)nicotinic acid (60 mg, crude) as colorless oil which was used in next step without purification.
[0761] Step 7: Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-6-(4-(2-(2-(((S)-1-((2S,4R)-4-hydroxy-2-(((S)-1-(4-(4-methylthiazol-5- yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)amino)-2- oxoethoxy)ethyl)piperazin-1-yl)nicotinamide:
Figure imgf000336_0001
[0762] To a stirred solution of -(4-(2-(2-(((S)-1-((2S,4R)-4-hydroxy-2-(((S)-1-(4-(4- methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2- yl)amino)-2-oxoethoxy)ethyl)piperazin-1-yl)nicotinic acid (60 mg, crude), 4-((1r,3r)-3-amino- 2,2,4,4-tetramethylcyclobutoxy)-2-chlorobenzonitrile hydrochloride (25 mg, 0.81 mmol), and N- ethyl-N-isopropylpropan-2-amine (40 mg, 0.32mmol) in anhydrous N,N-dimethylformamide (2 ml) was added HATU (2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium
hexafluorophosphate ) (61 mg, 0.16 mmol) at 0°C, the resulting mixture was allowed to warm to room temperature and stirred at room temperature for 20 min. TLC showed the reaction was complete. The mixture was partitioned between ethyl acetate (5 ml) and water (5 ml). The organic layer was collected, washed with brine (5ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue, which was purified by TLC (eluted with 10 % methanol in dichloromethane) to afford N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)- 2,2,4,4-tetramethylcyclobutyl)-6-(4-(2-(2-(((S)-1-((2S,4R)-4-hydroxy-2-(((S)-1-(4-(4- methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2- yl)amino)-2-oxoethoxy)ethyl)piperazin-1-yl)nicotinamide. (11.5 mg, yield 15%) as white solid. LC_MS: (ES+): m/z 996.4 [M+H]+. tR = 2.442 min. 1HNMR (400 MHz, CD3OD): ^ 1.08 (s, 9H). 1.23 (s, 6H), 1.29 (s, 6H), 1.51-1.58 (m, 3H), 1.98-2.06 (m, 1H), 2.18-2.25 (m, 1H), 2.49 (s, 3H), 2.90 (s, 6H), 3.76-3.88 (m, 8H), 4.06-4.15 (m, 3H), 4.29 (s, 1H), 4.41-4.46 (s, 1H), 4.57-4.26 (m, 1H), 4.71 (s, 1H), 5.01-5.04 (m, 1H), 6.88-7.00 (m, 2H), 7.14 (s, 1H), 7.42-7.45 (m, 4H), 7.74 (d, J = 8.4 Hz, 1H), 8.00 (d, J = 6.0 Hz, 1H), 8.64 (s, 1H), 8.89 (s, 1H). Chemical Formula:
C52H66ClN9O7S, Molecular Weight: 996.65.
[0763] Unless otherwise noted, Examples 594-770 were synthesized according to similar procedures described above for the synthesis of Example 608, by utilizing corresponding starting materials and reagents.
[0764] Table 18: Additional Exemplary Compounds
Figure imgf000338_0001
Figure imgf000339_0001
Figure imgf000340_0001
Figure imgf000341_0001
Figure imgf000342_0001
Figure imgf000343_0001
Figure imgf000344_0001
Figure imgf000345_0001
Figure imgf000346_0001
$^^^^^^^^^^^^^$#^^
Figure imgf000347_0001
Figure imgf000348_0001
$^^^^^^^^^^^^^$#^^
Figure imgf000349_0001
Figure imgf000350_0001
Figure imgf000351_0001
^^^^^^^^^^^^^^^^^^^^^^^^
Figure imgf000352_0001
Figure imgf000353_0001
Figure imgf000354_0001
Figure imgf000355_0001
Figure imgf000356_0001
Figure imgf000357_0001
Figure imgf000358_0001
Figure imgf000359_0001
Figure imgf000360_0001
$^^^^^^^^^^^^^$#^^
Figure imgf000361_0001
$^^^^^^^^^^^^^$#^^
Figure imgf000362_0001
Figure imgf000363_0001
Figure imgf000364_0001
Figure imgf000365_0001
Figure imgf000366_0001
Figure imgf000367_0001
Figure imgf000368_0001
Figure imgf000369_0001
Figure imgf000370_0001
Figure imgf000371_0001
Figure imgf000372_0001
Figure imgf000373_0001
Figure imgf000374_0001
Figure imgf000375_0001
Figure imgf000376_0001
Figure imgf000377_0001
Figure imgf000378_0001
Figure imgf000379_0001
Figure imgf000380_0001
Figure imgf000381_0001
Figure imgf000382_0001
Figure imgf000383_0001
Figure imgf000384_0001
Figure imgf000385_0001
Figure imgf000386_0001
Figure imgf000387_0001
Figure imgf000388_0001
Figure imgf000389_0001
$^^^^^^^^^^^^^$#^^
Figure imgf000390_0001
Figure imgf000391_0001
Figure imgf000392_0001
Figure imgf000393_0001
Figure imgf000394_0001
Figure imgf000395_0001
Figure imgf000396_0001
Figure imgf000397_0001
^^!^^"^^^^^^^
^^^^^^^^^%^^^^^^^^!^^^^
^^^^^^^^^^^^^^^^^^^^^^^^
^^^^^^^^^^^
^^^$'^ ^^^^^^^^^^^^^^^
^^^^^^^^^^^^
^^^^^^^^^^^^^^^^^ ^^^^^
770 ^^^^^^^^^^^^^^^^^^^^^ 1010.5 1012.4
5 ^^^$^^^^%$^^^^^^^^^^
^^^$+^$^#^^$^^^^^^^^^^
^^^^^^^ ^^^^^^^^^^
^^#^^^^^^^^^^$#$^^^^^
^^^^^^^^$#^^
Figure imgf000398_0001
[0766] Step 1: Synthesis of (2S,6R)-tert-butyl 4-(5-(methoxycarbonyl) pyridin-2-yl)-2,6- dimethylpiperazine-1-carboxylate.
Figure imgf000398_0002
[0767] A mixture of methyl 6-fluoronicotinate (200 mg, 0.93 mmol), (2S,6R)-tert-butyl 2,6- dimethylpiperazine-1-carboxylate (289 mg, 1.87 mmol) and N-ethyl-N-isopropylpropan-2-amine (362 mg, 2.80 mmol) in dimethyl sulfoxide (2 ml) was stirred at 100°C for 16 hours. TLC showed the reaction was complete. The reaction mixture was partitioned between ethyl acetate (20 ml) and water (10 ml). The organic layer was collected, and the aqueous layer was extracted with ethyl acetate (15 ml x 2). The combined organic layers were washed with brine (10 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash column chromatography (eluted with 20-40% ethyl acetate in hexane) to afford (2S,6R)-tert-butyl 4-(5-(methoxycarbonyl)pyridin-2-yl)-2,6- dimethylpiperazine-1-carboxylate (300 mg, yield 92%) as light yellow solid. LC_MS: (ES+): m/z 350.2 [M+H]+. tR = 3.020 min.
[0768] Step 2: Synthesis of methyl 6-((3S,5R)-3,5-dimethylpiperazin-1-yl)nicotinate TFA salt.
Figure imgf000399_0001
[0769] A mixture of (2S,6R)-tert-butyl 4-(5-(methoxycarbonyl)pyridin-2-yl)-2,6- dimethylpiperazine -1-carboxylate (300 mg, 0.86 mmol) and 2,2,2-trifluoroacetic acid (2 ml) in dichloromethane (2 ml) was stirred at room temperature for 2 hours. TLC showed the reaction was complete. The volatiles were evaporated under reduced pressure. The residue was taken up in dichloromethane (15 ml) and washed with aqueous sodium carbonate (sat, 20 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give methyl 6-((3S,5R)- 3,5-dimethylpiperazin-1-yl) nicotinate (180 mg, yield 84%) as light yellow solid which was used in next step directly without further purification.
[0770] Step 3: Synthesis of methyl 6-((3S, 5R)-4-(2-(2-(tert-butoxy)-2- oxoethoxy)ethyl)-3, 5- dimethylpiperazin-1-yl)nicotinate.
Figure imgf000399_0002
[0771] To a solution of methyl 6-((3S,5R)-3,5-dimethylpiperazin-1-yl)nicotinate(180 mg, 0.72 mmol), triethylamine(146 mg, 1.44 mmol) in acetonitrile (2 ml) was added tert-butyl 2-(2- (tosyloxy)ethoxy)acetate (238 mg, 0.72 mmol). The resulting mixture was stirred at 70°C for 16 hours. TLC showed the reaction was complete. To the reaction mixture was added water (20 ml) and ethyl acetate (20 ml). The organic layer was collected, and the aqueous layer was extracted with ethyl acetate (10 ml x 2). The combined organic layers were washed with brine (30 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by preparative TLC (eluting with 5% methanol in dichloromethane) to afford methyl 6-((3S, 5R)-4-(2-(2-(tert-butoxy)-2- oxoethoxy)ethyl)-3, 5-dimethylpiperazin-1- yl)nicotinate (75 mg, yield 25%) as light yellow solid. LC_MS: (ES+): m/z 408.3 [M+H]+. tR = 2.059 min.
[0772] Step 4: Synthesis of methyl 4-(5-(3-aminopropoxy)pentyl)benzoate.
Figure imgf000400_0001
[0773] A mixture of methyl 6-((3S, 5R)-4-(2-(2-(tert-butoxy)-2- oxoethoxy)ethyl)-3, 5- dimethylpiperazin-1-yl)nicotinate (75 mg, 0.18 mmol) and 2,2,2-trifluoroacetic acid (1 ml) in dichloromethane (1 ml) was stirred at room temperature for 2 hours. TLC showed the reaction was complete. The volatiles were evaporated under reduced pressure to afford methyl 4-(5-(3- aminopropoxy)pentyl)benzoate (64.8 mg, yield 100%) as light yellow solid which was used to the next step without further purification.
[0774] Step 5: Synthesis of methyl 6-((3S,5R)-4-(2-(2-(((2S)-1-((4R)-4-hydroxy -2-(((S)-1-(4- (4-methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2- yl)amino)-2-oxoethoxy)ethyl)-3,5-dimethylpiperazin-1-yl)nicotinate.
Figure imgf000401_0001
[0775] To a stirred solution of methyl 4-(5-(3-aminopropoxy)pentyl)benzoate (64.8 mg, 0.18 mmol), (4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-((S)-1-(4-(4-methylthiazol-5- yl)phenyl)ethyl)pyrrolidine-2-carboxamide hydrochloride (88.6 mg, 0.18 mmol), and N-ethyl-N- isopropylpropan-2-amine (143 mg, 1.1 mmol) in anhydrous N,N-dimethylformamide (1 ml) was added HATU (2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium
hexafluorophosphate) (139 mg, 0.37 mmol) at 0°C, the resulting mixture was allowed to warm to room temperature and stirred for 40 min. TLC showed the reaction was complete. The mixture was partitioned between ethyl acetate (25 ml) and water (15 ml). The organic layer was collected, washed with brine (15 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluted with 5 % methanol in dichloromethane) to afford methyl 6-((3S,5R)-4-(2-(2-(((2S)-1-((4R)-4- hydroxy -2-(((S)-1-(4-(4-methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3- dimethyl-1-oxobutan-2-yl)amino)-2-oxoethoxy)ethyl)-3,5-dimethylpiperazin-1-yl)nicotinate (140 mg, yield 98%).
[0776] Step 6: Synthesis of N-((1r,3R)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-6-((3S,5R)-4-(2-(2-(((2S)-1-((4R)-4-hydroxy-2-(((S)-1-(4-(4- methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2- yl)amino)-2-oxoethoxy)ethyl)-3,5-dimethylpiperazin-1-yl)nicotinamide.
Figure imgf000402_0001
[0777] A mixture of methyl 6-((3S,5R)-4-(2-(2-(((2S)-1-((4R)-4-hydroxy -2-(((S)-1-(4-(4- methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2- yl)amino)-2-oxoethoxy)ethyl)-3,5-dimethylpiperazin-1-yl)nicotinate (140 mg, 0.18 mmol) and lithium hydroxide monohydrate (15 mg, 0.36 mmol) in tetrahydrofuran (2 ml)-water (0.5 ml)- methanol (0.5 ml) was stirred at 45°C overnight. TLC showed the reaction was complete. The mixture solution was concentrated and the residue was acidified with diluted hydrochloride acid (1N) till pH 5-6, extracted with dichloromethane (10 ml × 3). The combined organic layers were washed with brine (10 ml), dried over sodium sulfate and concentrated. The residue was taken up in anhydrous N,N-dimethylformamide (2 ml), followed by N-ethyl-N-isopropylpropan-2-amine (132 mg, 1.02 mmol), 4-((1r,3r)-3-amino-2,2,4,4-tetramethylcyclobutoxy)-2-chlorobenzonitrile hydrochloride (54 mg, 0.17 mmol), and HATU (2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3- tetramethyluronium hexafluorophosphate) (129 mg, 0.34 mmol) at 0°C, the resulting mixture was allowed to warm to room temperature and stirred for 20 minutes. TLC showed the reaction was complete. The mixture was partitioned between ethyl acetate (30 ml) and water (15 ml). The organic layer was collected, washed with brine (10 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluted with 10% methanol in dichloromethane) to afford N-((1r,3R)-3-(3- chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-6-((3S,5R)-4-(2-(2-(((2S)-1-((4R)-4- hydroxy-2-(((S)-1-(4-(4-methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3- dimethyl-1-oxobutan-2-yl)amino)-2-oxoethoxy)ethyl)-3,5-dimethylpiperazin-1-yl)nicotinamide (16.3 mg, yield 9.3%) as white solid. 1H NMR (400 MHz, CD3OD): ^ 0.93 (s, 9H), 1.12 (s, 6H), 1.17 (s, 12H) 1.42-1.44 (m, 3H), 1.87-1.89 (m, 1H), 2.05-2.08 (m, 1H), 2.38 (s, 3H), 2.66-2.73 (m, 2H), 2.81 (m, 2H), 3.04 (m, 2H), 3.58-3.65 (m, 3H), 3.71-3.74 (m, 1H), 3.87-3.91 (m, 2H), 4.04 (m, 1H), 4.12-4.25(m, 3H), 4.32 (s, 1H), 4.41-4.51 (m, 1H), 4.60 (m, 1H), 4.87-4.93 (m, 1H), 6.74-6.76 (m, 1H), 6.86-6.90 (m, 1H), 7.04 (s, 1H), 7.21-7.35 (m, 4H), 7.61-7.62 (d, J = 8.8 Hz, 1H), 7.85-7.87 (m, 1H), 8.48 (br, 1H), 8.77 (br, 1H), 8.98 (s, 1H)). Chemical Formula: C53H68ClN9O8S; Molecular Weight: 1026.68.
[0778] Unless otherwise noted, Examples 771-793 were synthesized according to similar procedures described above for the synthesis of Example 786, by utilizing corresponding starting materials and reagents. [0779] Table 19: Additional Exemplary Compounds
Figure imgf000403_0001
Figure imgf000404_0001
Figure imgf000405_0001
[0780] Table 20: Exemplary Compounds.
Figure imgf000405_0002
Figure imgf000406_0001
Figure imgf000407_0001
Figure imgf000408_0001
Figure imgf000409_0001
^^^^$^^^$^^^$#^^ [0781] Synthesis of Example 794:
Figure imgf000410_0001
[0782] Step 1: Synthesis of benzyl 6-fluoronicotinate.
Figure imgf000410_0002
[0783] A mixture of 6-fluoronicotinic acid (10 g, 70.9 mmol), benzyl bromide (18 g, 106 mmol) and potassium carbonate (18.2 g, 141 mmol) in N,N-dimethylformamide (100 ml) was stirred at room temperature for 12 hours. TLC showed the reaction was complete. The reaction mixture was allowed partitioned between water (100 ml) and ethyl acetate (100 ml). The organic layer was collected, washed with brine (100 ml x 2), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash
chromatography (eluted with 20% ethyl acetate in hexane) to afford benzyl 6-fluoronicotinate (15 g, yield 93%) as yellow solid. 1HNMR (400 MHz, CDCl3): ^ 5.37 (s, 2H), 6.96-6.98(m, 1H), 7.34-7.45 (m, 5H), 8.38-8.43 (m, 1H), 8.90-8.91 (m, 1H). Chemical Formula: C13H10FNO2, Molecular Weight: 231.22.
[0784] Step 2: Synthesis of tert-butyl 4-(5-((benzyloxy)carbonyl)pyridin-2-yl)piperazine-1- carboxylate.
Figure imgf000411_0001
[0785] A mixture of benzyl 6-fluoronicotinate (15 g, 65 mmol), di-tert-butyl piperazine-1,4- dicarboxylate (12 g, 65mmol) and N-ethyl-N-isopropylpropan-2-amine (16.6g, 129 mmol) in dry 1-methylpyrrolidin-2-one (80 ml) was stirred at 80°C for 12 hours. TLC showed the reaction was complete. The reaction mixture was partitioned between water (100 ml) and ethyl acetate (100 ml). The organic layer was collected, washed with brine (100 ml x 2), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to afford a crude residue which was purified by silica gel flash chromatography (eluted with 20-50% ethyl acetate in hexane) to afford tert-butyl 4-(5-((benzyloxy)carbonyl)pyridin-2-yl)piperazine-1-carboxylate (20 g, yield 80%) as white solid.
[0786] Step 3: Synthesis of 6-(4-(tert-butoxycarbonyl)piperazin-1-yl)nicotinic acid.
Figure imgf000411_0002
[0787] A mixture of tert-butyl 4-(5-((benzyloxy)carbonyl)pyridin-2-yl)piperazine-1-carboxylate (2.0 g, 5.03 mmol) and palladium on carbon (10%, 200 mg) in ethanol (20 ml) was stirred at 30°C overnight under hydrogen atmosphere (hydrogen balloon). TLC showed the reaction was complete. Palladium on carbon was removed through filtration and washed with ethanol (20 ml x 2). The combined filtrates were concentrated under reduced pressure to give 6-(4-(tert- butoxycarbonyl)piperazin-1-yl)nicotinic acid (1.6 g, crude) as colorless oil which was used in next step without purification.
[0788] Step 4: Synthesis of tert-butyl 4-(5-(((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)piperazine-1-carboxylate.
Figure imgf000412_0001
[0789] To a stirred solution of 6-(4-(tert-butoxycarbonyl)piperazin-1-yl)nicotinic acid (300 mg, 0.97 mmol), 4-((1r,3r)-3-amino-2,2,4,4-tetramethylcyclobutoxy)-2-chlorobenzonitrile
hydrochloride (306 mg, 0.97 mmol), and N-ethyl-N-isopropylpropan-2-amine (309 mg, 2.4 mmol) in anhydrous N,N-dimethylformamide (8 ml) was added HATU (2-(7-Aza-1H- benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate) (684 mg, 1.8 mmol) at 0°C, the resulting mixture was allowed to warm to room temperature and stirred for 20 min. TLC showed the reaction was complete. The mixture was partitioned between ethyl acetate (50 ml) and water (80ml). The organic layer was collected, washed with brine (10ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluted with 10 % methanol in dichloromethane) to afford tert-butyl 4-(5-(((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)piperazine-1-carboxylate (400 mg, yield 72%) as white solid.
[0790] Step 5: Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-6-(pi
Figure imgf000412_0002
[0791] A mixture of tert-butyl 4-(5-(((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)piperazine-1-carboxylate (80 mg, 0.14 mmol) in hydrogen chloride in dioxane solution (4M, 2 ml) was stirred at room temperature for 2 hours. TLC showed the reaction was complete. The volatiles were evaporated under reduced pressure. The residue was taken up in dichloromethane (20 ml) and washed with aqueous sodium bicarbonate solution (1N, 5 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by pre-TLC (eluted with 10 % methanol in dichloromethane) to afford [N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-6-(piperazin-1-yl)nicotinamide (32 mg, yield 50%) as white solid.
LC_MS: (ES+): m/z 468.6 [M+H]+. tR = 2.285 min. 1HNMR (400 MHz, CD3OD): ^ 1.07-1.38 (m, 12H), 3.12-3.40 (m, 4H), 3.51-3.86 (m, 1H), 3.94 (br, 3H), 4.17-4.30 (m, 2H), 6.99-7.15 (m, 2H), 7.74(s, 1H), 8.05(s, 1H), 8.48-8.68 (m, 2H). Chemical Formula: C25H30ClN5O2; Molecular Weight: 467.99.
[0792] Step 6: Synthesis of tert-butyl 3-(4-(5-(((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)carbamo l ridin-2- l i erazin-1- l azetidine-1-carboxylate.
Figure imgf000413_0001
[0793] A mixture of tert-butyl 3-oxoazetidine-1-carboxylate (87 mg, 0.51 mmol), N-((1r,3r)-3- (3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)-6-(piperazin-1-yl)nicotinamide (200 mg, 0.43 mmol), acetic acid (45 mg, 0.75 mmol) in methanol (10 ml) was stirred at room temperature for 30 min, followed by addition of sodium cyanoborohydride (53 mg, 0.86 mmol) at room temperature. The resulting mixture was stirred at room temperature for 4 hours. The mixture was partitioned between ethyl acetate (50 ml) and water (30 ml). The organic layer was collected, washed with brine (20 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to afford crude residue which was purified by silica gel flash chromatography (eluted with 10 % methanol in dichloromethane) to afford tert-butyl 3-(4-(5-(((1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)piperazin-1-yl)azetidine-1- carboxylate (143 mg, yield 52%) as white solid.
[0794] Step 7: Synthesisi of 6-(4-(azetidin-3-yl)piperazin-1-yl)-N-((1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)nicotinamide hydrochloride.
Figure imgf000414_0001
[0795] A solution of tert-butyl 3-(4-(5-(((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)piperazin-1-yl)azetidine-1-carboxylate (140 mg, 0.22mmol) in hydrogen chloride in 1,4-dioxane (4M, 5 ml) was stirred at room temperature for 6 hours. TLC showed the reaction was complete. The volatiles were removed under reduced pressure to give 6-(4-(azetidin-3-yl)piperazin-1-yl)-N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)- 2,2,4,4-tetramethylcyclobutyl)nicotinamide hydrochloride (130 mg, crude) as white solid which was used in next step without further purification.
[0796] Step 8: Synthesis of tert-butyl 2-(3-(4-(5-(((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)piperazin-1-yl)azetidin-1-yl)acetate.
Figure imgf000414_0002
[0797] To a stirred solution of 6-(4-(azetidin-3-yl)piperazin-1-yl)-N-((1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)nicotinamide hydrochloride (130 mg, crude), N- ethyl-N-isopropylpropan-2-amine (148 mg, 1.14 mmol) in acetonitrile (5 ml) was added tert-butyl 2-bromoacetate (45 mg, 0.23 mmol) at room temperature. The resulting mixture was stirred at room temperature overnight. TLC showed the reaction was complete. The mixture was partitioned between ethyl acetate (30 ml) and water (20 ml). The organic layer was collected, washed with brine (10 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by preparative TLC (eluted with 10 % methanol in dichloromethane) to afford tert-butyl 2-(3-(4-(5-(((1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)piperazin-1-yl)azetidin-1- yl)acetate (70 mg, yield 49%, over 2 steps) as yellow solid.
[0798] Step 9: Synthesisi of 2-(3-(4-(5-(((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)piperazin-1-yl)azetidin-1-yl)acetic acid.
Figure imgf000415_0001
[0799] A solution of tert-butyl 2-(3-(4-(5-(((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)piperazin-1-yl)azetidin-1-yl)acetate (65 mg, 0.10 mmol) in hydrogen chloride in 1,4-dioxane (4M, 5 ml) was stirred at room temperature for 2 hours. TLC showed the reaction was complete. The volatiles were removed under reduced pressure to give 2-(3-(4-(5-(((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)piperazin-1-yl)azetidin-1-yl)acetic acid (60 mg, crude) as white solid which was used in next step without further purification.
[0800] Step 10: Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-6-(4-(1-(2-(((2S)-1-((4R)-4-hydroxy-2-(((S)-1-(4-(4-methylthiazol-5- yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)amino)-2- oxoethyl)azetidin-3-yl)pi
Figure imgf000415_0002
[0801] To a stirred solution of 2-(3-(4-(5-(((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)piperazin-1-yl)azetidin-1-yl)acetic acid (60 mg, crude), (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-((S)-1-(4-(4-methylthiazol- 5-yl)phenyl)ethyl)pyrrolidine-2-carboxamide hydrochloride (49 mg, 0.10 mmol), and N-ethyl-N- isopropylpropan-2-amine (78 mg, 0.6 mmol) in anhydrous N,N-dimethylformamide (2 ml) was added HATU (2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium
hexafluorophosphate) (58 mg, 0.15 mmol) at 0°C, the resulting mixture was allowed to warm to room temperature and stirred at room temperature for 20 min. TLC showed the reaction was complete. The mixture was partitioned between ethyl acetate (30 ml) and water (20 ml). The organic layer was collected, washed with brine (20 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by prep. HPLC to afford N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)-6-(4-(1-(2-(((2S)- 1-((4R)-4-hydroxy-2-(((S)-1-(4-(4-methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)- 3,3-dimethyl-1-oxobutan-2-yl)amino)-2-oxoethyl)azetidin-3-yl)piperazin-1-yl)nicotinamide (15 mg, yield 13.6%, over 2 steps) as white solid. LC_MS: (ES+): m/z 1007.5 [M+H]+. tR = 2.397 min. 1H NMR (400 MHz, CD3OD): ^ 0.93, 0.95 (two singles, 9H), 1.12(s, 6H), 1.18 (s, 6H),1.41- 1.48 (m, 3H), 1.85-1.94 (m, 2H), 2.07-2.11 (m, 1H), 2.22-2.33 (m, 1H), 2.34-2.38 (m, 6H), 3.01- 3.14 (m, 3H), 3.17-3.19 (m, 2H), 3.55-3.66 (m, 6H), 3.71-3.76 (m, 1H), 4.01-4.06 (m, 1H), 4.18 (s, 1H), 4.31-4.37 (m, 1H), 4.47-4.54 (m, 2H), 4.88-4.92 (m, 1H), 6.74 (d, J = 8.8 Hz, 1H), 6.88 (dd, J = 2.4, 8.8 Hz, 1H), 7.03 (d, J = 2.4 Hz, 1H), 7.27-7.36 (m, 4H), 7.62 (d, J = 8.8 Hz, 1H), 7.87 (dd, J = 2.0, 9.2 Hz, 1H), 8.51 (d, J = 2.0 Hz, 1H), 8.77 (s, 1H). Chemical Formula:
C53H67ClN10O6S; Molecular Weight: 1007.68. [0802] Synthesis of Example 795:
Figure imgf000416_0001
[0803] Step 1: Synthesis of benzyl 3-(4-(2-tert-butoxy-2-oxoethyl)piperazin-1-yl)azetidine-1- carboxylate.
Figure imgf000417_0001
[0804] To a stirred solution of benzyl 3-oxoazetidine-1-carboxylate (300 mg, 1.46 mmol), tert- butyl 2-(piperazin-1-yl)acetate (307 mg, 1.53 mmol) and acetic acid glacial (1 d) in methanol (4 ml) was added sodium cyanoborohydride (276 mg, 4.39 mmol) at room temperature. The mixture was stirred at room temperature for 1.5 hours. TLC showed the reaction was complete. The reaction mixture was partitioned between ethyl acetate (50 ml) and water (25 ml). The organic layer was collected, washed with brine (25 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluted with 20-30 % ethyl acetate in hexane) to afford benzyl 3-(4-(2-tert- butoxy-2-oxoethyl)piperazin-1-yl)azetidine-1-carboxylate (145 mg, yield 25%) as white solid. 1H NMR (400 MHz, CDCl3): ^ 1.46 (s, 9H), 2.43 (br, 4H), 2.62 (br, 4H), 3.11 (s, 2H), 3.13-3.18 (m, 1H), 3.87-3.90 (m, 2H), 3.99-4.03 (m, 2H), 5.08 (s, 2H), 7.30-7.35 (m, 5H). Chemical Formula: C21H31N3O4; Molecular Weight: 389.49.
[0805] Step 2: Synthesis of tert-butyl 2-(4-(azetidin-3-yl)piperazin-1-yl)acetate.
Figure imgf000417_0002
[0806] A mixture of benzyl 3-(4-(2-tert-butoxy-2-oxoethyl)piperazin-1-yl)azetidine-1- carboxylate (145 mg, 0.37 mmol) and palladium hydroxide on carbon (10%, 15 mg) in ethanol (5 ml) was stirred at room temperature overnight under hydrogen atmosphere (hydrogen balloon). TLC showed the reaction was complete. Palladium on carbon was removed through filtration and washed with methanol (5 ml x2). The combined filtrates were concentrated under reduced pressure to give tert-butyl 2-(4-(azetidin-3-yl)piperazin-1-yl)acetate (85 mg, yield 89%) as colorless oil which was used in next step directly without further purification. 1H NMR (400 MHz, CDCl3): ^ 1.46 (s, 9H), 2.40 (br, 4H), 2.60 (br, 4H), 2.87-2.95 (m, 1H), 3.09-3.11 (m, 2H), 3.16-3.27 (m, 1H), 3.45-3.50 (m, 1H), 3.52-3.56 (m, 1H), 3.59-3.63 (m, 1H). Chemical Formula: C13H25N3O2; Molecular Weight: 255.36.
[0807] Step 3: Synthesis of methyl 6-(3-(4-(2-tert-butoxy-2-oxoethyl)piperazin-1-yl)azetidin-1- yl)nicotinate.
Figure imgf000418_0001
[0808] A mixture of tert-butyl 2-(4-(azetidin-3-yl)piperazin-1-yl)acetate (85 mg, 0.33 mmol), methyl 6-fluoronicotinate (103 mg, 0.66 mmol) and triethylamine (135 mg, 1.33 mmol) in dimethyl sulfoxide (2 ml) was stirred at 110°C for 5 hours. TLC showed the reaction was complete. The reaction mixture was partitioned between ethyl acetate (20 ml) and water (10 ml). The organic layer was collected, washed with brine (10 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluted with 5 % methanol in dichloromethane) to afford methyl 6-(3-(4-(2- tert-butoxy-2-oxoethyl)piperazin-1-yl)azetidin-1-yl)nicotinate (63 mg, yield 48%) as yellow solid. LC_MS: (ES+): m/z 391.2 [M+H]+. tR = 1.532 min. 1H NMR (400 MHz, CDCl3):^ 1.46 (s, 9H), 2.52 (br, 4H), 2.64 (br, 4H), 3.12 (s, 2H), 3.33-3.40 (m, 1H), 3.86 (s, 3H), 3.95-3.99 (m, 2H), 4.13-4.17 (m, 2H), 6.21 (d, J = 8.8 Hz, 1H), 7.97-7.80 (m, 1H), 8.76-8.77 (m, 1H). Chemical Formula: C20H30N4O4; Molecular Weight: 390.48.
[0809] Step 4: Synthesis of methyl 6-(3-(4-(2-((2S)-1-((4R)-4-hydroxy-2-((S)-1-(4-(4- methylthiazol-5-yl)phenyl)ethylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-ylamino)- 2-oxoethyl)piperazin-1-yl)azetidin-1-yl)nicotinate.
Figure imgf000418_0002
[0810] A mixture of methyl 6-(3-(4-(2-tert-butoxy-2-oxoethyl)piperazin-1-yl)azetidin-1- yl)nicotinate (63 mg, 0.16 mmol) and 2,2,2-trifluoroacetic acid (1 ml) in dichloromethane (1 ml) was stirred at room temperature for 2 hour. TLC showed the reaction was complete. The volatiles were evaporated under reduced pressure. The residue was taken up in anhydrous N,N- dimethylformamide (2 ml), followed by sequentially addition of (2S,4R)-1-((S)-2-amino-3,3- dimethylbutanoyl)-4-hydroxy-N-((S)-1-(4-(4-methylthiazol-5-yl)phenyl)ethyl)pyrrolidine-2- carboxamide hydrochloride (78 mg, 0.16 mmol), N-ethyl-N-isopropylpropan-2-amine (104 mg, 0.80 mmol), and HATU (2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium
hexafluorophosphate) (184 mg, 0.48 mmol) at 0°C, the resulting mixture was allowed to warm up to room temperature and stirred for 1 hour. LCMS showed formation of desired product. The reaction mixture was partitioned between ethyl acetate (40 ml) and water (20 ml). The organic layer was collected, washed with brine (10 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluted with 10% methanol in dichloromethane-1% ammonium hydroxide) to afford methyl 6-(3-(4-(2-((2S)-1-((4R)-4-hydroxy-2-((S)-1-(4-(4-methylthiazol-5- yl)phenyl)ethylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-ylamino)-2- oxoethyl)piperazin-1-yl)azetidin-1-yl)nicotinate (88 mg, yield 72%) as yellow solid. LC_MS: (ES+): m/z 761.4 [M+H]+. tR = 1.893 min. 1H NMR (400 MHz, CDCl3):^ 1.07 (s, 9H), 1.48 (d, J = 6.8 Hz, 3H), 1.99-2.11 (m, 2H), 2.50 (br, 4H), 2.54 (s, 3H), 2.63 (br, 4H), 3.01-3.10 (m, 2H), 3.35-3.40 (m, 1H), 3.49 (s, 1H), 3.57-3.61 (m, 1H), 3.87 (s, 3H), 3.95-3.99 (m, 2H), 4.15-4.20 (m, 3H), 4.45 (d, J = 8.4 Hz, 1H), 4.51 (br, 1H), 4.76 (t, J = 7.6 Hz, 1H), 5.04-5.12 (m, 1H), 5.22 (d, J = 8.8 Hz, 1H), 7.36-7.42 (m, 5H), 7.80-7.83 (m, 1H), 7.99-8.01 (m, 1H), 8.68 (s, 1H), 8.76-8.77 (m, 1H). Chemical Formula: C39H52N8O6S; Molecular Weight: 760.95.
[0811] Step 5: Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-6-(3-(4-(2-((2S)-1-((4R)-4-hydroxy-2-((S)-1-(4-(4-methylthiazol-5- yl)phenyl)ethylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-ylamino)-2- oxoethyl)piperazin-1-yl)azetidin-1-yl)nicotinamide.
Figure imgf000420_0001
[0812] A mixture of methyl 6-(3-(4-(2-((2S)-1-((4R)-4-hydroxy-2-((S)-1-(4-(4-methylthiazol-5- yl)phenyl)ethylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-ylamino)-2- oxoethyl)piperazin-1-yl)azetidin-1-yl)nicotinate (88 mg, 0.12 mmol) and lithium hydroxide monohydrate (10 mg, 0.24 mmol) in tetrahydrofuran (2 ml)-water (0.5 ml)-methanol (0.5 ml) was stirred at room temperature overnight. TLC showed the reaction was complete. The mixture solution acidified with diluted hydrochloride acid (3N) till pH 5-6. The volatiles were evaporated under reduced pressure. The residue was taken up in N,N-dimethylformamide (1 ml), followed by sequentially addition of 4-((1r,3r)-3-amino-2,2,4,4-tetramethylcyclobutoxy)-2-chlorobenzonitrile hydrochloride (36 mg, 0.11 mmol), N-ethyl-N-isopropylpropan-2-amine (75 mg, 0.58 mmol), and HATU (2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate) (132 mg, 0.35 mmol) at 0°C, the resulting mixture was allowed to warm up to room temperature and stirred at room temperature for 30 minutes. LCMS showed formation of desired product. The reaction mixture was partitioned between ethyl acetate (20 ml) and water (10 ml). The organic layer was collected, washed with brine (10 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by pre-HPLC to afford N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)-6-(3-(4-(2-((2S)-1- ((4R)-4-hydroxy-2-((S)-1-(4-(4-methylthiazol-5-yl)phenyl)ethylcarbamoyl)pyrrolidin-1-yl)-3,3- dimethyl-1-oxobutan-2-ylamino)-2-oxoethyl)piperazin-1-yl)azetidin-1-yl)nicotinamide (17.2 mg, yield 15%) as white solid. LC_MS: (ES+): m/z 1007.4 [M+H]+. tR = 2.365 min. 1H NMR (400 MHz, CD3OD):^ 0.93, 0.95 (two singles, 9H), 1.12 (s, 6H), 1.18 (s, 6H), 1.41-1.48 (m, 3H), 1.82- 1.88 (m, 1H), 2.07-2.14 (m, 1H), 2.38 (s, 3H), 2.50-2.59 (m, 8H), 3.03 (s, 2H), 3.32-3.35 (m, 1H), 3.63-3.67 (m, 1H), 3.75-3.77 (m, 1H), 3.86-3.89 (m, 2H), 4.03-4.05 (m, 1H), 4.09-4.12 (m, 2H), 4.18 (s, 1H), 4.34 (br, 1H), 4.45-4.49 (m, 1H), 4.54 (s, 1H), 4.90-4.93 (m, 1H), 6.36 (d, J = 8.8 Hz, 1H), 6.87-6.90 (m, 1H), 7.02-7.03 (m, 1H), 7.27-7.36 (m, 4H), 7.58-7.63 (m, 2H), 7.86-7.88 (m, 1H), 8.11 (br, 1H), 8.44 (s, 1H), 8.50 (d, J = 7.6 Hz, 1H), 8.78 (s, 1H). Chemical Formula: C53H67ClN10O6S; Molecular Weight: 1007.68.
[0813] Unless otherwise noted, Examples 796-808 were synthesized according to similar procedures described above for the synthesis of Examples 794 and 795, by utilizing
corresponding starting materials and reagents.
[0814] Table 21: Additional Exemplary Compounds.
Figure imgf000421_0001
Figure imgf000422_0001
[0815] Table 22: Exemplary Compounds Measured Ex. Mass Ion Data # Structure Compound Name
m/z m/z (1) (2) ^^^^^^^^"^^^^^^^^^^^^^^^^
^^^^^^^^^^^^^^^^^^^ ^^^^^
^^^^^"^^"^^^^^^^^^^!^^^^
^^^!^^"^^^^^^#^^^^^^^^^^!^^
^^^^^^^^^^^^^^^^$^%^^^^^
^^^^^^^^^^^^^^^^^^^^^^^^^^
798 ^^^^^^$#$^^^^^^^^^^^^ 950.9 952.9
#$^^^^^^^^^^^^^ ^^^^^^
^^^^^$^^^^^^^^^^^^^^^^^
^^^^
#$^%^^$^^^^^)^^^^^*^^^^^^^
^^^^^^$^^^$^^^$#^^
^^^^^^^^^^^^^^^^^^^^^^^^^
^^^^^^^^^^^^^^^^^^^^^^
^^^^^^^^^^^^^^^^^ ^^^^^^^
^^^^^^^^#$'^ ^^^^^^^^^!^^^^
^^^!^^"^^^^^^#^^^^^^^^^^!^^
799 1030.
^^^^^^^^^^^^^^^^$^%^^^^^ 4
^^^^^^^^^^^^^^^^^^^^^^^^^^
^^^^^^$#$^^^^^^^^^^^^
Figure imgf000422_0002
#$^^^^^^^^^^^^^ ^^^^^^
^^^^^$^^^^^^
Figure imgf000423_0001
^^^^^^^^^^^$#^^
Figure imgf000424_0001
Figure imgf000425_0001
^^^^^^^^^^^$#^^
Figure imgf000426_0001
[0816] Synthesis of Example 809:
Figure imgf000427_0001
[0817] Step 1: Synthesis of benzyl 2 ):
Figure imgf000427_0002
[0818] A mixture of piperazine (5.6 g, 65.4 mmol) and benzyl 2-bromoacetate (5 g, 21.8 mmol) in ethanol (50 ml) was stirred at 45oC for 16 hours. TLC showed the reaction was complete. The reaction mixture was partitioned between water (50 ml) and ethyl acetate (100 ml). The organic layer was collected, washed with brine (30 ml x 2), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluted with 8% methanol in dichloromethane) to afford benzyl 2-(piperazin-1- yl)acetate (2.5 g, yield 50%) as colorless oil. LC_MS: (ES+): m/z 235.4 [M+H]+. tR = 1.305 min. 1HNMR (400 MHz, CDCl3): ^ 2.56 (t, J = 4.8 Hz, 4H), 2.93 (t, J = 5 Hz, 4H), 3.25 (s, 2H), 5.16 (s, 2H), 7.33-7.36 (m, 5H). Chemical Formula: C13H18N2O2; Molecular Weight: 234.29. [0819] Step 2. Synthesis of tert-butyl 6-(4-(2-(benzyloxy)-2-oxoethyl)piperazin-1-yl)-2- azaspiro[3.3]heptane-2-carboxylate.
Figure imgf000428_0001
[0820] A mixture of tert-butyl 6-oxo-2-azaspiro[3.3]heptane-2-carboxylate (450 mg, 2.1 mmol), benzyl 2-(piperazin-1-yl)acetate (498 mg, 2.1 mmol) and acetic acid (120 mg, 2.0 mmol) in methanol (15 ml) was stirred at room temperature for 30 minutes, followed by addition of sodium cyanoborohydride (256 mg, 4.3 mmol) at room temperature. The resulting mixture was stirred at room temperature for 1 hour. The reaction mixture was diluted with ethyl acetate (30 ml), washed with water (10 ml x 2), brine (10 ml) and dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash column chromatography (eluted with 3-5 % methanol in dichloromethane) to afford tert-butyl 6-(4-(2- (benzyloxy)-2-oxoethyl)piperazin-1-yl)-2-azaspiro[3.3]heptane-2-carboxylate (570 mg, yield 62%) as white solid. LC_MS: (ES+): m/z 430.7 [M+H]+. tR = 1.917 min. 1HNMR (400 MHz, CDCl3): ^ 1.42 (s, 9H), 2.07-2.12 (m, 2H), 2.27-2.30 (m, 2H), 2.64-2.68 (m, 5H), 2.89-2.90 (m, 4H), 3.27 (s, 2H), 3.81 (s, 2H), 3.92 (s, 2H), 5.16 (s, 2H), 7.32-7.37 (m, 5H). Chemical Formula: C24H35N3O4; Molecular Weight: 429.55.
[0821] Step 3: Synthesis of benzyl 2-(4-(2-azaspiro[3.3]heptan-6-yl)piperazin-1-yl)acetate hydrochloride:
Figure imgf000428_0002
[0822] A solution of tert-butyl 6-(4-(2-(benzyloxy)-2-oxoethyl)piperazin-1-yl)-2- azaspiro[3.3]heptane-2-carboxylate (570 mg, 1.3 mmol) in hydrogen chloride in 1,4-dioxane (4M, 5 ml) was stirred at room temperature for 1 hour. TLC showed the reaction was complete. The volatiles were removed under reduced pressure to give the benzyl 2-(4-(2-azaspiro[3.3]heptan-6- yl)piperazin-1-yl)acetate hydrochloride (400 mg, crude) as white solid which was used in next step without purification.
[0823] Step 4: Synthesis of methyl 6-(6-(4-(2-(benzyloxy)-2-oxoethyl)piperazin-1-yl)-2- azaspiro[3.3]heptan-2-yl)nicotinate.
Figure imgf000429_0001
[0824] A mixture of benzyl 2-(4-(2-azaspiro[3.3]heptan-6-yl)piperazin-1-yl)acetate
hydrochloride (400 mg, crude), methyl 6-fluoronicotinate (170 mg, 1.1 mmol) and N-ethyl-N- isopropylpropan-2-amine (282 mg, 2.2 mmol) in dry 1-methyl-2-pyrrolidinone (8 ml) was stirred at 80°C for 16 hours. TLC showed the reaction was complete. The reaction mixture was partitioned between water (50 ml) and ethyl acetate (30 ml). The organic layer was collected, washed with brine (10 ml x 2), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluted with 0-8% methanol in dichloromethane) to afford methyl 6-(6-(4-(2-(benzyloxy)-2- oxoethyl)piperazin-1-yl)-2-azaspiro[3.3]heptan-2-yl)nicotinate (120 mg, yield 23%) as white solid. LC_MS: (ES+): m/z 465.4 [M+H]+. tR = 1.747 min. 1HNMR (400 MHz, CDCl3): ^ 2.11- 2.16 (m, 2H), 2.37-2.41 (m, 5H), 2.62-2.71 (m, 5H), 3.27 (s, 2H), 3.86 (s, 3H), 4.01 (s, 2H), 4.12 (s, 2H), 5.16 (s, 2H), 6.17 (d, J = 9.2 Hz, 1H), 7.32-7.36 (m, 6H), 7.96-7.98 (m, 1H), 8.75 (d, J = 2.0 Hz, 1H). Chemical Formula: C26H32N4O4; Molecular Weight: 464.56.
[0825] Step 5: Synthesis of 2-(4-(2-(5-(methoxycarbonyl)pyridin-2-yl)-2-azaspiro[3.3]heptan-6- yl)piperazin-1-yl)acetic acid.
Figure imgf000429_0002
[0826] A mixture of methyl 6-(6-(4-(2-(benzyloxy)-2-oxoethyl)piperazin-1-yl)-2- azaspiro[3.3]heptan-2-yl)nicotinate (120 mg, 0.26 mmol) and palladium on carbon (10%, 10 mg) in methanol (10 ml) was stirred at 40°C for 2 hours under hydrogen atmosphere (hydrogen balloon). TLC showed the reaction was complete. Palladium on carbon was removed through filtration and washed with methanol (10 ml x 2). The filtrate was concentrated under reduced pressure to give 2-(4-(2-(5-(methoxycarbonyl)pyridin-2-yl)-2-azaspiro[3.3]heptan-6-yl)piperazin- 1-yl)acetic acid (70 mg, crude) as colorless oil which was used in next step without purification. LC_MS: (ES+): m/z 375.2 [M+H]+. tR = 0.975 min. Chemical Formula: C19H26N4O4; Molecular Weight: 374.43. [0827] Step 6: Synthesis of methyl 6-(6-(4-(2-((S)-1-((2S,4R)-4-hydroxy-2-((S)-1-(4-(4- methylthiazol-5-yl)phenyl)ethylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-ylamino)- 2-oxoethyl)piperazin-1-yl)-2-azaspiro[3.3]heptan-2-yl)nicotinate:
Figure imgf000430_0001
[0828] To a stirred solution of 2-(4-(2-(5-(methoxycarbonyl)pyridin-2-yl)-2-azaspiro[3.3]heptan- 6-yl)piperazin-1-yl)acetic acid (70 mg, crude), (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4- hydroxy-N-((S)-1-(4-(4-methylthiazol-5-yl)phenyl)ethyl)pyrrolidine-2-carboxamide
hydrochloride (83 mg, 0.18 mmol), and N-ethyl-N-isopropylpropan-2-amine (69 mg, 0.54 mmol) in anhydrous N,N-dimethylformamide (3 ml) was added HATU (2-(7-Aza-1H-benzotriazole-1- yl)-1,1,3,3-tetramethyluronium hexafluorophosphate) (102 mg, 0.27 mmol) at 0°C, the resulting mixture was allowed to warm to room temperature and stirred at room temperature for 20 mins. TLC showed the reaction was complete. The reaction mixture was partitioned between ethyl acetate (20 ml) and water (10 ml). The organic layer was collected, and the aqueous layer was extracted with ethyl acetate (15 ml x 2). The combined organic layers were washed with brine (10 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluted with 8 % methanol in dichloromethane) to afford methyl 6-(6-(4-(2-((S)-1-((2S,4R)-4-hydroxy-2-((S)-1-(4-(4- methylthiazol-5-yl)phenyl)ethylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-ylamino)- 2-oxoethyl)piperazin-1-yl)-2-azaspiro[3.3]heptan-2-yl)nicotinate (110 mg, yield 74%) as white solid. LC_MS: (ES+): m/z 801.4 [M+H]+. tR = 1.912 min. 1HNMR (400 MHz, CDCl3): ^ 1.09 (s, 9H), 1.50 (d, J = 7.2 Hz, 3H), 2.01-2.04 (m, 2H), 2.10-2.18 (m, 3H), 2.24 (t, J = 7.6 Hz, 1H), 2.40-2.44 (m, 3H), 2.55 (s, 3H), 2.60-2.62 (m, 3H), 2.74 (t, J = 7.8 Hz, 1H), 3.05 (d, J = 6.0 Hz, 1H), 3.58-3.62 (m, 1H), 3.88 (s, 3H), 4.03 (s, 2H), 4.13-4.17 (m, 3H), 4.23 (d, J = 11.6 Hz, 1H), 4.45-4.53 (m, 2H), 4.78 (t, J = 7.8 Hz, 1H), 5.10 (t, J = 7.2 Hz, 1H), 5.37 (t, J = 4.8 Hz, 1H), 6.20 (d, J = 8.8 Hz, 1H), 7.38-7.44 (m, 4H), 7.85 (d, J = 8.4 Hz, 1H), 7.98-8.01 (m, 1H), 8.69 (s, 1H), 8.77 (s, 1H). Chemical Formula: C42H56N8O6S; Molecular Weight: 801.01.
[0829] Step 7: Synthesis of (6-(6-(4-(2-((S)-1-((2S,4R)-4-hydroxy-2-((S)-1-(4-(4-methylthiazol- 5-yl)phenyl)ethylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-ylamino)-2- oxoethyl)piperazin-1-yl)-2-azaspiro[3.3]heptan-2-yl)nicotinic acid:
Figure imgf000431_0001
[0830] A mixture of methyl 6-(6-(4-(2-((S)-1-((2S,4R)-4-hydroxy-2-((S)-1-(4-(4-methylthiazol- 5-yl)phenyl)ethylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-ylamino)-2- oxoethyl)piperazin-1-yl)-2-azaspiro[3.3]heptan-2-yl)nicotinate (110 mg, crude) and lithium hydroxide monohydrate (30 mg, 0.55 mmol) in tetrahydrofuran (4 ml)-water (1 ml)-methanol (1 ml) was stirred at room temperature for 16 hours. TLC showed the reaction was complete. The mixture solution was acidified with diluted hydrochloride acid (3N) till pH 3-4, and extracted with dichloromethane (10 ml × 2). The combined organic layers were washed with brine (10 ml), dried over sodium sulfate and concentrated under reduced pressure to give the 6-(6-(4-(2-((S)-1- ((2S,4R)-4-hydroxy-2-((S)-1-(4-(4-methylthiazol-5-yl)phenyl)ethylcarbamoyl)pyrrolidin-1-yl)- 3,3-dimethyl-1-oxobutan-2-ylamino)-2-oxoethyl)piperazin-1-yl)-2-azaspiro[3.3]heptan-2- yl)nicotinic acid (100 mg, crude) as white solid which was used in next step without purification. LC_MS: (ES+): m/z 787.5 [M+H]+. tR = 1.773 min. Chemical Formula: C41H54N8O6S; Molecular Weight: 786.98.
[0831] Step 8: Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-6-(6-(4-(2-((S)-1-((2S,4R)-4-hydroxy-2-((S)-1-(4-(4-methylthiazol-5- yl)phenyl)ethylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-ylamino)-2- oxoethyl)piperazin-1-yl)-2-azaspiro[3.3]heptan-2-yl)nicotinamide.
Figure imgf000432_0001
[0832] To a stirred solution of 6-(6-(4-(2-((S)-1-((2S,4R)-4-hydroxy-2-((S)-1-(4-(4- methylthiazol-5-yl)phenyl)ethylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-ylamino)- 2-oxoethyl)piperazin-1-yl)-2-azaspiro[3.3]heptan-2-yl)nicotinic acid (100 mg, crude), 4-((1r,3r)- 3-amino-2,2,4,4-tetramethylcyclobutoxy)-2-chlorobenzonitrile hydrochloride (44 mg, 0.14 mmol), and N-ethyl-N-isopropylpropan-2-amine (72 mg, 0.56 mmol) in anhydrous N,N- dimethylformamide (3 ml) was added HATU (2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3- tetramethyluronium hexafluorophosphate) (79 mg, 0.21 mmol) at 0°C, the resulting mixture was allowed to warm to room temperature and stirred for 20 min. TLC showed the reaction was complete. The reaction mixture was partitioned between ethyl acetate (20 ml) and water (10 ml). The organic layer was collected, and the aqueous layer was extracted with ethyl acetate (15 ml x 2). The combined organic layers were washed with brine (10 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluted with 8 % methanol in dichloromethane) to afford N- ((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)-6-(6-(4-(2-((S)-1-((2S,4R)- 4-hydroxy-2-((S)-1-(4-(4-methylthiazol-5-yl)phenyl)ethylcarbamoyl)pyrrolidin-1-yl)-3,3- dimethyl-1-oxobutan-2-ylamino)-2-oxoethyl)piperazin-1-yl)-2-azaspiro[3.3]heptan-2- yl)nicotinamide (52.5 mg, yield 37%, two steps) as white solid. LC_MS: (ES+): m/z 1047.5
[M+H]+. tR = 2.385 min. 1HNMR (400 MHz, CD3OD): ^ 1.07 (s, 9H), 1.23 (s, 6H), 1.30 (s, 6H), 1.53-1.60 (m, 3H), 1.93-1.99 (m, 1H), 2.22-2.27 (m, 1H), 2.49-2.63 (m, 9H), 2.72-3.03 (m, 8H), 3.19-3.26 (m, 2H), 3.48-3.64 (m, 1H), 3.74-3.78 (m, 1H), 3.87 (d, J = 11.2 Hz, 1H), 4.09 (s, 2H), 4.15-4.19 (m, 3H), 4.31 (s, 1H), 4.46 (br, 1H), 4.59 (t, J = 8.4 Hz, 1H), 4.66 (s, 1H), 5.01-5.04 (m, 1H), 6.44 (d, J = 8.8 Hz, 1H), 6.99-7.02 (m, 1H), 7.14 (d, J = 2.0 Hz, 1H), 7.41-7.46 (m, 4H), 7.74 (d, J = 8.8 Hz, 1H), 7.97-8.00 (m, 1H), 8.54 (s, 1H), 8.90 (s, 1H). Chemical Formula: C56H71ClN10O6S; Molecular Weight: 1047.74.
[0833] Unless otherwise noted, Examples 810-812 were synthesized according to similar procedures described above for the synthesis of Example 809, by utilizing corresponding starting materials and reagents.
[0834] Table 23: Additional Exemplary Compounds
Figure imgf000433_0001
Figure imgf000434_0001
[0835] Synthesis of Example 813:
Figure imgf000435_0001
[0836] Step 1: Synthesis of (3S,5R)-benzyl 3,5-dimethylpiperazine-1-carboxylate.
Figure imgf000435_0002
[0837] To a stirred solution of (2S,6R)-2,6-dimethylpiperazine (32.7 g, 291.4 mmol) in dichloromethane (100 ml) was added benzyl (2,5-dioxopyrrolidin-1-yl) carbonate (11 g, 43.78 mmol) at 0°C. The reaction mixture was stirred at 0 °C for 1 hour. TLC showed the reaction was complete. The resulting reaction mixture was allowed to warm up to room temperature and partitioned between dichloromethane (50 ml) and water (60 ml); the organic layer was collected and the aqueous layer was extracted with ethyl acetate (30 ml). The combined organic layers were washed with brine (40 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluted with 10-50 % ethyl acetate in hexane) to afford (3S,5R)-benzyl 3,5-dimethylpiperazine-1- carboxylate (6 g, yield 55%) as colorless oil. 1H NMR (400 MHz, CDCl3): ^ 1.06 (d, J = 6.0 Hz, 6H), 2.32-2.46 (m, 2H), 2.78-2.80 (m, 2H), 3.95-4.10 (m, 2H), 5.14-5.15 (m, 2H), 7.29-7.38 (m, 5H). Chemical Formula: C14H20N2O2; Molecular Weight: 248.32.
[0838] Step 2: Synthesis of (3S,5R)-benzyl 4-(5-(methoxycarbonyl)pyridin-2-yl)-3,5- dimethylpiperazine-1-carboxylate.
Figure imgf000436_0001
[0839] To a stirred solution of (3S,5R)-benzyl 3,5-dimethylpiperazine-1-carboxylate (1.0 g, 4.0 mmol), sodium 2-methylpropan-2-olate (768.8 mg, 8.0 mmol) and methyl 6-bromonicotinate (951.7 mg, 4.4 mmol) in toluene (10 ml) was added tri-tert-butylphosphine tetrafluoroborate (115.6 mg, 0.4 mmol) and Pd2(dba)3(Tris(dibenzylideneacetone)dipalladium) (366.3 mg, 0.4 mmol) at room temperature under nitrogen atmosphere; the mixture was degassed with nitrogen three times. The reaction mixture was stirred at 100°C for 2 hours. TLC showed the reaction was complete. The resulting reaction mixture was cooled to room temperature and partitioned between ethyl acetate (40 ml) and water (40 ml); the organic layer was collected and the aqueous layer was extracted with ethyl acetate (30 ml x 2). The combined organic layers were washed with brine (40 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash chromatography (eluted with 10-30 % ethyl acetate in hexane) to afford (3S,5R)-benzyl 4-(5-(methoxycarbonyl)pyridin-2-yl)-3,5- dimethylpiperazine-1-carboxylate (250 mg, yield 16%) as yellow oil. 1H NMR (400 MHz, CDCl3): ^ 1.24 (s, 6H), 3.16 (s, 2H), 3.86 (s, 3H), 4.01-4.19 (m, 2H), 4.55 (br, 2H), 5.16-5.22 (m, 2H), 6.51 (d, J = 8.8 Hz, 1H), 7.33-7.39 (m, 5H).8.02-8.04 (m, 1H), 8.82 (d, J = 2.0 Hz, 1H). Chemical Formula: C21H25N3O4; Molecular Weight: 383.44. LC_MS: (ES+): m/z 384.5 [M+H]+. tR = 2.947 min.
[0840] Step 3: Synthesis of methyl 6-((2S,6R)-2,6-dimethylpiperazin-1-yl)nicotinate.
Figure imgf000436_0002
[0841] A mixture of palladium on carbon (10%, 50 mg) and (3S,5R)-benzyl 4-(5- (methoxycarbonyl)pyridin-2-yl)-3,5-dimethylpiperazine-1-carboxylate (250 mg, 0.65 mmol) in methanol (30 ml) was stirred at 40°C for 1 hour under hydrogen atmosphere (hydrogen balloon). TLC showed the reaction was complete. Palladium on carbon was removed through filtration and washed with methanol (10 ml x2). The combined filtrates were concentrated under reduced pressure to afford methyl 6-((2S,6R)-2,6-dimethylpiperazin-1-yl)nicotinate (170 mg, crude) as yellow oil which was used in next step directly without further purification. [0842] Step 4: Synthesis of methyl 6-((2R,6S)-4-(2-(2-(tert-butoxy)-2-oxoethoxy)ethyl)-2,6- dimethylpiperazin-1-yl)nicotinate.
Figure imgf000437_0001
[0843] A mixture of methyl 6-((2S,6R)-2,6-dimethylpiperazin-1-yl)nicotinate (162 mg, 0.65 mmol), tert-butyl 2-(2-(tosyloxy)ethoxy)acetate (257.7 mg, 0.78 mmol), triethylamine (131.3 mg, 1.3 mmol) and potassium iodide (10 mg, 0.06 mmol) in N,N-dimethylformamide (6 ml) was stirred at 50°C for 12 hours. TLC showed the reaction was complete. The reaction mixture was partitioned between ethyl acetate (30 ml) and water (20 ml). The organic layer was collected, and the aqueous layer was extracted with ethyl acetate (15 ml x 2). The combined organic layers were washed with brine (10 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by which was purified by silica gel flash chromatography (eluted with 0-30% ethyl acetate in hexane) to afford methyl 6-((2R,6S)-4-(2-(2- (tert-butoxy)-2-oxoethoxy)ethyl)-2,6-dimethylpiperazin-1-yl)nicotinate (250 mg, crude) as yellow oil which was used in next step directly without further purification.
[0844] Step 5: Synthesis of methyl 6-((2R,6S)-4-(2-(2-(((2S)-1-((4R)-4-hydroxy-2-(((S)-1-(4- (4-methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2- yl)amino)-2-oxoethoxy)ethyl)-2,6-dimethylpiperazin-1-yl)nicotinate.
Figure imgf000437_0002
[0845] A mixture of m methyl 6-((2R,6S)-4-(2-(2-(tert-butoxy)-2-oxoethoxy)ethyl)-2,6- dimethylpiperazin-1-yl)nicotinate (200 mg, 0.49 mmol) and 2,2,2-trifluoroacetic acid (2 ml) in dichloromethane (3 ml) was stirred at room temperature for 2 hours. TLC showed the reaction was complete. The volatiles were evaporated under reduced pressure. The residue was taken up in dry N,N-dimethylformamide (2 ml), followed by sequential addition of N-ethyl-N- isopropylpropan-2-amine (127.4 mg, 0.98 mmol), (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)- 4-hydroxy-N-((S)-1-(4-(4-methylthiazol-5-yl)phenyl)ethyl)pyrrolidine-2-carboxamide hydrochloride (235 mg, 0.49 mmol), and HATU (2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3- tetramethyluronium hexafluorophosphate) (279.5 mg, 0.74 mmol) at 0°C, the resulting mixture was allowed to warm up to room temperature and stirred for 10 minutes. TLC showed the reaction was complete. The mixture was partitioned between ethyl acetate (10 ml) and water (10 ml). The organic layers was collected, washed with brine (10 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by silica gel flash column chromatography (eluted with 0-5% methanol in dichloromethane) to afford methyl 6-((2R,6S)-4-(2-(2-(((2S)-1-((4R)-4-hydroxy-2-(((S)-1-(4-(4-methylthiazol-5- yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)amino)-2- oxoethoxy)ethyl)-2,6-dimethylpiperazin-1-yl)nicotinate (180 mg, yield 47%) as yellow solid. 1H NMR (400 MHz, CD3OD): ^ 1.03-1.09 (m, 9H), 1.35-1.38 (m, 6H), 1.51-1.61 (m, 3H), 1.95-2.02 (m, 1H), 2.20-2.25 (m, 1H), 2.36-2.39 (m, 2H), 2.50 (s, 3H), 2.72 (t, J = 5.6 Hz, 2H), 2.91-3.02 (m, 2H), 3.72-3.82 (m, 3H), 3.83-4.03 (m, 4H), 4.05-4.17 (m, 2H), 4.37-4.56 (m, 3H), 4.57-4.62 (m, 1H), 4.72 (s, 1H), 5.00-5.03 (m, 1H), 6.69-6.73 (m, 1H), 7.40-7.48 (m, 4H), 8.00-8.04 (m, 1H), 8.72 (d, J = 2.4 Hz, 1H), 8.89 (s, 1H). Chemical Formula: C40H55N7O7S; Molecular Weight: 777.97. LC_MS: (ES+): m/z 778.4 [M+H]+. tR = 2.155 min.
[0846] Step 6: Synthesis of [N-((1r,3R)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-6-((2R,6S)-4-(2-(2-(((2S)-1-((4R)-4-hydroxy-2-(((S)-1-(4-(4- methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2- yl)amino)-2-oxoethoxy)eth l -26-dimeth l i erazin-1- l nicotinamide.
Figure imgf000438_0001
[0847] A mixture of methyl 6-((2R,6S)-4-(2-(2-(((2S)-1-((4R)-4-hydroxy-2-(((S)-1-(4-(4- methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2- yl)amino)-2-oxoethoxy)ethyl)-2,6-dimethylpiperazin-1-yl)nicotinate (170 mg, 0.22 mmol) and lithium hydroxide monohydrate (36.7 mg, 0.88 mmol) in tetrahydrofuran (4 ml)-water (1 ml)- methanol (1 ml) was stirred at 40°C for 3 hours. TLC showed the reaction was complete. The reaction mixture was acidified with diluted hydrochloride acid (3N) till pH 3-4 and evaporated under reduced pressure. The residue was taken up in dry N,N-dimethylformamide (2 ml), followed by sequential addition of N-ethyl-N-isopropylpropan-2-amine (46.7 mg, 0.33 mmol), 4- ((1r,3r)-3-amino-2,2,4,4-tetramethylcyclobutoxy)-2-chlorobenzonitrile hydrochloride (69.4 mg, 0.22 mmol), and HATU (2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium
hexafluorophosphate) (92.5 mg, 0.33 mmol) at 0°C, the resulting mixture was allowed to warm up to room temperature and stirred at room temperature for 10 minutes. TLC showed the reaction was complete. The mixture was partitioned between ethyl acetate (20 ml) and water (10 ml). The organic layer was collected, washed with brine (20 ml), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude residue which was purified by preparative TLC (eluted with 10% methanol in dichloromethane) to afford N-((1r,3R)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)-6-((2R,6S)-4-(2-(2-(((2S)-1-((4R)-4-hydroxy-2- (((S)-1-(4-(4-methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1- oxobutan-2-yl)amino)-2-oxoethoxy)ethyl)-2,6-dimethylpiperazin-1-yl)nicotinamide (100 mg, 44%) as white solid. 1H NMR (400 MHz, CD3OD): ^ 0.94, 0.95 (two singles, 9H), 1.12 (s, 6H), 1.18 (s, 6H), 1.21-1.29 (m, 6H), 1.40-1.48 (m, 3H), 1.87 (s, 1H), 2.07-2.15 (m, 1H), 2.23-2.31 (m, 2H), 2.38 (s, 3H), 2.60-2.62 (m, 2H), 2.85 (d, J = 12.8 Hz, 2H), 3.60-3.79 (m, 4H), 3.93-4.06 (m, 3H), 4.18 (s, 1H), 4.35-4.39 (m, 3H), 4.45-4.52 (m, 1H), 4.60 (s, 1H), 4.88-4.93 (m, 1H), 6.59- 6.63 (m, 1H), 6.88 (dd, J = 8.8, 2.4 Hz, 1H), 7.03 (d, J = 2.4 Hz, 1H), 7.26-7.36 (m, 4H), 7.49 (d, J = 8.8 Hz, 1H), 7.62 (d, J = 8.8 Hz, 1H), 7.82-7.86 (m, 1H), 8.47-8.50 (m, 1H), 8.52 (d, J = 2.4 Hz, 1H), 8.77 (s, 1H). Chemical Formula: C54H70ClN9O7S; Molecular Weight: 1024.71.
LC_MS: (ES+): m/z 1024.5 [M+H]+. tR = 2.548 min.
[0848] Unless otherwise noted, Examples 814-824 were synthesized according to similar procedures described above for the synthesis of Example 813, by utilizing corresponding starting materials and reagents.
[0849] Table 24: Exemplary Compounds.
Figure imgf000439_0001
Figure imgf000440_0001
Figure imgf000441_0001
Figure imgf000442_0001
Figure imgf000443_0001
[0850] Synthesis of Example 825.
Figure imgf000444_0001
[0851] Step 1: Synthesis of tert-butyl 4-[[4-(5-[[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl carbamo l ridin-2- l i erazin-1- l meth l i eridine-1-carboxylate.
Figure imgf000444_0002
[0852] Into a 20-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 6-(piperazin-1-yl)-N-[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]pyridine-3-carboxamide (260.0 mg, 0.56 mmol, 1.00 equiv), tert-butyl 4- (bromomethyl)piperidine-1-carboxylate (233.0 mg, 0.84 mmol, 1.50 equiv), CH3CN (5 mL), potassium carbonate (230.0 mg, 1.66 mmol, 3.00 equiv), NaI (89.0 mg, 1.00 equiv). The resulting solution was stirred for 24 hours at 100°C. The resulting solution was extracted with ethyl acetate and washed with water. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:1). The collected fractions were combined and concentrated under vacuum. This resulted in 218.0 mg (59%) of tert-butyl 4-[[4-(5-[[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperazin-1-yl]methyl]piperidine-1-carboxylate as a yellow solid. LC-MS (ES+): m/z 665.65 [(MH+], tR = 1.56 min, (1.9 minute run).
[0853] Step 2: Synthesis of 6-[4-(piperidin-4-ylmethyl)piperazin-1-yl]-N-[(1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetrameth lc clobut l ridine-3-carboxamide.
Figure imgf000445_0001
[0854] Into a 50-mL round-bottom flask, was placed tert-butyl 4-[[4-(5-[[(1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperazin-1- yl]methyl]piperidine-1-carboxylate (218.0 mg, 0.33 mmol, 1.00 equiv), dichloromethane (5 mL), trifluoroacetic acid (2 mL). The resulting solution was stirred for 1 h at room temperature. The resulting mixture was concentrated under vacuum. This resulted in 170.0 mg (92%) of 6-[4- (piperidin-4-ylmethyl)piperazin-1-yl]-N-[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]pyridine-3-carboxamide as yellow oil.
[0855] Step 3: Synthesis of tert-butyl 2-(4-[[4-(5-[[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobut l carbamo l ridin-2- l i erazin-1- l meth l i eridin-1-yl)acetate.
Figure imgf000445_0002
[0856] Into a 50-mL round-bottom flask, was placed 6-[4-(piperidin-4-ylmethyl)piperazin-1-yl]- N-[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyridine-3-carboxamide (170.0 mg, 0.30 mmol, 1.00 equiv), dichloromethane (5 mL), DIEA (155.0 mg, 1.20 mmol, 4.00 equiv), tert-butyl 2-bromoacetate (117.0 mg, 0.60 mmol, 2.00 equiv). The resulting solution was stirred for 1.5 h at room temperature. The resulting solution was extracted with dichloromethane and washed with water. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with dichloromethane/methanol (12:1). The collected fractions were combined and concentrated under vacuum. This resulted in 200.0 mg (98%) of tert-butyl 2-(4-[[4-(5-[[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperazin-1-yl]methyl]piperidin-1-yl)acetate as a yellow solid.
[0857] Step 4: Synthesis of 2-(4-[[4-(5-[[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperazin-1-yl]methyl]piperidin-1-yl)acetic acid.
Figure imgf000446_0001
[0858] Into a 100-mL round-bottom flask, was placed tert-butyl 2-(4-[[4-(5-[[(1r,3r)-3-(3-chloro- 4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperazin-1- yl]methyl]piperidin-1-yl)acetate (200.0 mg, 0.29 mmol, 1.00 equiv), dichloromethane (57.7 mL), trifluoroacetic acid (21.4 mL). The resulting solution was stirred for 1 overnight at room temperature. The resulting mixture was concentrated under vacuum. This resulted in 170.0 mg (93%) of 2-(4-[[4-(5-[[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperazin-1-yl]methyl]piperidin-1-yl)acetic acid as yellow oil.
[0859] Step 5: Synthesis of 6-(4-[[1-([[(2S)-1-[(2S,4R)-4-hydroxy-2-[[(1S)-1-[4-(4-methyl-1,3- thiazol-5-yl)phenyl]ethyl]carbamoyl]pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl]methyl)piperidin-4-yl]methyl]piperazin-1-yl)-N-[(1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyridine-3-carboxamide.
Figure imgf000446_0002
[0860] Into a 50-mL round-bottom flask, was placed 2-(4-[[4-(5-[[(1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperazin-1- yl]methyl]piperidin-1-yl)acetic acid (170.0 mg, 0.27 mmol, 1.00 equiv), (2S,4R)-1-[(2S)-2- amino-3,3-dimethylbutanoyl]-4-hydroxy-N-[(1S)-1-[4-(4-methyl-1,3-thiazol-5- yl)phenyl]ethyl]pyrrolidine-2-carboxamide hydrochloride (131.0 mg, 0.27 mmol, 1.00 equiv), N,N-dimethylformamide (5 mL), DIEA (169.0 mg, 1.31 mmol, 4.00 equiv), BoP (145.0 mg, 1.20 equiv). The resulting solution was stirred for 2 hours at room temperature. The reaction was then quenched by the water. The resulting solution was extracted with ethyl acetate and washed with water. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Shield RP18 OBD Column, 5um,19*150mm; mobile phase, Water(10MMOL/L NH4HCO3) and ACN (52.0% ACN up to 73.0% in 8 min); Detector, UV 254nm. This resulted in 63.0 mg (22%) of 6-(4-[[1-([[(2S)-1-[(2S,4R)-4-hydroxy-2-[[(1S)-1-[4-(4-methyl-1,3-thiazol-5- yl)phenyl]ethyl]carbamoyl]pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl]methyl)piperidin-4-yl]methyl]piperazin-1-yl)-N-[(1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyridine-3-carboxamide as a white solid. 1H-NMR (400 MHz, CD3OD ) ^8.89 (s, 1H), 8.62 (s, 1H), 7.99-7.96 (m, 1H), 7.75-7.73 (d, J=8.4Hz, 1H), 7.49-7.36 (m, 4H), 7.14 (s, 1H), 7.01-6.94 (m, 1H), 6.85-6.83 (m, 1H), 5.05-5.00 (m, 1H), 4.62 (s, 1H), 4.60-4.41 (m, 2H), 4.29 (s, 1H), 4.11 (s, 1H), 4.89-4.85 (m, 1H), 3.78-3.62 (m, 5H), 3.05- 3.01 (m, 2H), 2.94-2.84 (m, 2H), 2.58-2.52 (m, 4H), 2.43 (s, 3H), 2.20-2.18 (m, 5H), 2.00-1.81 (m, 3H), 1.70-1.50 (m, 4H), 1.38-1.30 (m, 2H), 1.28 (s, 6H), 1.22 (s, 6H), 1.05 (s, 9H); LC-MS (ES+): m/z 1050.05 [(MH+], tR = 1.95 min, (3.0 minute run. Chemical Formula:
C56H73ClN10O6S. Molecular Weight: 1048.51.
[0861] Synthesis of Example 829:
Figure imgf000448_0001
[0862] Step 1: Synthesis of benzyl 6-[4-(hydroxymethyl)piperidin-1-yl]pyridine-3-carboxylate.
Figure imgf000448_0002
[0863] Into a 100-mL round-bottom flask, was placed benzyl 6-chloropyridine-3-carboxylate (2.0 g, 8.08 mmol, 1.00 equiv), N,N-dimethylformamide (30 mL), piperidin-4-ylmethanol (927.4 mg, 8.05 mmol, 1.00 equiv), potassium carbonate (3.3 g, 23.88 mmol, 3.00 equiv). The resulting solution was stirred for 16 hours at 100°C in an oil bath. The resulting mixture was concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (v: v = 1: 1). This resulted in 1.72 g (65%) of benzyl 6-[4-(hydroxymethyl)piperidin-1- yl]pyridine-3-carboxylate as colorless oil. LC-MS (ES+): m/z 327.30 [MH+], tR =1.12 min (3.0 minute run).
[0864] Step 2: Synthesis of benzyl 6-[4-([[(4-methylbenzene)sulfonyl]oxy]methyl)piperidin-1- yl]pyridine-3-carboxylate.
Figure imgf000449_0001
[0865] Into a 100-mL round-bottom flask, was placed benzyl 6-[4-(hydroxymethyl)piperidin-1- yl]pyridine-3-carboxylate (1.7 g, 5.21 mmol, 1.00 equiv), dichloromethane (30 mL),
triethylamine (1.1 g, 10.87 mmol, 2.00 equiv), 4-methylbenzene-1-sulfonyl chloride (1.2 g, 6.29 mmol, 1.20 equiv), 4-dimethylaminopyridine (190.9 mg, 1.56 mmol, 0.30 equiv). The resulting solution was stirred for 16 hours at room temperature. The resulting mixture was concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (v: v = 1: 2). This resulted in 1.9 g (76%) of benzyl 6-[4-([[(4- methylbenzene)sulfonyl]oxy]methyl)piperidin-1-yl]pyridine-3-carboxylate as a white solid. LC-MS (ES+): m/z 481.35 [MH+], tR =2.74 min (5.0 minute run).
[0866] Step 3: Synthesis of benzyl 6-(3-(2-(4-(2-ethoxy-2-oxoethyl)piperazin-1- yl)ethyl)pyrrolidin-1-yl)ni in
Figure imgf000449_0002
[0867] Into a 100-mL round-bottom flask, was placed benzyl 6-[4-([[(4-methylbenzene) sulfonyl]oxy]methyl)piperidin-1-yl]pyridine-3-carboxylate (800.0 mg, 1.66 mmol, 1.00 equiv), N-methyl pyrrolidone (10 mL), potassium carbonate (688.6 mg, 4.98 mmol, 3.00 equiv), ethyl 2- (piperazin-1-yl)acetate (286.1 mg, 1.66 mmol, 1.00 equiv). The resulting solution was stirred for 16 hours at 120°CC in an oil bath. The reaction was then quenched by water (50 mL), extracted with ethyl acetate (50 mL x 3)^washed with water (50 mL) and brine (50 mL). The mixture was dried over anhydrous sodium sulfate. The solids were filtered out. The resulting mixture was concentrated under vacuum. The residue was applied onto a silica gel column with
dichloromethane/methanol (9/1). This resulted in 450.0 mg (56%) of benzyl 6-(3-(2-(4-(2- ethoxy-2-oxoethyl)piperazin-1-yl)ethyl)pyrrolidin-1-yl)nicotinate as yellow oil. LC-MS (ES+): m/z 481.05 [MH+], tR =2.54 min (4.6 minute run).
[0868] Step 4: Synthesis of 6-(3-(2-(4-(2-ethoxy-2-oxoethyl)piperazin-1-yl)ethyl)pyrrolidin-1- yl)nicotinic acid.
Figure imgf000450_0001
[0869] Into a 100-mL round-bottom flask, was placed benzyl 6-(3-(2-(4-(2-ethoxy-2- oxoethyl)piperazin-1-yl)ethyl)pyrrolidin-1-yl)nicotinate (450.0 mg, 0.94 mmol, 1.00 equiv), methanol (10 mL), palladium carbon (400.0 mg). The flask was then vacuumed and flushed with hydrogen. The reaction mixture was hydrogenated at room temperature for 24 hours under hydrogen atmosphere using a hydrogen balloon, then filtered through a Celite pad and concentrated under reduced pressure. The resulting mixture was concentrated under vacuum. This resulted in 320.0 mg (88%) of 6-(3-(2-(4-(2-ethoxy-2-oxoethyl)piperazin-1- yl)ethyl)pyrrolidin-1-yl)nicotinic acid as yellow oil. LC-MS (ES+): m/z 391.10 [MH+], tR =0.64 min (2.0 minute run).
[0870] Step 5: Synthesis of ethyl 2-(4-(2-(1-(5-(((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobut l carbamo l ridin-2- l rrolidin-3- l eth l i erazin-1-yl)acetate.
Figure imgf000450_0002
[0871] Into a 50-mL round-bottom flask, was placed 6-(3-(2-(4-(2-ethoxy-2-oxoethyl)piperazin- 1-yl)ethyl)pyrrolidin-1-yl)nicotinic acid (200.0 mg, 0.51 mmol, 1.00 equiv), N,N- dimethylformamide (5 mL), 2-chloro-4-[(1r,3r)-3-amino-2,2,4,4- tetramethylcyclobutoxy]benzonitrile hydrochloride (161.5 mg, 0.51 mmol, 1.00 equiv),
N,N,N,N-Tetramethyl-O-(7-azabenzotriazol-1-yl)uronium hexafluorophospate (292.3 mg, 0.77 mmol, 1.50 equiv), N,N-diisopropylethylamine (198.5 mg, 1.54 mmol, 3.00 equiv). The resulting solution was stirred for 1 hour at room temperature. The reaction was then quenched by water (30 mL), extracted with 3x30 mL of ethyl acetate (30 mL x 3), washed with water (30 mL) and brine (30 mL), dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with dichloromethane/methanol (v: v = 9: 1). This resulted in 300.0 mg (90%) of ethyl 2-(4-(2-(1-(5-(((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)pyrrolidin-3-yl)ethyl)piperazin-1-yl)acetate as yellow oil. LC-MS (ES+): m/z 651.4 [MH+], tR =1.09 min (2.0 minute run).
[0872] Step 6: Synthesis of 2-(4-(2-(1-(5-(((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)pyrrolidin-3-yl)ethyl)piperazin-1-yl)acetic acid.
Figure imgf000451_0001
[0873] Into a 50-mL round-bottom flask, was placed ethyl 2-(4-(2-(1-(5-(((1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)pyrrolidin-3- yl)ethyl)piperazin-1-yl) (300.0 mg, 0.46 mmol, 1.00 equiv), methanol (10 mL), water (3 mL), lithium hydroxide (110.6 mg, 4.62 mmol, 10.00 equiv). The resulting solution was stirred for 5 h at room temperature. The pH value of the solution was adjusted to 5-6 with 1 mol/L hydrogen chloride, concentrated under vacuum. The solids were filtered out. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Shield RP18 OBD Column, 5um,19*150mm; mobile phase, water(10 mmol/L ammonium bicarbonate) and acetonitrile (30.0% acetonitrile up to 55.0% in 8 min); Detector, UV 254nm. This resulted in 150.0 mg (52%) of 2-(4-(2-(1-(5-(((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)pyrrolidin-3-yl)ethyl)piperazin-1-yl)acetic acid as colorless oil. LC-MS (ES+): m/z 623.60 [MH+], tR =0.97 min (1.9 minute run).
[0874] Step 7: Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-6-(3-(2-(4-(2-(((S)-1-((2S,4R)-4-hydroxy-2-(((S)-1-(4-(4-methylthiazol- 5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)amino)-2- oxoethyl)piperazin-1-yl)ethyl)pyrrolidin-1-yl)nicotinamide.
Figure imgf000452_0001
[0875] Into a 25-mL round-bottom flask, was placed 2-(4-(2-(1-(5-(((1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)pyrrolidin-3- yl)ethyl)piperazin-1-yl)acetic acid (140.0 mg, 0.22 mmol, 1.00 equiv), N,N-dimethylformamide (2 mL), (2S,4R)-1-[(2S)-2-amino-3,3-dimethylbutanoyl]-4-hydroxy-N-[(1S)-1-[4-(4-methyl-1,3- thiazol-5-yl)phenyl]ethyl]pyrrolidine-2-carboxamide hydrochloride (108.1 mg, 0.22 mmol, 1.00 equiv), N,N,N,N-Tetramethyl-O-(7-azabenzotriazol-1-yl)uronium hexafluorophospate (128.1 mg, 0.34 mmol, 1.50 equiv), N,N-diisopropylethylamine (87.0 mg, 0.67 mmol, 3.00 equiv). The resulting solution was stirred for 1 hour at room temperature. T The reaction was then quenched by water (20 mL), extracted with ethyl acetate (20 mL x 3) and concentrated under vacuum. The solids were filtered out. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Shield RP18 OBD Column, 5um, 19*150mm; mobile phase, water (10mmol/L ammonium bicarbonate) and acetonitrile (48.0% acetonitrile up to 63.0% in 8 min); Detector, UV 254nm. This resulted in 76.6 mg (32%) of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)- 2,2,4,4-tetramethylcyclobutyl)-6-(3-(2-(4-(2-(((S)-1-((2S,4R)-4-hydroxy-2-(((R)-2-hydroxy-1-(4- (4-methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2- yl)amino)-2-oxoethyl)piperazin-1-yl)ethyl)pyrrolidin-1-yl)nicotinamide as a white solid. 1H NMR (300 MHz, CD3OD): ^ 8.56 (s, 1H), 8.55 (s, 1H), 7.95-7.92 (m, 1H), 7.72 (d, J = 8.7 Hz, 1H), 7.51-7.37 (m, 4H), 7.13-7.12 (m, 1H), 7.00-6.96 (m, 1H), 6.53 (d, J = 8.7 Hz, 1H), 5.02-5.00 (m, 1H), 4.67-4.36 (m, 3H), 4.28 (s, 1H), 4.14 (s, 1H), 3.90-3.55 (m, 4H), 3.50-3.40 (m, 1H), 3.20-3.08 (m, 3H), 2.75-2.44 (m, 13H), 2.40-2.17 (m, 3H), 2.00-1.90 (m, 1H), 1.80-1.68 (m, 3H), 1.61-1.48 (m, 3H), 1.28-1.22 (m, 12H), 1.05 (s, 9H); LC-MS (ES+): m/z 1050.50 [MH+]; HPLC: tR =9.78 min (15.0 minute run). ^^^^^^^^^^^^^^^^^^C56H73ClN10O6S. Molecular Weight: 1048.51.
[0876] Unless otherwise noted, Examples 826-832 were synthesized according to similar procedures described above for the synthesis of Examples 825 and 829, by utilizing corresponding starting materials and reagents.
[0877] Table 25: Additional Exemplary Compounds
Figure imgf000453_0001
Figure imgf000454_0001
Figure imgf000455_0001
[0878] Synthesis of Example 833:
Figure imgf000456_0001
Figure imgf000456_0002
[0879] Step 1: Synthesis of benzyl 6-[4-(2-hydroxyethyl)piperidin-1-yl]pyridine-3-carboxylate.
Figure imgf000456_0003
[0880] Into a 100-mL round-bottom flask, was placed benzyl 6-chloropyridine-3-carboxylate (1.0 g, 4.04 mmol, 1.00 equiv), 2-(piperidin-4-yl)ethan-1-ol (520.0 mg, 4.02 mmol, 1.00 equiv), N,N-dimethylformamide (15 mL), potassium carbonate (1.7 g, 12.30 mmol, 3.00 equiv). The resulting solution was stirred for 4 hours at 100°C. The resulting solution was extracted with ethyl acetate and washed with water. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:1). The collected fractions were combined and concentrated under vacuum. This resulted in 1.0 g (73%) of benzyl 6-[4-(2-hydroxyethyl)piperidin-1-yl]pyridine-3- carboxylate as a yellow solid.
[0881] Step 2: Synthesis of benzyl 6-[4-(2-[[(4-methylbenzene)sulfonyl]oxy]ethyl)piperidin-1- yl]pyridine-3-carboxylate.
Figure imgf000457_0001
[0882] Into a 100-mL round-bottom flask, was placed benzyl 6-[4-(2-hydroxyethyl)piperidin-1- yl]pyridine-3-carboxylate (500.0 mg, 1.47 mmol, 1.00 equiv), dichloromethane (10 mL), TsCl (419.0 mg, 2.20 mmol, 1.50 equiv), TEA (446.0 mg, 4.41 mmol, 2.00 equiv), 4- dimethylaminopyridine (54.0 mg, 0.44 mmol, 0.20 equiv). The resulting solution was stirred for 4 hours at room temperature. The resulting solution was extracted with dichloromethane and washed with water. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:2). The collected fractions were combined and concentrated under vacuum. This resulted in 660.0 mg (91%) of benzyl 6-[4-(2-[[(4-methylbenzene)sulfonyl]oxy]ethyl)piperidin-1- yl]pyridine-3-carboxylate as a yellow solid.
[0883] Step 3: Synthesis of benzyl 6-(4-[2-[4-(2-ethoxy-2-oxoethyl)piperazin-1- yl]ethyl]piperidin-1-yl)pyridine-3-carboxylate.
Figure imgf000457_0002
[0884] Into a 100-mL round-bottom flask, was placed benzyl 6-[4-(2-[[(4- methylbenzene)sulfonyl]oxy]ethyl)piperidin-1-yl]pyridine-3-carboxylate (660.0 mg, 1.33 mmol, 1.00 equiv), ethyl 2-(piperazin-1-yl)acetate hydrochloride (278.8 mg, 1.34 mmol, 1.00 equiv), CH3CN (10 mL), potassium carbonate (553.0 mg, 4.00 mmol, 3.00 equiv), NaI (200.0 mg, 1.00 equiv). The resulting solution was stirred for 3 hours at 100°C. The resulting solution was extracted with ethyl acetate and washed with water. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with dichloromethane/methanol (15:1). The collected fractions were combined and concentrated under vacuum. This resulted in 467.0 mg (71%) of benzyl 6-(4-[2-[4-(2-ethoxy-2- oxoethyl)piperazin-1-yl]ethyl]piperidin-1-yl)pyridine-3-carboxylate as yellow oil. LC-MS (ES+): m/z 495.30 [(MH+], tR = 0.71 min, (1.9 minute run). [0885] Step 4: Synthesis of 6-(4-[2-[4-(2-ethoxy-2-oxoethyl)piperazin-1-yl]ethyl]piperidin-1- yl)pyridine-3-carboxylic acid.
Figure imgf000458_0001
[0886] Into a 50-mL round-bottom flask, was placed benzyl 6-(4-[2-[4-(2-ethoxy-2- oxoethyl)piperazin-1-yl]ethyl]piperidin-1-yl)pyridine-3-carboxylate (467.0 mg, 0.94 mmol, 1.00 equiv), ethanol (10 mL), Palladium carbon (90.0 mg) was added under nitrogen atmosphere, the flask was then vacuumed and flushed with hydrogen. The reaction mixture was hydrogenated at 35°C for 1 overnight under hydrogen atmosphere using a hydrogen balloon, then filtered through a Celite pad and concentrated under reduced pressure. This resulted in 380.0 mg (99%) of 6-(4-[2- [4-(2-ethoxy-2-oxoethyl)piperazin-1-yl]ethyl]piperidin-1-yl)pyridine-3-carboxylic acid as a yellow solid.
[0887] Step 5: Synthesis of ethyl 2-(4-[2-[1-(5-[[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperidin-4-yl]ethyl]piperazin-1-yl)acetate.
Figure imgf000458_0002
[0888] Into a 50-mL round-bottom flask, was placed 6-(4-[2-[4-(2-ethoxy-2-oxoethyl)piperazin- 1-yl]ethyl]piperidin-1-yl)pyridine-3-carboxylic acid (180.0 mg, 0.44 mmol, 1.00 equiv), 2-chloro- 4-[(1r,3r)-3-amino-2,2,4,4-tetramethylcyclobutoxy]benzonitrile hydrochloride (140.0 mg, 0.44 mmol, 1.00 equiv), N,N-dimethylformamide (5 mL), DIEA (230.0 mg, 1.78 mmol, 4.00 equiv), BoP (237.0 mg, 1.20 equiv). The resulting solution was stirred for 2 hours at room temperature. The reaction was then quenched by the water. The resulting solution was extracted with ethyl acetate and washed with water. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with
dichloromethane/methanol (12:1). The collected fractions were combined and concentrated under vacuum. This resulted in 230.0 mg (78%) of ethyl 2-(4-[2-[1-(5-[[(1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperidin-4- yl]ethyl]piperazin-1-yl)acetate as yellow oil. [0889] Step 6: Synthesis of 2-(4-[2-[1-(5-[[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperidin-4-yl]ethyl]piperazin-1-yl)acetic acid.
Figure imgf000459_0001
[0890] Into a 50-mL round-bottom flask, was placed ethyl 2-(4-[2-[1-(5-[[(1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperidin-4- yl]ethyl]piperazin-1-yl)acetate (125.0 mg, 0.19 mmol, 1.00 equiv), ethanol (3 mL), LiOH (43.0 mg, 1.80 mmol, 10.00 equiv), water(1 mL). The resulting solution was stirred for 1 overnight at 25°C. The resulting mixture was concentrated under vacuum. This resulted in 110.0 mg (92%) of 2-(4-[2-[1-(5-[[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperidin-4-yl]ethyl]piperazin-1-yl)acetic acid as yellow oil.
[0891] Step 7: Synthesis of 6-(4-[2-[4-([[(2S)-1-[(2S,4R)-4-hydroxy-2-[[(1S)-1-[4-(4-methyl- 1,3-thiazol-5-yl)phenyl]ethyl]carbamoyl]pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl]methyl)piperazin-1-yl]ethyl]piperidin-1-yl)-N-[(1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyridine-3-carboxamide.
Figure imgf000459_0002
[0892] Into a 50-mL round-bottom flask, was placed 2-(4-[2-[1-(5-[[(1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperidin-4- yl]ethyl]piperazin-1-yl)acetic acid (110.0 mg, 0.17 mmol, 1.00 equiv), (2S,4R)-1-[(2S)-2-amino- 3,3-dimethylbutanoyl]-4-hydroxy-N-[(1S)-1-[4-(4-methyl-1,3-thiazol-5- yl)phenyl]ethyl]pyrrolidine-2-carboxamide hydrochloride (83.0 mg, 0.17 mmol, 1.00 equiv), N,N-dimethylformamide (5 mL), DIEA (89.0 mg, 0.69 mmol, 4.00 equiv), BoP (92.0 mg, 1.20 equiv). The resulting solution was stirred for 2 hours at room temperature. The reaction was then quenched by the water. The resulting solution was extracted with ethyl acetate and washed with water. The mixture was dried over anhydrous sodium sulfate. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Shield RP18 OBD Column, 5um,19*150mm; mobile phase, Water(10MMOL/L NH4HCO3) and ACN (55.0% ACN up to 71.0% in 8 min); Detector, UV 254nm. This resulted in 80.0 mg (44%) of 6-(4-[2-[4-([[(2S)-1- [(2S,4R)-4-hydroxy-2-[[(1S)-1-[4-(4-methyl-1,3-thiazol-5-yl)phenyl]ethyl]carbamoyl]pyrrolidin- 1-yl]-3,3-dimethyl-1-oxobutan-2-yl]carbamoyl]methyl)piperazin-1-yl]ethyl]piperidin-1-yl)-N- [(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyridine-3-carboxamide as a white solid. H-NMR (400 MHz, CD3OD ) ^8.88 (s, 1H), 8.59 (s, 1H), 7.96-7.93 (m, 1H), 7.74- 7.72 (m, 1H), 7.47-7.36 (m, 4H), 7.14 (s, 1H), 7.00-6.98 (m, 1H), 6.85-6.83 (m, 1H), 5.05-5.00 (m, 1H), 4.62 -4.41 (m, 5H), 4.27 (s, 1H), 4.12 (s, 1H), 3.87-3.72 (m, 2H), 3.05-3.02 (m, 2H), 2.96-2.89 (m, 2H), 2.62-2.45 (s, 13H), 2.22-2.16 (m, 1H), 2.00-1.81 (m, 3H), 1.60-1.47 (m, 6H), 1.27-1.21 (m, 14H), 1.04 (s, 9H); LC-MS (ES+): m/z 1063.60 [(MH+], tR = 2.87 min, (5.6 minute run). Chemical Formula: C57H75ClN10O6S. Molecular Weight 1062.53.
[0893] Unless otherwise noted, Examples 834-837 were synthesized according to similar procedures described above for the synthesis of Example 833, by utilizing corresponding starting materials and reagents.
[0894] Table 26: Exemplary Compounds
Figure imgf000460_0001
Figure imgf000461_0001
Figure imgf000462_0001
[0895] Synthesis of Example 838:
Figure imgf000463_0001
[0896] Step 1: Synthesis of 3-(benzyloxy)propanal.
Figure imgf000463_0002
[0897] Into a 500-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 3-(benzyloxy)propan-1-ol (11.62 g, 69.91 mmol, 1.00 equiv), dichloromethane (250 mL). This was followed by the addition of DMP (32.65 g, 76.98 mmol, 1.10 equiv), in portions at 0°C. The resulting solution was stirred for 3 hours at 25°C. The resulting mixture was washed with 2x200 mL of Na2S2O3. The resulting mixture was washed with 1x200 mL of sodium bicarbonate. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1/3). This resulted in 10 g (87%) of 3-(benzyloxy)propanal as colorless oil.
[0898] Step 2: Synthesis of (E)-N-[3-(benzyloxy)propylidene]hydroxylamine.
Figure imgf000464_0004
[0899] Into a 250-mL round-bottom flask, was placed NH2OH.HCl (12.51 g, 3.00 equiv), H2O/MeOH (90/30 mL), NaOAc (14.76 g, 3.00 equiv). This was followed by the addition of a solution of 3-(benzyloxy)propanal (9.84 g, 59.93 mmol, 1.00 equiv) in methanol (10 mL) dropwise with stirring at 0°C in 5 minutes. The resulting solution was stirred for 5 hours at 25°C. The resulting mixture was concentrated under vacuum. The resulting solution was extracted with 2x150 mL of dichloromethane and the organic layers combined. The resulting mixture was washed with 1x150 mL of brine. The mixture was dried over anhydrous sodium sulfate. The resulting mixture was concentrated under vacuum.This resulted in 10 g (93%) of (E)-N-[3- (benzyloxy)propylidene]hydroxylamine as light yellow oil.
[0900] Step 3: Synthesis of (Z)-3-(benzyloxy)-N-hydroxypropcarbonimidoyl chloride.
Figure imgf000464_0001
[0901] Into a 250-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed (E)-N-[3-(benzyloxy)propylidene]hydroxylamine (9 g, 50.22 mmol, 1.00 equiv), N,N-dimethylformamide (60 mL), NCS (8.04 g, 60.21 mmol, 1.20 equiv). The resulting solution was stirred for 3 hours at 25oC.The reaction mixture was used to next step without purification.
[0902] Step 4: Synthesis of (Z)-3-(benzyloxy)-N-hydroxypropcarbonimidoyl chloride.
Figure imgf000464_0002
[0903] Into a 250-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed (E)-N-[3-(benzyloxy)propylidene]hydroxylamine (9 g, 50.22 mmol, 1.00 equiv), N,N-dimethylformamide (60 mL), NCS (8.04 g, 60.21 mmol, 1.20 equiv). The resulting solution was stirred for 3 hours at 25oC.The reaction mixture was used to next step without purification.
[0904] Step 5: Synthesis of 2-[3-[2-(benzyloxy)ethyl]-1,2-oxazol-5-yl]acetic acid.
Figure imgf000464_0003
[0905] Into a 500-mL round-bottom flask, was placed 2-[3-[2-(benzyloxy)ethyl]-1,2-oxazol-5- yl]ethan-1-ol (6.48 g, 26.20 mmol, 1.00 equiv), acetone (120 mL), Jones reagent (8 g CrO3/80 ml H2O/12 ml H2SO4). The resulting solution was stirred for 3 hours at 25oC. The reaction was then quenched by the addition of 80 mL of. The resulting mixture was concentrated under vacuum. The resulting solution was diluted with 50 mL of water. The resulting solution was extracted with 2x100 mL of dichloromethane and the organic layers combined and dried over anhydrous sodium sulfate. This resulted in 6 g crude product(88%) of 2-[3-[2-(benzyloxy)ethyl]-1,2-oxazol- 5-yl]acetic acid as yellow oil.
[0906] Step 6: Synthesis of ethyl 2-[3-[2-(benzyloxy)ethyl]-1,2-oxazol-5-yl]acetate.
Figure imgf000465_0001
[0907] Into a 250-mL round-bottom flask, was placed 2-[3-[2-(benzyloxy)ethyl]-1,2-oxazol-5- yl]acetic acid (5.22 g, 19.98 mmol, 1.00 equiv), ethanol (100 mL), sulfuric acid (2 mL). The resulting solution was stirred for 2 hours at 70oC. The resulting mixture was concentrated under vacuum. The pH value of the solution was adjusted to 8 with sodium carbonate. The resulting solution was extracted with 2x50 mL of ethyl acetate and the organic layers combined and dried over anhydrous sodium sulfate. The resulting mixture was concentrated under vacuum.The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1/10). This resulted in 2.5 g (43%) of ethyl 2-[3-[2-(benzyloxy)ethyl]-1,2-oxazol-5-yl]acetate as colorless oil. LC-MS (ES+): m/z 290.00 [MH+], tR = 1.40 min, (2.70 minute run).
[0908] Step 7: Synthesis of ethyl 2-[3-[2-(benzyloxy)ethyl]-1,2-oxazol-5-yl]-3-methylbutanoate.
Figure imgf000465_0002
[0909] Into a 25-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed ethyl 2-[3-[2-(benzyloxy)ethyl]-1,2-oxazol-5-yl]acetate (347 mg, 1.20 mmol, 1.00 equiv), tetrahydrofuran (15 mL). This was followed by the addition of t-BuOK (1M in THF)(1.44 mL, 1.20 equiv) dropwise with stirring at 0oC. To this was added 2-iodopropane (245 mg, 1.44 mmol, 1.20 equiv) dropwise with stirring at 0oC. The resulting solution was stirred for 10 min at 0oC. The resulting solution was allowed to react, with stirring, for an additional 3 hours at 25oC. The reaction was then quenched by the addition of 10 mL of NH4Cl. The resulting solution was extracted with 2x15 mL of ethyl acetate and the organic layers combined and dried over anhydrous sodium sulfate. The resulting mixture was concentrated under vacuum. This resulted in 0.22 g (55%) of ethyl 2-[3-[2-(benzyloxy)ethyl]-1,2-oxazol-5-yl]-3- methylbutanoate as colorless oil.
[0910] Step 8: Synthesis of ethyl 2-[3-(2-hydroxyethyl)-1,2-oxazol-5-yl]-3-methylbutanoate.
Figure imgf000466_0001
[0911] Into a 25-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed ethyl 2-[3-[2-(benzyloxy)ethyl]-1,2-oxazol-5-yl]-3-methylbutanoate (199 mg, 0.60 mmol, 1.00 equiv), dichloromethane (10 mL). This was followed by the addition of BBr3 (1.2 mL, 2.00 equiv) dropwise with stirring at -78oC. The resulting solution was stirred for 20 minutes at -78oC. The reaction was then quenched by the addition of water. The resulting solution was extracted with 2x10 mL of dichloromethane and the organic layers combined and dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1/1). This resulted in 0.13 g (90%) of ethyl 2-[3-(2-hydroxyethyl)-1,2-oxazol-5-yl]-3-methylbutanoate as colorless oil.
[0912] Step 9: Synthesis of ethyl 3-methyl-2-[3-(2-[[(4-methylbenzene)sulfonyl]oxy]ethyl)-1,2- oxazol-5-yl]butanoate.
Figure imgf000466_0002
[0913] Into a 25-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed ethyl 2-[3-(2-hydroxyethyl)-1,2-oxazol-5-yl]-3-methylbutanoate (121 mg, 0.50 mmol, 1.00 equiv), dichloromethane (10 mL), triethylamine (152 mg, 1.50 mmol, 3.00 equiv), TsCl (238 mg, 1.25 mmol, 2.50 equiv), 4-dimethylaminopyridine (12 mg, 0.10 mmol, 0.20 equiv). The resulting solution was stirred for 12 hours at 25oC. The reaction was then quenched by the addition of 10 mL of water. The resulting solution was extracted with 2x15 mL of dichloromethane and the organic layers combined. The resulting mixture was washed with 1x15 mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1/3). This resulted in 0.123 g (62%) of ethyl 3-methyl-2-[3-(2-[[(4- methylbenzene)sulfonyl]oxy]ethyl)-1,2-oxazol-5-yl]butanoate as a white solid.
[0914] Step 10: Synthesis of ethyl 3-methyl-2-(3-[2-[4-(5-[[(1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperazin-1-yl]ethyl]-1,2- oxazol-5-yl)b
Figure imgf000467_0001
[0915] Into a 15-mL sealed tube purged and maintained with an inert atmosphere of nitrogen, was placed. This was followed by the addition of CH3CN (6 mL), 6-(piperazin-1-yl)-N-[(1r,3r)- 3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyridine-3-carboxamide (152 mg, 0.32 mmol, 1.20 equiv), potassium carbonate (112 mg, 0.81 mmol, 3.00 equiv), The resulting solution was stirred for 2 min at 25oC.Ethyl 3-methyl-2-[3-(2-[[(4- methylbenzene)sulfonyl]oxy]ethyl)-1,2-oxazol-5-yl]butanoate (107 mg, 0.27 mmol, 1.00 equiv) and NaI (0.041 g, 1.00 equiv)was added. The resulting solution was allowed to react, with stirring, for an additional 12 hours at 60oC. The reaction mixture was cooled to 25°C. The solids were filtered out. The resulting mixture was concentrated under vacuum. The residue was applied onto a silica gel column with dichloromethane/methanol (10/1). This resulted in 0.17 g (91%) of ethyl 3-methyl-2-(3-[2-[4-(5-[[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperazin-1-yl]ethyl]-1,2-oxazol-5-yl)butanoate as a white solid. LC-MS (ES+): m/z 691.35 [MH+], tR = 1.02 min, (1.90 minute run).
[0916] Step 11: Synthesis of 3-methyl-2-(3-[2-[4-(5-[[(1r,3r)-3-(3-chloro-4-cyanophenoxy)- 2,2,4,4-tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperazin-1-yl]ethyl]-1,2-oxazol-5- yl)butanoic acid.
Figure imgf000468_0001
[0917] Into a 25-mL round-bottom flask, was placed ethyl 3-methyl-2-(3-[2-[4-(5-[[(1r,3r)-3-(3- chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperazin-1- yl]ethyl]-1,2-oxazol-5-yl)butanoate (159 mg, 0.23 mmol, 1.00 equiv), MeOH/H2O(5ml/0.5ml). LiOH (17 mg, 0.71 mmol, 3.00 equiv). The resulting solution was stirred for 12 hours at 25oC. The resulting mixture was concentrated under vacuum. The residue was applied onto a silica gel column with dichloromethane/methanol (1/5). This resulted in 0.133 g (87%) of 3-methyl-2-(3- [2-[4-(5-[[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperazin-1-yl]ethyl]-1,2-oxazol-5-yl)butanoic acid as a white solid. LC-MS (ES+): m/z 663.30 [MH+], tR = 0.94 min, (1.90 minute run).
[0918] Step 12: Synthesis of 6-[4-[2-(5-[1-[(2S,4R)-4-hydroxy-2-[[(1S)-1-[4-(4-methyl-1,3- thiazol-5-yl)phenyl]ethyl]carbamoyl]pyrrolidin-1-yl]-3-methyl-1-oxobutan-2-yl]-1,2-oxazol-3- yl)ethyl]piperazin-1-yl]-N-[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]pyridine-3-carboxamide.
Figure imgf000468_0002
[0919] Into a 25-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 3-methyl-2-(3-[2-[4-(5-[[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperazin-1-yl]ethyl]-1,2-oxazol-5-yl)butanoic acid (133 mg, 0.20 mmol, 1.00 equiv), N,N-dimethylformamide (5 mL), T3P (50% in EA)(0.254 g, 2.00 equiv), DIEA (77 mg, 0.60 mmol, 3.00 equiv), (2S,4R)-4-hydroxy-N-[(1S)-1-[4-(4- methyl-1,3-thiazol-5-yl)phenyl]ethyl]pyrrolidine-2-carboxamide (79 mg, 0.24 mmol, 1.20 equiv). The resulting solution was stirred for 12 hours at 25oC. The solids were filtered out. The crude product ( mL) was purified by Prep-HPLC with the following conditions (Column:
XBridge Shield RP18 OBD Column, 5um,19*150mm;Mobile Phase A:Water(10MMOL/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 20 mL/min; Gradient: 46% B to 64% B in 8 min; 220 nm; Rt: 7.58 min): This resulted in 0.0363 g (19%) of 6-[4-[2-(5-[1-[(2S,4R)-4-hydroxy-2- [[(1S)-1-[4-(4-methyl-1,3-thiazol-5-yl)phenyl]ethyl]carbamoyl]pyrrolidin-1-yl]-3-methyl-1- oxobutan-2-yl]-1,2-oxazol-3-yl)ethyl]piperazin-1-yl]-N-[(1r,3r)-3-(3-chloro-4-cyanophenoxy)- 2,2,4,4-tetramethylcyclobutyl]pyridine-3-carboxamide as a white solid. 1H NMR (300 MHz, CDCl3): ^ 8.66 (s, 1H), 8.56-8.55(m, 1H), 7.91-7.87 (m, 1H), 7.52-7.60 (m, 1H), 7.46-7.30 (m, 5H), 6.97-6.96 (m, 1H), 6.82-6.78 (m, 1H), 6.70-6.58 (m, 1H), 6.18 (d,1H,J = 8.7 Hz), 6.06 (d,1H,J = 8.1 Hz),5.14-4.85 (m, 1H), 4.84-4.52 (m, 2H), 4.14(d,1H,J = 8.1 Hz),4.04 (s, 1H), 3.81-3.42 (m, 7H), 3.05-2.81 (m, 3H), 2.80-2.55 (m, 5H), 2.54-2.36 (m, 5H), 2.25-2.10 (m, 1H), 2.03-1.88 (m, 1H), 1.52-1.33 (m, 3H), 1.30-1.14 (m, 12H), 1.08-1.01 (m, 3H), 0.92-0.89 (m, 3H); LC-MS (ES+): m/z 976.40 [MH+], tR = 1.51 min, (3.00 minute run). Chemical Formula: C52H62ClN9O6S [975.42/977.42].
[0920] Unless otherwise noted, Examples 839-840 were synthesized according to similar procedures described above for the synthesis of Example 838, by utilizing corresponding starting materials and reagents.
[0921] Table 27: Additional Exemplary Compounds
Figure imgf000469_0001
Figure imgf000470_0001
0922 S nthesis of Exam le 841:
Figure imgf000470_0002
[0923] Step 1: ^-^^^^#^^"-^-^^-^^ ^^^-^-^^^^^^^^^^!^"-^#^#^#^-^^^^^^^^^^ ^^^ ^$%^^ "-+-^^-^^- ^^-^^-^^^^"-^-^^^^#^8"-^-^^(^^!^-^-^^^8"-^-^^(^^!^-^-^^-^^-^^^^^ ^^^^'^ -0- yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)amino)-2- oxoethyl i erazin-1- l eth l rrolidin-1- l nicotinamide.
Figure imgf000471_0001
[0924] Into a 25-mL round-bottom flask, was placed 2-(4-(2-(1-(5-(((1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)carbamoyl)pyridin-2-yl)pyrrolidin-3- yl)ethyl)piperazin-1-yl)acetic acid (85.0 mg, 0.14 mmol, 1.00 equiv), N,N-dimethylformamide (2 mL), (2S,4R)-1-[(2S)-2-amino-3,3-dimethylbutanoyl]-4-hydroxy-N-[(1R)-2-hydroxy-1-[4-(4- methyl-1,3-thiazol-5-yl)phenyl]ethyl]pyrrolidine-2-carboxamide hydrochloride (67.8 mg, 0.14 mmol, 1.00 equiv), N,N,N,N-Tetramethyl-O-(7-azabenzotriazol-1-yl)uronium
hexafluorophospate (77.8 mg, 0.20 mmol, 1.50 equiv), N,N-diisopropylethylamine (52.8 mg, 0.41 mmol, 3.00 equiv). The resulting solution was stirred for 1 hour at room temperature. The reaction was then quenched by water (20 mL), extracted with ethyl acetate (20 mL x3) and concentrated under vacuum. The solids were filtered out. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Prep OBD C18 Column,
19*250mm,5um; mobile phase, water (10 mmol/L ammonium bicarbonate) and acetonitrile (44.0% acetonitrile up to 60.0% in 8 min); Detector, UV 254nm. This resulted in 53.2 mg (37%) of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)-6-(3-(2-(4-(2-(((S)-1- ((2S,4R)-4-hydroxy-2-(((R)-2-hydroxy-1-(4-(4-methylthiazol-5- yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)amino)-2- oxoethyl)piperazin-1-yl)ethyl)pyrrolidin-1-yl)nicotinamide as a light yellow solid. 1H NMR (300 MHz, CD3OD): ^ 8.85 (s, 1H), 8.56-8.48 (m, 1H), 7.91 (m, 1H), 7.69 (d, J = 8.7 Hz, 1H), 7.43 (m, 4H), 7.10 (m, 1H), 6.95 (m, 1H), 6.50 (d, J = 9.0 Hz, 1H), 5.00 (m, 1H), 4.64-4.52 (m, 2H), 4.43 (s, 1H), 4.24 (s, 1H), 4.10 (s, 1H), 3.93-3.51 (m, 7H), 3.42 (m, 1H), 3.15-3.01 (m, 3H), 2.60 (s, 8H), 2.45 (s, 3H), 2.38- 2.12 (m, 2H), 2.04-1.89 (m, 1H), 1.77-1.65 (m, 3H), 1.25 (s, 6H), 1.19 (s, 6H), 1.01 (s, 9H); LC-MS (ES+): m/z 1065.70 [MH+], tR =2.96 min (5.2 minute run). Chemical Formula: C56H73ClN10O7S; Molecular Weight: 1064.51.
[0925] Synthesis of Example 845:
Figure imgf000472_0001
[0926] Step 1: Synthesis of benzyl 6-(piperazin-1-yl)nicotinate.
Figure imgf000473_0001
[0927] A mixture of tert-butyl 4-(5-((benzyloxy)carbonyl)pyridin-2-yl)piperazine-1-carboxylate (20 g, 50.3 mmol) and 2,2,2-trifluoroacetic acid (15 ml) in dichloromethane (50 ml) was stirred at room temperature for 2 hours. TLC showed the reaction was complete. The volatiles were evaporated under reduced pressure. The residue was taken up in dichloromethane (100 ml) and washed with aqueous sodium bicarbonate (sat.20 ml x 2). The organic layer was collected, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give benzyl 6- (piperazin-1-yl)nicotinate (15 g, yield 95%) as yellow solid which was used in next step without further purification. 1HNMR (400 MHz, CDCl3): ^ 2.90-2.92 (m, 4H), 3.60-3.63 (m, 4H), 5.31 (s, 2H), 6.54 (d, J = 9.2 Hz, 1H), 7.30-7.42(m, 5H), 8.00-8.03 (m, 1H), 8.44 (s, 1H). Chemical Formula: C17H19N3O2, Molecular Weight: 297.35.
[0928] Step 2: Synthesis of benzyl 6-[4-[3-(benzyloxy)cyclobutyl]piperazin-1-yl]pyridine-3- carboxylate.
Figure imgf000473_0002
. [0929] Into a 50 mL round-bottom flask, was placed benzyl 6-(piperazin-1-yl)pyridine-3- carboxylate 2,2,2-trifluoroacetate salt (800 mg, 2.02 mmol, 1.00 equiv), dichloromethane (20 mL), 3-(benzyloxy)cyclobutan-1-one (704 mg, 4.00 mmol, 1.50 equiv), sodium
triacetoxyborohydride (1.7 g, 8.02 mmol, 3.00 equiv). The resulting solution was stirred for 2 hour at room temperature. The reaction was then quenched by the addition of 5 mL water. The resulting solution was extracted with dichloromethane and the organic layers combined. The resulting mixture was washed with saturate sodium chloride. The mixture was dried over anhydrous sodium sulfate. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:5). The collected fractions were combined and concentrated under vacuum. This resulted in 469 mg (51%) of benzyl 6-[4-[3-(benzyloxy)cyclobutyl]piperazin-1- yl]pyridine-3-carboxylate as a white solid. LC-MS: (ES+): m/z 458.15 [MH+], tR = 0.69min, (1.90 minute run).
[0930] Step 3: Synthesis of 6-[4-[3-(benzyloxy)cyclobutyl]piperazin-1-yl]pyridine-3-carboxylic acid.
Figure imgf000474_0001
[0931] Into a 50 mL round-bottom flask, was placed benzyl 6-[4-[3- (benzyloxy)cyclobutyl]piperazin-1-yl]pyridine-3-carboxylate (469 mg, 1.02 mmol, 1.00 equiv), ethanol (20 mL), Palladium carbon (100 mg), hydrogen. The resulting solution was stirred for overnight at 30oC in an oil bath. The solids were filtered out. The resulting mixture was concentrated under vacuum. This resulted in 611 mg (162%) of 6-[4-[3- (benzyloxy)cyclobutyl]piperazin-1-yl]pyridine-3-carboxylic acid as a yellow solid.
[0932] Step 4: Synthesis of 6-[4-[3-hydroxycyclobutyl]piperazin-1-yl]pyridine-3-carboxylic acid.
Figure imgf000474_0002
[0933] Into a 50 mL round-bottom flask, was placed 6-[4-[3-(benzyloxy)cyclobutyl]piperazin-1- yl]pyridine-3-carboxylic acid (610 mg, 1.66 mmol, 1.00 equiv), dichloromethane (10 mL). This was followed by the addition of a solution of tribromoborane (623 mg, 2.49 mmol, 1.50 equiv) in dichloromethane (2.5 mL) dropwise with stirring at 0oC in 10 min. The resulting solution was stirred for 0.5 hours at room temperature. The reaction was then quenched by the addition of sodium bicarbonate. The resulting solution was diluted with 10 mL of methanol. The solids were filtered out. The crude product ( 460 mg) was purified by Prep-HPLC with the following conditions: Column, 5um,19*150mm; mobile phase, Water(10mmol/L NH4HCO3) and acetonitrile (48.0% acetonitrile up to 62.0% in 8 min); Detector, UV 220nm. This resulted in 120 mg (26%) of 6-[4-[3-hydroxycyclobutyl]piperazin-1-yl]pyridine-3-carboxylic acid as a white solid.
[0934] Step 5: Synthesis of 6-[4-(3-hydroxycyclobutyl)piperazin-1-yl]-N-[(1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyridine-3-carboxamide.
Figure imgf000475_0001
[0935] Into a 50 mL round-bottom flask, was placed 6-[4-[3-(benzyloxy)cyclobutyl]piperazin-1- yl]-N-[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyridine-3- carboxamide (322 mg, 0.51 mmol, 1.00 equiv), dichloromethane (10 mL). This was followed by the addition of a solution of BBr3 (190 mg, 1.50 equiv) in dichloromethane (4 mL) dropwise with stirring at 0oC in 10 minutes. The resulting solution was stirred for 1 day at room temperature. The reaction was then quenched by the addition of sodium bicarbonate. The resulting solution was extracted with dichloromethane and the organic layers combined. The resulting mixture was washed with saturate sodium chloride. The mixture was dried over anhydrous sodium sulfate. The residue was applied onto a silica gel column with dichloromethane/methanol (20:1). The collected fractions were combined and concentrated under vacuum. This resulted in 200 mg (73%) of 6-[4-(3-hydroxycyclobutyl)piperazin-1-yl]-N-[(1r,3r)-3-(3-chloro-4-cyanophenoxy)- 2,2,4,4-tetramethylcyclobutyl]pyridine-3-carboxamide as a yellow solid.
[0936] Step 6: Synthesis of tert-butyl 2-[3-[4-(5-[[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamo l ridin-2- l i erazin-1- l c clobutoxy]acetate.
Figure imgf000475_0002
[0937] Into a 50 mL round-bottom flask, was placed 6-[4-(3-hydroxycyclobutyl)piperazin-1-yl]- N-[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyridine-3-carboxamide (180 mg, 0.33 mmol, 1.00 equiv), tert-butyl 2-diazoacetate (71.4 mg, 0.50 mmol, 1.50 equiv), dichloromethane (20 mL), Rh2(OAc)4 (14.7 mg, 0.10 equiv). The resulting solution was stirred for 1 overnight at room temperature. The reaction was then quenched by the addition of water. The resulting solution was extracted with dichloromethane and the organic layers combined. The resulting mixture was washed with saturate sodium chloride. The mixture was dried over anhydrous sodium sulfate. The residue was applied onto a silica gel column with
dichloromethane/methanol (10:1). The collected fractions were combined and concentrated under vacuum. This resulted in 150 mg (69%) of tert-butyl 2-[3-[4-(5-[[(1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperazin-1- yl]cyclobutoxy]acetate as a yellow oil. LC-MS: (ES+): m/z 652.35 [MH+], tR = 1.04min, (1.90 minute run).
[0938] Step 7: Synthesis of 2-[3-[4-(5-[[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl] carbamo l ridin-2- l i erazin-1- l c clobutoxy]acetic acid.
Figure imgf000476_0001
[0939] Into a 50 mL round-bottom flask, was placed tert-butyl 2-[3-[4-(5-[[(1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperazin-1- yl]cyclobutoxy]acetate (150 mg, 0.23 mmol, 1.00 equiv), dichloromethane (10 mL),
trifluoroacetic acid (2 mL). The resulting solution was stirred for 2 hours at room temperature. The resulting mixture was concentrated under vacuum. This resulted in 136 mg (99%) of 2-[3- [4-(5-[[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]carbamoyl]pyridin-2- yl)piperazin-1-yl]cyclobutoxy]acetic acid as yellow oil.
[0940] Step 8: Synthesis of 6-[4-[3-([[(2S)-1-[(2S,4R)-4-hydroxy-2-[[(1S)-1-[4-(4-methyl-1,3- thiazol-5-yl)phenyl]ethyl]carbamoyl]pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl]methoxy) cyclobutyl]piperazin-1-yl]-N-[(1r,3r)-3-(3-chloro-4-cyanophenoxy)- 2,2,4,4-tetramethylcyclobutyl]pyridine-3-carboxamide.
Figure imgf000477_0001
[0941] Into a 50 mL round-bottom flask, was placed 2-[3-[4-(5-[[(1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]carbamoyl]pyridin-2-yl)piperazin-1- yl]cyclobutoxy]acetic acid (126 mg, 0.21 mmol, 1.00 equiv), (2S,4R)-1-[(2S)-2-amino-3,3- dimethylbutanoyl]-4-hydroxy-N-[(1S)-1-[4-(4-methyl-1,3-thiazol-5-yl)phenyl]ethyl]pyrrolidine- 2-carboxamide (94.1 mg, 0.21 mmol, 1.00 equiv), N,N-dimethylformamide (10 mL), DIEA (81.9 mg, 0.63 mmol, 3.00 equiv), BOP (93.7 mg, 1.00 equiv). The resulting solution was stirred for 1 hour at room temperature. The reaction was then quenched by the addition of. The resulting solution was extracted with DCM:MeOH=10:1 and the organic layers combined. The resulting mixture was washed with saturate sodium chloride. The mixture was dried over anhydrous sodium sulfate. The crude product (210 mg) was purified by Prep-HPLC with the following conditions: Column, XBridge Shield RP18 OBD Column, 5um,19*150mm; mobile phase, Water(10 mmol/L NH4HCO3) and acetonitrile (48.0% acetonitrileup to 62.0% in 8 min);
Detector, UV 220nm.50 mg product was obtained and concentrated under vacuum. This resulted in 50 mg (23%) of 6-[4-[3-([[(2S)-1-[(2S,4R)-4-hydroxy-2-[[(1S)-1-[4-(4-methyl-1,3-thiazol-5- yl)phenyl]ethyl]carbamoyl]pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl]methoxy)cyclobutyl]piperazin-1-yl]-N-[(1r,3r)-3-(3-chloro-4-cyanophenoxy)- 2,2,4,4-tetramethylcyclobutyl]pyridine-3-carboxamide as yellow oil.
[0942] Step 9: Synthesis of N-[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]-6-[4-[(1s,3s)-3-([[(2S)-1-[(2S,4R)-4-hydroxy-2-[[(1S)-1-[4-(4-methyl- 1,3-thiazol-5-yl)phenyl]ethyl] carbamoyl] pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2- yl]carbamoyl]methoxy)cyclobutyl]piperazin-1-yl]pyridine-3-carboxamide.
Figure imgf000478_0001
[0943] Into a 50 mL round-bottom flask, was placed 6-[4-[3-([[(2S)-1-[(2S,4R)-4-hydroxy-2- [[(1S)-1-[4-(4-methyl-1,3-thiazol-5-yl)phenyl]ethyl]carbamoyl]pyrrolidin-1-yl]-3,3-dimethyl-1- oxobutan-2-yl]carbamoyl]methoxy)cyclobutyl]piperazin-1-yl]-N-[(1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyridine-3-carboxamide (50 mg, 0.05 mmol, 1.00 equiv). The resulting solution was stirred for 1 minute at room temperature. The crude product (50 mg) was purified by Chiral-Prep-HPLC with the following conditions: Column,
CHIRALPAK IF, 2*25cm,5um; mobile phase, 2-Methyl-2-methoxy propane and
isopropylamine- (hold 45.0% isopropylamine- in 32 min); Detector, UV 220/254nm.31.5 mg product was obtained, and concentrated under vacuum. This resulted in 31.5 mg (63%) of N- [(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]-6-[4-[(1s,3s)-3-([[(2S)-1- [(2S,4R)-4-hydroxy-2-[[(1S)-1-[4-(4-methyl-1,3-thiazol-5-yl)phenyl]ethyl]carbamoyl]pyrrolidin- 1-yl]-3,3-dimethyl-1-oxobutan-2-yl]carbamoyl]methoxy)cyclobutyl]piperazin-1-yl]pyridine-3- carboxamide as a white solid. 1H NMR (400 MHz, CD3OD) ^ 8.87 (s, 1H), 8.61- 8.59(m, 1H), 7.98 - 7.92(m, 1H), 7.74- 7.70 (m, 1H), 7.57 - 7.38 (m, 4H), 7.13- 7.10 (m, 1H), 6.99 - 6.96(m, 1H), 6.85 - 6.81(m, 1H), 5.02 - 4.99(m, 1H), 4.72 - 4.66(m, 1H), 4.62- 4.57(m, 1H), 4.46- 4.38(m, 1H), 4.27(m, 1H), 4.13(m, 1H), 4.00- 3.88(m, 4H), 3.86- 3.67 (m, 5H), 2.60- 2.42(m, 10H), 2.22- 2.15(m, 1H), 2.02- 1.90(m, 3H), 1.57- 1.46(m, 3H), 1.27- 1.21(d, J= 24.4Hz, 12H), 1.05 - 1.02(S, 9H); LC-MS (ES+): m/z 1022.5[MH+], tR = 4.294min, (5.60 minute run).
Chemical Formula: C54H68ClN9O7S; Molecular Weight 1021.47.
[0944] Unless otherwise noted, Examples 842-852 were synthesized according to similar procedures described above for the synthesis of Examples 841 and 845, by utilizing
corresponding starting materials and reagents.
[0945] Table28: Additional Exemplary Compounds Ex. Measured # Structure Compound Name Mass Ion Data
Figure imgf000479_0001
^^^^^^^^"^^^^^^^^^^^^^^^^
^^^^^^^^^^^^^^^^^^^^^^ ^^^^^^^^^^^^^^^^^ ^^^^^^^ 844
Figure imgf000480_0001
^^^^$^^^$^^^$#^^ [0946] Table 29: Additional Exemplary Compounds
Figure imgf000480_0002
Figure imgf000481_0001
Figure imgf000482_0001
[0947] Synthesis of Example 853:
Figure imgf000483_0001
Figure imgf000483_0002
[0948] Step 1: Synthesis of benzyl 6-[4-(hydroxymethyl)piperidin-1-yl]pyridine-3-carboxylate:
Figure imgf000483_0003
[0949] Into a 100-mL round-bottom flask, was placed benzyl 6-chloropyridine-3-carboxylate (4.0 g, 16.1 mmol, 1.00 equiv), N,N-dimethylformamide (30 mL), piperidin-4-ylmethanol (1.85 mg, 16.1 mmol, 1.00 equiv), potassium carbonate (6.6 g, 48.0 mmol, 3.00 equiv). The resulting solution was stirred for 16 hours at 100 °C in an oil bath. The resulting solution was extracted with ethyl acetate (100 mL x 2) and the organic layers combined and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1/1). This resulted in 3.42 g (65%) of benzyl 6-[4-(hydroxymethyl)piperidin-1-yl]pyridine-3- carboxylate as colorless oil.
[0950] Step 2: Synthesis of benzyl 6-(4-formylpiperidin-1-yl)pyridine-3-carboxylate:
Figure imgf000484_0001
[0951] Into a 250-mL round-bottom flask, was placed benzyl 6-[4-(hydroxymethyl)piperidin-1- yl]pyridine-3-carboxylate (3.42 g, 10.6 mmol, 1.00 equiv) and 100 mL dichloromethane.1,1,1- Triacetoxy)-1,1-dihydro-1,2-benziodoxol-3(1H)-one (6.7 g, 15.8 mmol, 1.50 equiv) was added slowly. The resulting solution was stirred for 4 hours at room temperature. The reaction was then quenched by the addition of 100 mL of saturated Na2S2O3. The resulting solution was extracted with dichloromethane (100 mL x 2) and the organic layers combined. The mixture was dried over anhydrous sodium sulfate. The resulting mixture was concentrated under vacuum. This resulted in 3.06 g (67%) of benzyl 6-(4-formylpiperidin-1-yl)pyridine-3-carboxylate as yellow oil.
[0952] Step 3: Synthesis of benzyl 6-(4-[[4-(2-ethoxy-2-oxoethyl)piperazin-1- yl]methyl]piperidin-1-yl ridine-3-carbox late:
Figure imgf000484_0002
[0953] Into a 100-mL round-bottom flask, was placed benzyl benzyl 6-(4-formylpiperidin-1- yl)pyridine-3-carboxylate (3.06 g, 9.44 mmol, 1.00 equiv) and ethyl 2-(piperazin-1-yl)acetate hydrochloride (2.0 g, 9.44 mmol, 1.00 equiv). The resulting solution was stirred at room temperature for 5 minutes and then sodium triacetoxyborohydride (6.0 g, 28.32 mmol, 3.00 equiv) was added. The reaction was then quenched by the addition of 100 mL of water. The resulting solution was extracted with ethyl acetate (100 mL x 2) and the organic layers combined and concentrated under vacuum. The residue was applied onto a silica gel column with dichloromethane/methanol (9/1). This resulted in 2.3 g (51%) of benzyl 6-(4-[[4-(2-ethoxy-2- oxoethyl)piperazin-1-yl]methyl]piperidin-1-yl)pyridine-3-carboxylate as yellow oil. LC-MS (ES+): m/z 481.50 [MH+], tR = 1.25 min (1.9 minute run).
[0954] Step 4: Synthesis of 6-(4-[[4-(2-ethoxy-2-oxoethyl)piperazin-1-yl]methyl]piperidin-1- yl)pyridine-3-carboxylic acid:
Figure imgf000485_0001
[0955] To a solution of benzyl 6-(4-[[4-(2-ethoxy-2-oxoethyl)piperazin-1-yl]methyl]piperidin-1- yl)pyridine-3-carboxylate (2.3 g, 4.79 mmol, 1.00 equiv) in 100 mL EtOH was added Pd/C (10%, 1.0 g) under nitrogen atmosphere in a 250 ml round bottom flask. The flask was then vacuumed and flushed with hydrogen. The reaction mixture was hydrogenated at room temperature for 2 hours under hydrogen atmosphere using a hydrogen balloon, then filtered through a Celite pad and concentrated under reduced pressure. This resulted in 1.3251 g (71 %) of 6-(4-[[4-(2-ethoxy-2-oxoethyl)piperazin-1-yl]methyl]piperidin-1-yl)pyridine-3-carboxylic acid as a white solid. 1H NMR (300 MHz, CD3OD): 8.64 (d, J = 2.4 Hz, 1H), 7.97 (dd, J = 9.1, 2.4 Hz, 1H), 6.75 (d, J = 9.1 Hz, 1H), 4.40 (d, J = 13.3 Hz, 2H), 4.15 (q, J = 7.1 Hz, 2H), 3.26 (s, 2H), 2.92 (td, J = 12.9, 2.6 Hz, 2H), 2.69 (s, 8H), 2.41 (d, J = 6.9 Hz, 2H), 1.89 (dd, J = 30.6, 10.6 Hz, 3H), 1.29-1.10 (m, 5H); LC-MS (ES+): m/z 391.35 [MH+], tR = 0.70 min (3.0 minute run). Chemical Formula: C20H30N4O4; Molecular Weight 390.23.
[0956] Steps 5-7: Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-6-(4-((4-(2-(((S)-1-((2S,4R)-4-hydroxy-2-(((S)-1-(4-(4-methylthiazol-5- yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)amino)-2- oxoethyl)piperazin-1-yl)methyl)piperidin-1-yl)nicotinamide: Steps 5-7 were synthesized according to similar procedures described above for the synthesis of examples 829 and 833, by using corresponding starting materials and reagents.
[0957] Synthesis of Example 864:
Figure imgf000486_0001
[0958] Step 1: Synthesis of Tert-Butyl 2-(4-(2-hydroxyethyl)piperazin-1-yl)acetate
Figure imgf000486_0002
[0959] To a solution of 2-(piperazin-1-yl)ethanol (10 g, 0.077 mol, 1.0 equiv) in THF (60 mL) were added a solution of tert-butyl 2-bromoacetate (14.9 g, 0.077 mol, 1.0 equiv) in THF (10 mL) and TEA (15.5 g, 0154 mol, 2.0 equiv) sequentially at 0oC. The solution was stirred at 50oC for 16 hours. The solvent was concentrated under reduced pressure. The residue was diluted with DCM and washed with saturated aqueous ammonium chloride. The combined organic layers were dried (Na2SO4), filtered and concentrated under reduced pressure to afford [007] tert-Butyl 2-(4-(2-hydroxyethyl)piperazin-1-yl)acetate (16 g, 85.1% yield) as a light yellow liquid. 1H NMR (400 MHz, CDCl3): δ 3.61 (t, J = 5.6 Hz, 2H), 3.12 (s, 2H), 2.54–2.60 (m, 8H), 1.47 (s, 9H). Chemical Formula: C12H24N2O3; Molecular Weight 244.33;
[0960] Step 2: Synthesis of Tert-Butyl 2-(4-(2-chloroethyl)piperazin-1-yl)acetate
Figure imgf000487_0001
[0961] To a solution of tert-Butyl 2-(4-(2-hydroxyethyl)piperazin-1-yl)acetate (3 g, 0.012 mol, 1.0 equiv) in DCM (50 mL) were added TEA (3.6 g, 0.036 mol, 3.0 equiv) and TsCl (4.7 g, 0.024 mol, 2.0 eq). The mixture was stirred at 20oC for 4 hours. The solution was quenched with water, and then extracted with DCM. The combined organic layers were dried (Na2SO4), filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE/EtOAc from 1:0 to 5:1, and then 1:1) to afford the desired product (1.6 g, 49.5% yield) as a light yellow liquid. 1H NMR (400 MHz, CDCl3): δ 3.58 (t, J = 7.2 Hz, 2H), 3.10 (s, 2H), 2.74 (t, J = 7.2 Hz, 2H), 2.60 (s, 8H), 1.46 (s, 9H).Chemical Formula:
C12H23ClN2O2; Molecular Weight: 262.78;
[0962] Step 3: Synthesis of 5-Acetyl-1H-pyrazole-3-carboxylic acid
Figure imgf000487_0002
[0963] To a solution of InCl3 (1.3 g, 5.882 mmol, 0.2 equiv) in water (60 mL) were added 2- ethoxy-2-oxoethanediazonium (4.1 g, 0.0324 mol, 1.1 equiv) and but-3-yn-2-one (2 g, 0.0294 mol, 1.0 equiv). The solution was stirred at 25oC for 4 hours. The mixture was extracted with EtOAc. The combined organic layers were washed with brine. The organic phase was dried (Na2SO4), filtered and concentrated under reduced pressure to afford the crude product (3.9 g). Then the crude product (3.9g, 0.0214mol, 1equiv) was dissolved in THF/H2O (v/v = 10:1, 80mL). LiOH (3.6 g, 0.0857mol, 4.0 equiv) was added. The mixture was stirred at 25oC for 16 hours. The pH of the solution was adjusted to 7~8 with aq.HCl (2M). The solution was concentrated under reduced pressure. The residue was washed with THF. The combined organic layers were concentrated under reduced pressure to afford 5-Acetyl-1H-pyrazole-3-carboxylic acid (3 g, 66.2% yield) as a brown solid. 1H NMR (400 MHz, MeOD): δ 7.01 (s, 1H), 2.54 (s, 3H). Chemical Formula: C6H6N2O3; Molecular Weight: 154.12. [0964] Step 4: Synthesis of (S)-5-Acetyl-N-(1-(3-(3-chloro-4-cyanophenyl)-1H-pyrazol-1- yl)propan-2-yl)-1H-pyrazole-3-carboxamide
Figure imgf000488_0001
[0965] To a solution of 5-Acetyl-1H-pyrazole-3-carboxylic acid (1.27 g, 70%, 5.76 mmol, 1.5 equiv) in DMF (10 mL) were added DIEA (1.5 g, 11.52 mmol, 3.0 equiv) and 3-(3-chloro-4- cyanophenyl)-1H-pyrazol (1 g, 3.84 mmol, 1.0 equiv). 5min later, HATU (1.75 g, 4.60 mmol, 1.2 equiv) was added. The mixture was stirred at 25oC for 30 minutes. Then it was quenched with water. The solution was extracted with EtOAc. The combined organic layers were washed with water and brine. The organic phase was dried (Na2SO4), filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE/EtOAc = 1/1) to afford (S)-5-Acetyl-N-(1-(3-(3-chloro-4-cyanophenyl)-1H-pyrazol-1-yl)propan-2-yl)-1H- pyrazole-3-carboxamide (350 mg, 22.9% yield) as a grey solid. 1H NMR (400 MHz, MeOD): δ 7.89 (s, 1H), 7.77–7.89 (m, 1H), 7.70 (s, 1H), 7.69 (s, 1H), 7.24 (s, 1H), 7.76 (d, J = 2.4Hz, 1H), 4.31 (s, 1H), 4.07–4.12 (m, 2H), 2.51 (s, 3H), 1.22–1.28 (m, 3H).
[0966] Chemical Formula: C19H17ClN6O2; Molecular Weight 396.83;
[0967] Step 5: Synthesis of 5-Acetyl-N-((S)-1-(3-(3-chloro-4-cyanophenyl)-1H-pyrazol-1- yl)propan-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carboxamide
Figure imgf000488_0002
[0968] To a solution of (S)-5-Acetyl-N-(1-(3-(3-chloro-4-cyanophenyl)-1H-pyrazol-1-yl)propan- 2-yl)-1H-pyrazole-3-carboxamide (350 mg, 0.883 mmol, 1.0 equiv) in DCM (10 mL) were added DHP (111 mg, 1.330 mmol, 1.5 equiv) and TsOH (30 mg, 0.0176 mmol, 0.2 equiv). The mixture was stirred at 20oC for 20 hours. The solution was quenched with water and extracted with EtOAc. The combined organic layers were washed with aq.NaHCO3. The organic phase was dried (Na2SO4), filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE/EtOAc from 10:1 to 5:1) to afford 5-Acetyl-N-((S)-1- (3-(3-chloro-4-cyanophenyl)-1H-pyrazol-1-yl)propan-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H- pyrazole-3-carboxamide (320 mg, 75.4% yield) as a grey solid. [0969] 1H NMR (400 MHz, MeOD): δ 7.80 (s, 1H),7.83–7.97 (m, 1H), 7.75–7.77 (m, 1H), 7.71 (d, J = 2.4 Hz, 1H), 7.35 (s, 1H), 6.76 (d, J = 4.4 Hz, 1H), 6.24–6.24 (d, J = 2.0 Hz, 1H), 4.31 (s, 1H), 4.07–4.12 (m, 2H), 3.97–4.02 (m,1H), 3.78–3.81 (m, 1H), 2.51 (s, 3H), 2.31–2.41 (m, 1H), 1.71–1.72 (m, 2H), 1.59–1.61 (m, 2H), 1.22–1.28 (m, 3H).
[0970] Chemical Formula: C24H25ClN6O3; Molecular Weight: 480.95;
[0971] Step 6: Synthesis of N-((S)-1-(3-(3-Chloro-4-cyanophenyl)-1H-pyrazol-1-yl)propan- 2-yl)-5-((Z)-1-(hydroxyimino)ethyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3- carboxamide
Figure imgf000489_0001
[0972] To a solution of 5-Acetyl-N-((S)-1-(3-(3-chloro-4-cyanophenyl)-1H-pyrazol-1- yl)propan-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carboxamide (320 mg, 0.665 mmol, 1.0 equiv) in EtOH (20 mL) were added NaOAc (1.09 g, 13.304 mmol, 20 equiv) and NH2OH·HCl (924 mg, 13.304 mmol, 20 equiv). The solution was stirred at 20oC for 20 hours, and then concentrated under reduced pressure. The residue was dissolved in EtOA and washed with brine. The organic layer was dried (Na2SO4), filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (DCM/MeOH = 100/1) to afford N-((S)-1-(3-(3-Chloro-4-cyanophenyl)-1H-pyrazol-1-yl)propan-2-yl)-5-((Z)-1- (hydroxyimino)ethyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carboxamide (100 mg, 30.3% yield) as a white solid.
[0973] 1H NMR (400 MHz, MeOD): δ 8.17–8.20 (m, 1H), 7.95 (s, 1H), 7.72–7.79 (m, 1H), 7.68–7.71 (m, 1H), 6.83 (d, J = 2.0 Hz, 1H), 6.71–6.73 (m, 1H), 6.08 (d, J = 1.6 Hz, 1H), 4.31 (s, 1H), 4.07–4.12 (m, 2H), 3.97–4.02 (m, 1H), 3.78–3.81 (m, 1H), 2.09–3.31 (m, 3H), 1.92–1.99 (m, 4H), 1.58 (s, 3H), 1.21–1.28 (m, 3H).
[0974] Chemical Formula: C24H26ClN7O3; Molecular Weight: 495.96;
[0975] Step 7: Synthesis of (S,E)-Tert-Butyl 2-(4-(2-(((1-(3-((1-(3-(3-chloro-4-cyanophenyl)- 1H-pyrazol-1-yl)propan-2-yl)carbamoyl)-1H-pyrazol-5-yl)ethylidene)amino)oxy)ethyl)- piperazin-1-yl)acetate
Figure imgf000489_0002
[0976] To a solution of N-((S)-1-(3-(3-Chloro-4-cyanophenyl)-1H-pyrazol-1-yl)propan-2-yl)-5- ((Z)-1-(hydroxyimino)ethyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carboxamide (100 mg, 0.2016 mmol, 1.0 equiv) was added NaH (15 mg, 0.6048 mmol, 3.0 equiv) at 20oC. After 10 min, tert-Butyl 2-(4-(2-chloroethyl)piperazin-1-yl)acetate (79.5 mg, 0.3024 mmol, 1.5 equiv) was added. The mixture was stirred at 20oC for 20 hours. The solution was quenched with water, and then extracted with EtOAc. The combined organic layers were washed with brine. The organic layer was dried (Na2SO4), filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (DCM/MeOH = 100:1) to afford (S,E)-tert- Butyl 2-(4-(2-(((1-(3-((1-(3-(3-chloro-4-cyanophenyl)-1H-pyrazol-1-yl)propan-2-yl)carbamoyl)- 1H-pyrazol-5-yl)ethylidene)amino)oxy)ethyl)- piperazin-1-yl)acetate (60 mg, 41.2% yield) as a white solid, which was used into the next step without further purification. [0977] Step 8: Synthesis of N-((S)-1-(3-(3-Chloro-4-cyanophenyl)-1H-pyrazol-1-yl)propan- 2-yl)-5-((E)-1-((2-(4-(2-(((S)-1-((2S,4R)-4-hydroxy-2-(((S)-1-(4-(4-methylthiazol-5- yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)amino)-2- oxoethyl)piperazin-1-yl)ethoxy)imino)ethyl)-1H-pyrazole-3-carboxamide
Figure imgf000490_0001
[0978] To a solution of (S,E)-tert-Butyl 2-(4-(2-(((1-(3-((1-(3-(3-chloro-4-cyanophenyl)-1H- pyrazol-1-yl)propan-2-yl)carbamoyl)-1H-pyrazol-5-yl)ethylidene)amino)oxy)ethyl)- piperazin- 1-yl)acetate (60 mg) in dioxane (2 mL) was added HCl-dioxane (4mL, 6N). The mixture was stirred at 20oC for 2 hours. The solution was concentrated in vacuo to afford the crude desired product (60 mg). Then the crude product (60 mg, 0.103mmol, 1.0equiv) was dissolved in DMF (5 mL). TEA (208 mg, 2.06 mmol, 20.0 equiv) and ULM-3 (55 mg, 0.124 mmol, 1.2 equiv) were added at 25oC. 5 minutes later, EDCI (197mg, 1.03mmol, 10 equiv) and HOBt (139mg, 1.03mmol, 10 equiv) were added. The resulting solution was stirred at 25oC for 16 hours. The mixture was quenched with water, and then extracted with EtOAc. The combined organic layers were washed with brine. The organic layer was dried (Na2SO4), filtered and concentrated under reduced pressure. The residue was purified by Prep.TLC (DCM/MeOH = 13:1) to afford N-((S)- 1-(3-(3-Chloro-4-cyanophenyl)-1H-pyrazol-1-yl)propan-2-yl)-5-((E)-1-((2-(4-(2-(((S)-1- ((2S,4R)-4-hydroxy-2-(((S)-1-(4-(4-methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin-1-yl)- 3,3-dimethyl-1-oxobutan-2-yl)amino)-2-oxoethyl)piperazin-1-yl)ethoxy)imino)ethyl)-1H- pyrazole-3-carboxamide (33.2 mg, 31.9% yield) as a white solid.
[0979] 1H NMR (400 MHz, MeOD): δ 8.86 (s, 1H),7.96 (s, 1H), 7.96 (s, 1H), 7.70–7.78 (m, 1H), 7.69 (s, 1H), 7.39–7.44 (m,4H), 6.75 (d, J = 2.4 Hz, 1H), 4.99–5.00 (m, 1H), 4.62 (s, 1H), 4.55–4.56 (m, 3H), 4.38–4.43 (m, 5H), 3.72–3.89 (m, 2H), 3.07 (s, 1H), 2.99 (s, 1H), 2.82–2.86 (m, 3H), 2.64–2.67 (m, 7H), 2.47 (s, 3H), 2.18 (s, 4H), 1.91–1.99 (m, 1H), 1.49 (d, J = 7.2 Hz, 2H), 1.25–1.30 (m, 6H), 1.04(s, 9H).
[0980] LC-MS: (ES+): m/z 1008.4 [M+H]; tR = 3.636 min.
[0981] Chemical Formula: C50H62ClN13O6S; Molecular Weight: 1008.63;
[0982] Unless otherwise noted, Examples 854-863 were synthesized according to similar procedures described above for the synthesis of Examples 853 and 864, by utilizing
corresponding starting materials and reagents. [0983] Table 30: Additional Exemplary Compounds
Figure imgf000491_0001
Figure imgf000492_0001
Figure imgf000493_0001
Figure imgf000494_0001
Figure imgf000495_0001
[0984] In certain embodiments, the description provides a compound having a structure selected from the group consisting of Examples 1-864 (see Tables 2-30), a salt, a polymorph, and prodrug thereof. In certain additional embodiments, the description provides a composition comprising at least one of the compounds of Examples 1-864, including a salt, polymorph, and prodrug thereof. In still additional embodiments, the description provides a therapeutic composition comprising at least one of the compounds of Examples 1-864, including a salt, a polymorph, and a prodrug thereof, and a pharmaceutically acceptable carrier.
[0985] Examples– In vitro and in vivo assays.
[0986] The experimental results presented below are made with reference to the Tables and Figures 1-7.
[0987] 1. Androgen Receptor ELISA Assay.
[0988] Compounds have been evaluated in this assay in LNCaP and/or VCaP cells utilizing similar protocols. The protocols used with VCaP cells are described below. The androgen receptor ELISA assay was performed using PathScan AR ELISA (Cell Signaling Catalog#12850) according to the following assay steps:
[0989] VCaP cells are seeded at 30,000 cells/well at a volume of 200 μL/well in VCaP assay medium [Phenol red free RPMI (Gibco Cat#11835-030); 5% Charcoal Stripped (Dextran treated) FBS (Omega Scientific, Cat#FB-04); Pen/Strep Life Technologies (Gibco Cat#: 10378-016); 0. 1nM R1881 (Sigma, Cat# R0908) is added upon the start of the assay, not during initial plating of the cells) in Corning 3904 plates. The cells are grown for a minimum of 3 days.
[0990] First, cells are dosed with compounds diluted in 0.1% DMSO– use a polypropylene plate according to the following protocol: (1)(i) make 1000x stock plate in DMSO; (ii) 20mM stock diluted 1/6.7 with DMSO (5 μL + 28.3 μL DMSO) =3mM into row H; (iii) perform serial dilutions in ½ log doses (10 μL of PROTAC + 20 μL DMSO) from row H towards row B. Reserve row A for DMSO; (iv) 7 doses total (final concentration in this 1000x plate will be 3 mM, 1 mM, 333 µM, 111 ^M, etc). (2)(i) Make 10x stock plate in media; (ii) transfer 2.5 μL of the 1000x stock to a new 10x stock plate (use 12 channel pipet, start at A (DMSO control) work thru H. When 247.5 μL of media is added to this plate, it will serve as a 10x stock; (iii) make media + 1nM R1881 for making 10x stock plate; (iv) add 247.5 μL of media with 1 nM R1881 to each well of the 10x stock plate, mix.
[0991] Then 22 μL of 10x stock is added to cells and incubated for 24h. 1x Cell Signaling Cell lysis buffer is made (Catalogue #9803; comes with the kit) - prepare for 50 μL/well. Keep on ice. Media is aspirated, and 50 μL lx cell lysis buffer/well is added. The cells are placed on ice for 10 minues. The solution is mixed and transferred to PCR plate, and centrifuged at 4C for 10 minutes at 4000 rpm.
[0992] 5 μL is transferred to fresh plate (use immediately or freeze -80C); 115 μL ELISA Dilutant is added (0.15ug/ml– 0.075ug/ml; comes with the PathScan ELISA).
[0993] Add 100 μL/well AR Elisa; cover and shake, 37C for 2hrs; dump, tap, wash 4x 200 μL ELISA wash buffer; add 100 μL/well mouse AR detection Ab; cover and shake, 37C for 1hr; dump, tap, wash 4x 200 μL ELISA wash buffer; add 100 μL/well anti-mouse– HRP conjugated Ab (comes with the kit); cover and shake, 37C for 30 min; allow TMB reagent to come to room temperature; dump, tap, wash 4x 200 μL Elisa wash buffer; tap; add 100 μL TMB, shake 5min– while watching color. Add the stop reagent when light blue color develops. Add 100 μL Stop solution; shake and read at 450nM.
[0994] Progression of prostate cancer in patients treated with anti-androgen therapy usually involves one of several mechanisms of enhanced Androgen Receptor (AR) signaling, including increased intratumoral androgen synthesis, increased AR expression and AR mutations.
PROTACs (PROteolysis TArgeting Chimera), which uses bi-functional molecules that simultaneously bind a target of choice and an E3 ligase, cause ubiquitination via induced proximity and degradation of the targeted, pathological protein. As opposed to traditional target inhibition, which is a competitive process, degradation is a progressive process. As such, it is less susceptible to increases in endogenous ligand, target expression, or mutations in the target. Thus this technology seems ideal for addressing the mechanisms of AR resistance in patients with prostate cancer.
[0995] AR PROTACs degrade AR in LNCaP and VCaP cells, with nM to pM potency, and had a >85% reduction in AR concentration (Dmax). Degradation was rapid, with 50% of AR lost within 15 minutes and maximal degradation observed by 4 hours. The duration of AR knockdown was long-lasting, with no recovery of AR observed over several days. The degradation process in cells was specific, as PROTACs with an inactive epimer for E3 ligase binding did not degrade AR. AR PROTACs induced rapid apoptosis and cell death in VCaP cells. In LNCap and VCaP cell systems, AR PROTACs were anti-proliferative under conditions in which enzalutamide was inactive, such as increasing concentrations of the AR agonist R1881 and cells containing the ARF876L mutation. AR PROTACs typically had t1/2 values of several hours and bioavailability of >50% after ip or sc injection. In mice, AR PROTACs have shown in vivo activity, including involution of seminal vesicles, reduction of AR protein levels in the prostate, and regression of VCaP tumors.
[0996] The following assay results were generated using the androgen receptor ELISA Assay described above, where compound potencies were charactrized in highest percentage of Androgen Receptor degradation (Dmax) observed and compound concentration that caused 50% Androgen Receptor degradation (DC50).
[0997] Table 31. Androgen Receptor degradation (Dmax) observed and compound concentration that caused 50% Androgen Receptor degradation (DC50). Dmax: + (Dmax 925%); ++ (26% 9 Dmax 950%); +++ (51% 9 Dmax 970%); ++++ (71% 9 Dmax ); DC50: A (Dmax 950nM); B (51nM 9DC509 500nM); C (501nM 9 DC50 ).
Figure imgf000497_0001
Figure imgf000498_0001
Figure imgf000499_0001
Figure imgf000500_0001
Figure imgf000501_0002
[0998] Table 32. Additional Androgen Receptor degradation (Dmax) observed and compound concentration that caused 50% Androgen Receptor degradation (DC50). Dmax: + (Dmax 9 25%); ++ (26% 9 Dmax 950%); +++ (51% 9 Dmax 970%); ++++ (71% 9 Dmax ); DC50: A (Dmax 950nM); B (51nM 9DC509500nM); C (501nM 9 DC50 ).
Figure imgf000501_0001
Figure imgf000502_0001
Figure imgf000503_0001
Figure imgf000504_0001
[0999] Table 33. Additional Androgen Receptor degradation (Dmax) observed and compound concentration that caused 50% Androgen Receptor degradation (DC50). Dmax: + (Dmax 925%); ++ (26% 9 Dmax 950%); +++ (51% 9 Dmax 970%); ++++ (71% 9 Dmax ); DC50: A (Dmax 950nM); B (51nM 9DC509500nM); C (501nM 9 DC50.
Figure imgf000505_0001
Figure imgf000506_0001
Figure imgf000507_0001
Figure imgf000508_0001
Figure imgf000509_0002
[01000] Table 34. Additional Androgen Receptor degradation (Dmax) observed and compound concentration that caused 50% Androgen Receptor degradation (DC50). Dmax: + (Dmax 925%); ++ (26% 9 Dmax 950%); +++ (51% 9 Dmax 970%); ++++ (71% 9 Dmax ); DC50: A (Dmax 950nM); B (51nM 9DC509500nM); C (501nM 9 DC50 ).
Figure imgf000509_0001
Figure imgf000510_0001
Figure imgf000511_0001
Figure imgf000512_0001
Figure imgf000513_0002
[01001] Table 35. Androgen Receptor degradation (Dmax) observed and compound concentration that caused 50% Androgen Receptor degradation (DC50). Dmax: + (Dmax ^ 25%); ++ (26%≤ Dmax ^50%); +++ (51%≤ Dmax ^70%); ++++ (71%≤ Dmax ); DC50: A (Dmax ^ 50nM); B (51nM ^DC50 ^ 500nM); C (501nM≤ DC50 )
Figure imgf000513_0001
Figure imgf000514_0001
Figure imgf000515_0001
Figure imgf000516_0001
Figure imgf000517_0001
Table 36. Additional Androgen Receptor degradation (Dmax) observed and compound concentration that caused 50% Androgen Receptor degradation (DC 50 ). Dmax: + (Dmax 9 25%); ++ (26% 9 Dmax 950%); +++ (51% 9 Dmax 970%); ++++ (71% 9 Dmax ); DC50: A (Dmax 950nM); B (51nM 9DC509 500nM); C (501nM 9 DC50 ).
Figure imgf000518_0001
Figure imgf000519_0001
Figure imgf000520_0001
Figure imgf000521_0001
Figure imgf000522_0001
Figure imgf000523_0001
Figure imgf000524_0001
Figure imgf000525_0001
Figure imgf000526_0002
[01002] 2. VCaP Cell Proliferation Assay.
[01003] VCaP cells are plated 7,500/well 200 μL/well in VCaP assay medium [Phenol red free RPMI (Gibco Cat#11835-030); 5% Charcoal Stripped (Dextran treated) FBS (Omega Scientific, Cat#FB-04); Pen/Strep Life Technologies (Gibco Cat#: 10378-016); 0.1nM R1881 (Sigma, Cat# R0908) is added upon the start of the assay, not during initial plating of the cells).
[01004] The assay was performed as follows: the cells are grown for a minimum of 3 days to deplete androgens; dosing of PROTACs and R1881 is performed as for AR ELISA; the baseline reading of Cell Titer Glo can be performed on day of dosing.
[01005] VCaP cells with 0.1 nM R1881 will double once in 4 days. Gently draw off 110 μL of media so as not to disturb the adherent cells; add 110 μL of CTG; incubate with slow shaking for 20 minutes; and read luminescence on a plate reader.
[01006] VCaP anti-proliferation data:
[01007] GI50 definition: A (GI50 9 50nM); B (51nM 9 GI50 9 250nM); C (251nM 9 GI50 )
[01008] Table 37. Inhibition of VCaP Proliferation.
Figure imgf000526_0001
Figure imgf000527_0002
[01009] Table 38. Inhibition of VCaP Proliferation.
Figure imgf000527_0001
[01010] These results support that both the difunctional compounds (ABM-L-ULM) and androgen receptor binding moieties (ABM-e) inhibit VCaP Proliferation.
[01011] 3. Apoptosis in VCaP cells.
[01012] Figure 2 illustrates that compounds as described herein induce apoptosis in VCaP cells. VCaP cells were cultured in Charcoal Stripped Serum containing media supplemented with 0.1 nM R1881 for 48 hrs. The degree of apoptosis was ascertained with CaspaseGlo assay (Promega). These results demonstrated that PROTACs are much more potent in inducing apoptosis than an AR antagonist enzalutamide. Further, the degree of AR degradation correlates with their ability to induce apoptosis in VCaP cells.
[01013] 4. Anti-proliferation in LNCaP F876L.
[01014] Figure 3 demonstrates the anti-proliferation in LNCaP F876L cells observed with treatment with a compounds as described herein. LNCaP cells transduced with AR F876L construct were cultured in Charcoal Stripped Serum containing media. Indicated doses of enzalutamide or Example 1 were added for 7 days. CellTiterGlo reagent (Promega) was employed to assess proliferation. As shown, LNCaP cells expressing F876L construct proliferate in response to increasing doses of enzalutamide, whereas Example 1 did not exhibit agonist activity. These results demonstrated that AR PROTACs do not possess agonist activity.
[01015] 5. PSA suppression in LNCaP F876L
[01016] Compounds as described herein also suppress PSA in LNCaP F876L cells (See Figure 4). LNCaP cells transduced with AR F876L construct were cultured in Charcoal Stripped Serum containing media supplemented with 0.1 nM R1881 for 7 days. Secreted PSA in the media was detected by PSA ELISA (Sigma). These results demonstrated that AR PROTAC is able to suppress the transcriptional activity of AR in F876L containing cells.
[01017] 6. Prostate involution in C57B6 mouse model.
[01018] Figure 5 demonstrates that compounds as described herein induce prostate involution in C57B6 mouse model. 12-week old male C57BL/6 mice were treated with AR PROTAC Example 163 and its inactive epimer analog Compound A which is unable to bind to VHL E3 ligase. Enzalutamide (PO, QD, 30 mpk), Example 163 (IP, QD, 1 and 3 mpk) and Compound A (IP, QD, 1 and 3 mpk) were administered for 10 days, upon which the prostates were isolated and weighed. PROTAC Example 163 demonstrated a significant reduction in prostate weights, whereas Compound A showed no significant activity. These results demonstrated that the ability of PROTAC Example 163 to degrade AR leads to significant prostate involution in mice at very low doses.
[01019] 7. Tumor growth inhibition in VCaP xenograft model.
[01020] Figure 6 illustrates tumor growth inhibition in a VCaP xenograft model, which was achieved with compounds as described herein. VCaP cells were implanted into CB17 scid mice subcutaneously. Once the tumors were palpable, the mice were castrated, leading to temporary tumor stasis. Upon regrowth of tumors, the mice were dosed with enzalutamide (PO, QD, 30 mpk) or AR PROTAC Example 163 (IP, QD, at 30, 10 and 3 mpk) as indicated. Tumor growth inhibition was observed in all treatment arms.
[01021] 8. AR degradation of PROTAC is E3 ligase dependent.
[01022] Figure 7A and Figure 7B demonsrates that AR degration achieved with compounds as described herein is E3 ligase dependent. For example, in Figure 7A, AR
PROTAC Example 1 was added to LNCaP cells at indicated concentrations for 24 hours in the presence or absence of 10 uM VHL E3 ligase ligand compound B. The presence of compound B competes with AR PROTAC Example 1 in VHL E3 ligase binding and greatly diminishes the AR degradation activity of AR PROTAC Example 1. In Figure 7B, LNCaP cells were treated with AR PROTAC Example 1 and its inactive epimer analog compound C which is unable to bind to VHL E3 ligase. While AR PROTAC Example 1 led to significant degradation of AR, compound C did not. These results demonstrated that AR PROTAC activity in AR degradation is VHL E3 ligase dependent.”
[01023] 9. PROTAC prodrug oral pharmacokinetics and PROTAC Subcutaneous phamacokinetics.
[01024] Representative Pharmacokinetic Procedure
[01025] Male CD-1 mice (6-8 weeks old, weighing 20-30 g, 3 per study) with free access to food and water were administered with the test article at 10 mg/kg either by oral gavage or sub-cutaneous injection in the formulation specified in tables 20 and 21, at 10 mL/kg.
[01026] Approximately 0.04 mL blood samples were collected from the dorsal metatarsal vein serially at 0.25, 0.5, 1, 2, 4, 8 and 24 h timepoints; heparin was used as the anticoagulant. The samples were centrifuged at 4000 g for 5 min at 4 °C then stored at -75 °C prior to analysis.
[01027] The plasma samples were analysed via an LC/MS/MS method quantitating for unchanged, administered test article, and/or a derivative species as appropriate. WinNonlin (PhoenixTM) was used for the pharmacokinetic calculations and modeling, to generate parameters such as Cmax and AUC.
[01028] Table 39: Examples of PROTAC prodrug pharmacokinetics (ESP-4: 5% EtOH, 5% solutol HS15 in PBS; ESD-45% EtOH, 15% solutol in D5W).
Figure imgf000529_0001
Figure imgf000530_0002
[01029] Table 40: Examples of PROTAC Subcutaneous pharmacokinetics (ELP-1: 5% EtOH, 20% labrasol in PBS; ESD-2: 5% EtOH, 20% solutol in D5W).
Figure imgf000530_0001
[01030] In summary, PROTACs designed to degrade AR are potent (low nM to pM), specific, rapid (within 2-4 hrs); long-lasting (days); active in vitro and in vivo, and have cellular efficacy superior to enzalutamide. AR PROTACs have efficacy in cell systems and work in vivo (AR degradation in prostate; prostate involution in prostate and seminal vesicle; tumor xenograft models). Thus, targeted degradation of AR may provide a novel mechanism for providing efficacious therapy for patients with prostate cancer for whom current therapies have failed.
[01031] SPECIFIC EMBODIMENTS
[01032] An aspect of the present disclosure provides a compound having the structure:
[ABM]-[L],
wherein ABM is an androgen receptor (AR) binding moiety, L is a chemical linker moiety, wherein the ABM comprises a structure selected from the group consisting of:
Figure imgf000531_0001
, wherein:
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CN, C^CH, CF3, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
Y1, Y2 are each independently NRY1, O, S;
Y3, Y4, Y5 are each independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2;
Q is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, optionally substituted with 0-6 RQ, each RQ is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C Q
1-6 alkoxyl), or 2 R groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); R1, R2, Ra, Rb, RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched,
optionally substituted by 1 or more halo, C1-6 alkoxyl), or R1, R2 together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); W2 is a bond, C1-6 alkyl, alicyclic, heterocyclic, aryl, heteroaryl, bicyclic, biaryl, biheteroaryl, biheterocyclic, each optionally substituted by 1, 2 or 3 RW2; and
each RW2 is independently H, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1- 3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN. [01033 In an as ect or embodiment described herein, the ABM comprises the structure:
Figure imgf000532_0001
,
wherein:
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, CF3, hydroxyl, nitro, CN, C^CH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
Y3, Y4, Y5 are each independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2;
Q is a 4 membered alicyclic ring with 0-2 heteroatoms, optionally substituted with 0-6 RQ, each RQ is independently H, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl);
W2 is a bond, C1-6 alkyl, alicyclic, heterocyclic, aryl, heteroaryl, bicyclic, biaryl,
biheteroaryl,or biheterocyclic, each optionally substituted by 1, 2 or 3 RW2; and each RW2 is independently H, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1- 3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN.
[01034] In any aspect or embodiment described herein, the [ABM]-[L] compound further comprises an E3 ubiquintin ligase binding moiety (ULM) coupled to the ABM or L or both.
[01035] In any aspect or embodiment described herein, the ULM comprises a hydroxyl prolyl moiety that binds Von Hippel-Lindau (VHL) E3 ubiquitin ligase. [01036] In any aspect or embodiment described herein, the ABM comprises the structure:
Figure imgf000533_0001
wherein:
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CN, CF3, C^CH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
each Y3 is independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2;
Q is a 3-6 alicyclic or aromatic membered ring with 0-4 heteroatoms, optionally substituted with 0-6 RQ, each RQ is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl);
W2 is a bond, C1-6 alkyl, alicyclic, heterocyclic, aryl, or heteroaryl, each optionally
substituted by 1, 2 or 3 RW2; and
each RW2 is independently H, halo, OH, NH2, CN, NRY1RY2, C1-6 alkyl (optionally substituted by 1 or more F), OC1-3alkyl (optionally substituted by 1 or more F).
[01037] In any aspect or embodiment described herein, the ABM comprises the structure:
Figure imgf000533_0002
wherein: W1 is aryl or heteroaryl, each optionally substituted by 1 or more halo, hydroxyl, nitro, CN, CF3,C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo);
each Y3 is independently a bond, O, NRY2, CRY1RY2, or C=O;
each RQ is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
RY1, RY2 are each independently H, OH, or C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl);
W2 is a bond, C1-6 alkyl, aryl, heteroaryl, alicyclic, heterocyclic, each optionally substituted by 1, 2 or 3 RW2; and
each RW2 is independently H, OH, NH2, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1-3alkyl (optionally substituted by 1 or more F).
[01038] Another aspect of the present disclosure provides a bifunctional compound comprising the chemical structure: ABM-L-ULM, wherein ABM is an androgen receptor (AR) binding moiety, L is absent (a bond) or a chemical linker, and ULM is an E3 ubiquitin ligase binding moiety,wherein the ABM comprises a structure selected from the group consisting of:
Figure imgf000534_0001
, wherein:
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CN, CF3, C^CH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
Y1, Y2 are each independently NRY1, O, S;
Y3, Y4, Y5 are each independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2;
Q is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, optionally substituted with 0-6 RQ, each RQ is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); R1, R2, Ra, Rb, RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched,
optionally substituted by 1 or more halo, C1-6 alkoxyl), or R1, R2 together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms;
W2 is a bond, C1-6 alkyl, aryl, heteroaryl, alicyclic, heterocyclic, bicyclic, biaryl, biheteroaryl, or biheterocyclic, each optionally substituted by 1, 2 or 3 RW2; and
each RW2 is independently H, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1- 3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN.
[01039 In an as ect or embodiment described herein, the ABM comprises the structure:
Figure imgf000535_0001
wherein:
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CF3, CN, C^CH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
Y3, Y4, Y5 are each independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2;
Q is a 4 membered alicyclic ring with 0-2 heteroatoms, optionally substituted with 0-6 RQ, each RQ,is independently H, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl);
W2 is a bond, C1-6 alkyl, alicyclic, heterocyclic, aryl, heteroaryl, bicyclic, biheterocyclic, biaryl, or biheteroaryl, each optionally substituted by 1, 2 or 3 RW2; and
each RW2 is independently H, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1- 3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN.
[01040] In any aspect or embodiment described herein, the ABM comprises the structure:
Figure imgf000536_0001
wherein:
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CN, CF3, C^CH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
each Y3 is independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2;
Q is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, optionally substituted with 0-6 RQ, each RQ,is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl);
W2 is a bond, C1-6 alkyl, alicyclic, heterocyclic, aryl, heteroaryl, each optionally substituted by 1, 2 or 3 RW2; and
each RW2 is independently H, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1- 3alkyl (optionally substituted by 1 or more F), OH, NH2, CN, NRY1RY2.
[01041] In any aspect or embodiment described herein, the ABM comprises the structure:
Figure imgf000537_0001
wherein:
W1 is aryl or heteroaryl, each optionally substituted by 1 or more halo, hydroxyl, nitro, CN, CF3, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo);
each Y3 is independently a bond, O, NRY2, CRY1RY2, or C=O;
each RQ is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
RY1, RY2 are each independently H, OH, or C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl);
W2 is a bond, C1-6 alkyl, aryl, or heteroaryl, each optionally substituted by 1, 2 or 3 RW2; and each RW2 is independently H, OH, NH2, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1-3alkyl (optionally substituted by 1 or more F).
[01042] In any aspect or embodiment described herein, the ULM comprises a hydroxyl prolyl moiety that binds Von Hippel-Lindau (VHL) E3 ubiquitin ligase (VLM) comprising the structure ULM-a:
wherein:
Figure imgf000537_0002
a dashed line indicates the attachment of at least one ABM, another ULM or VLM (i.e., ULM’ or VLM’), or a chemical linker moiety coupling at least one ABM, a ULM’ or VLM’ to the other end of the linker; X1, X2 are each independently a bond, O, NRY3, CRY3RY4, C=O, C=S, SO, SO2; RY3, RY4 are each independently H, inear or branched C1-6 alkyl, optionally substituted by 1 or more halo, optionally substituted C1-6 alkoxyl (e.g., optionally substituted with 0-3 RP groups);
RP is 0, 1, 2, or 3 groups, each independently H, halo, -OH, C1-3alkyl;
W3 is an optionally substituted–T-N(R1aR1b),–T-Aryl, an optionally substituted–T- Heteroaryl, an optionally substituted–T-Heterocycle, an optionally substituted -NR1-T- Aryl, an optionally substituted -NR1-T-Heteroaryl or an optionally substituted -NR1-T- Heterocycle, where T is covalently bonded to X1;
each R1 , R1a , R1b is independently H, a C1-C6 alkyl group (linear, branched, optionally
substituted by 1 or more halo, -OH), RY3C=O, RY3C=S, RY3SO, RY3SO2, N(RY3RY4)C=O, N(RY3RY4)C=S, N(RY3RY4)SO, N(RY3RY4)SO2;
W4 is an optionally substituted -NR1-T-Aryl, an optionally substituted -NR1-T-Heteroaryl group or an optionally substituted -NR1-T-Heterocycle, wherein -NR1 is covalently bonded to X2
; R1 is H or CH3, preferably H; and
T is an optionally substituted–(CH2)n- group, wherein each one of the methylene groups may be optionally substituted with one or two substituents, preferably selected from halogen, a C1-C6 alkyl group (linear, branched, optionally substituted by 1 or more halogen, -OH) or the sidechain of an amino acid as otherwise described herein, preferably methyl, which may be optionally substituted; and n is 0 to 6, often 0, 1, 2, or 3, preferably 0.
[01043] In any aspect or embodiment described herein, RY3 and RY4 are each
independently H, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl).
[01044] In any aspect or embodiment described herein, the ULM comprises the structure:
Figure imgf000539_0001
wherein:
W3 is optionally substituted aryl, optionally substituted heteroaryl, or
Figure imgf000539_0002
each R9 and R10 is independently hydrogen, optionally substituted alkyl, optionally
substituted cycloalkyl, optionally substituted hydroxyalkyl, optionally substituted heteroaryl, or haloalkyl; or R9, R10, and the carbon atom to which they are attached form an optionally substituted cycloalkyl;
R11 is optionally substituted heterocyclic, optionally substituted alkoxy, optionally
substituted heteroaryl, optionally substituted aryl, or
Figure imgf000539_0003
Figure imgf000539_0004
R12 is H or optionally substituted alkyl;
R13 is H, optionally substituted alkyl, optionally substituted alkylcarbonyl, optionally
substituted (cycloalkyl)alkylcarbonyl, optionally substituted aralkylcarbonyl, optionally substituted arylcarbonyl, optionally substituted (heterocyclyl)carbonyl, or optionally substituted aralkyl;
R14a, R14b, is each independently H, haloalkyl, or optionally substituted alkyl;
W5 is a phenyl or a 5-10 membered heteroaryl, R15 is H, halogen, CN, OH, NO2, NR14aR14b, OR14a, CONR14aR14b, NR14aCOR14b, SO2NR14aR14b, NR14aSO2R14b, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted haloalkoxy, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted
cycloheteroalkyl,
Figure imgf000540_0001
wherein R17 is H, halo, optionally substituted C3- 6cycloalkyl, optionally substituted C1-6alkyl, optionally substituted C1-6alkenyl, or C1- 6haloalkyl; Xa is S or O;
each R16 is independently halo, optionally substituted alkyl, haloalkyl, hydroxy, optionally substituted alkoxy, or haloalkoxy;
o is 0, 1, 2, 3, or 4;
each R18 is independently halo, optionally substituted alkoxy, cyano, optionally substituted alkyl, haloalkyl, haloalkoxy or a linker; and
p is 0, 1, 2, 3, or 4.
[01045] In an as ect or embodiment described herein the ULM comprises the structure:
Figure imgf000540_0002
,
wherein:
R9 is H;
R10 is iso ro l tert-butyl, sec-butyl, cyclopentyl, or cyclohexyl;
Figure imgf000540_0003
R11 is ; R12 is H;
R13 is H, optionally substituted alkyl, optionally substituted alkylcarbonyl, optionally
substituted (cycloalkyl)alkylcarbonyl, optionally substituted aralkylcarbonyl, optionally substituted arylcarbonyl, optionally substituted (heterocyclyl)carbonyl, or optionally substituted aralkyl;
R14a is H, haloalkyl, methyl, ethyl, isopropyl, cyclopropyl, or C1-C6 alkyl (linear, branched, optionally substituted), each optionally substituted with 1 or more halo, hydroxyl, nitro, CN, C1-C6 alkyl (linear, branched, optionally substituted), or C1-C6 alkoxyl (linear, branched, optionally substituted); and
R15 is
Figure imgf000541_0001
wherein R17 is H, halo, optionally substituted C3-6cycloalkyl, optionally substituted C1-6alkyl, optionally substituted C1-6alkenyl, or C1-6haloalkyl; and
Xa is S or O.
[01046] In any aspect or embodiment described herein, ULM is selected from the group consisting of:
(2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5- yl)benzyl)pyrrolidine-2-carboxamide;
(2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(thiazol-5- yl)benzyl)pyrrolidine-2-carboxamide;
(2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-((S)-1-(4-(4-methylthiazol-5- yl)phenyl)ethyl)pyrrolidine-2-carboxamide;
(2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(oxazol-5- yl)benzyl)pyrrolidine-2-carboxamide hydrochloride;
(2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methyloxazol-5- yl)benzyl)pyrrolidine-2-carboxamide;
(2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-N-(4-chlorobenzyl)-4-hydroxypyrrolidine-2- carboxamide hydrochloride;
(2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-N-(4-cyanobenzyl)-4-hydroxypyrrolidine-2- carboxamide hydrochloride; (2S,4R)-1-((S)-2-amino-3-methylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5- yl)benzyl)pyrrolidine-2-carboxamide hydrochloride;
(2S,4R)-1-((S)-2-amino-3-methylbutanoyl)-4-hydroxy-N-(4-(thiazol-5-yl)benzyl)pyrrolidine- 2-carboxamide hydrochloride;
(2S,4R)-1-((S)-2-amino-3-methylbutanoyl)-4-hydroxy-N-(4-(4-methyloxazol-5- yl)benzyl)pyrrolidine-2-carboxamide hydrochloride;
(2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(1-methyl-1H-pyrazol-5- yl)benzyl)pyrrolidine-2-carboxamide hydrochloride;
(2S,4R)-4-tert-butoxy-N-(2-hydroxy-4-(4-methylthiazol-5-yl)benzyl)-1-((S)-3-methyl-2-(1- oxoisoindolin-2-yl)butanoyl)pyrrolidine-2-carboxamide;
(2S,4R)-4-tert-butoxy-1-((S)-2-(6-fluoro-1-oxoisoindolin-2-yl)-3-methylbutanoyl)-N-(2- hydroxy-4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide;
(2S,4R)-4-tert-butoxy-1-((S)-2-(7-cyano-1-oxoisoindolin-2-yl)-3-methylbutanoyl)-N-(2- hydroxy-4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide; and
(2S,4R)-1-((S)-2-Amino-3,3-dimethylbutanoyl)-4-hydroxy-N-((R)-2-hydroxy-1-(4-(4- methylthiazol-5-yl)phenyl)ethyl)pyrrolidine-2-carboxamide hydrochloride.
[01047] In any aspect or embodiment described herein, the linker group (L) comprises a chemical structural unit represented by the formula:
-Aq- wherein:
q is an integer greater than 1; and
A is independently selected from the group consisting of a bond, CRL1RL2, O, S, SO, SO2, NRL3, SO2NRL3, SONRL3, CONRL3, NRL3CONRL4, NRL3SO2NRL4, CO, CRL1=CRL2, C^C, SiRL1RL2, P(O)RL1, P(O)ORL1, NRL3C(=NCN)NRL4, NRL3C(=NCN), NRL3C(=CNO2)NRL4, C3-11cycloalkyl optionally substituted with 0-6 RL1 and/or RL2 groups, C3-11heteocyclyl optionally substituted with 0-6 RL1 and/or RL2 groups, aryl optionally substituted with 0-6 RL1 and/or RL2 groups, heteroaryl optionally substituted with 0-6 RL1 and/or RL2 groups; wherein
RL1, RL2, RL3, RL4 and RL5 are each, independently, selected from the group consisting of H, halo, C1-8alkyl, OC1-8alkyl, SC1-8alkyl, NHC1-8alkyl, N(C1-8alkyl)2, C3- 11cycloalkyl, aryl, heteroaryl, C3-11heterocyclyl, OC1-8cycloalkyl, SC1-8cycloalkyl, NHC1-8cycloalkyl, N(C1-8cycloalkyl)2, N(C1-8cycloalkyl)(C1-8alkyl), OH, NH2, SH, SO2C1-8alkyl, P(O)(OC1-8alkyl)(C1-8alkyl), P(O)(OC1-8alkyl)2, CC-C1-8alkyl, CCH, CH=CH(C1-8alkyl), C(C1-8alkyl)=CH(C1-8alkyl), C(C1-8alkyl)=C(C1- 8alkyl)2, Si(OH)3, Si(C1-8alkyl)3, Si(OH)(C1-8alkyl)2, COC1-8alkyl, CO2H, halogen, CN, CF3, CHF2, CH2F, NO2, SF5, SO2NHC1-8alkyl, SO2N(C1-8alkyl)2, SONHC1- 8alkyl, SON(C1-8alkyl)2, CONHC1-8alkyl, CON(C1-8alkyl)2, N(C1- 8alkyl)CONH(C1-8alkyl), N(C1-8alkyl)CON(C1-8alkyl)2, NHCONH(C1-8alkyl), NHCON(C1-8alkyl)2, NHCONH2, N(C1-8alkyl)SO2NH(C1-8alkyl), N(C1-8alkyl) SO2N(C1-8alkyl)2, NH SO2NH(C1-8alkyl), NH SO2N(C1-8alkyl)2, and NH SO2NH2; and
wherein when q is greater than 1, RL1 or RL2 each, independently, can be linked to another A group to form cycloalkyl and/or heterocyclyl moeity that can be further substituted with 0-4 RL5 groups.
[01048] In any aspect or embodiment described herein, the linker (L) comprises the following chemical structure:
wherein:
Figure imgf000543_0001
WL1 and WL2 are each independently a 4-8 membered ring with 0-4 heteroatoms, optionally substituted with RQ, each RQ is independently a H, halo, OH, CN, CF3, NH2, carboxyl, C1-C6 alkyl (linear, branched, optionally substituted), C1-C6 alkoxy (linear, branched, optionally substituted), or 2 RQ groups taken together with the atom they are attached to, form a 4-8 membered ring system containing 0-4 heteroatoms;
YL1 is each independently a bond, C1-C6 alkyl (linear, branched, optionally substituted) and optionally one or more C atoms are replaced with O; or C1-C6 alkoxy (linear, branched, optionally substituted);
n is 0-10; and
a dashed line indicates the attachment point to the PTM or ULM moieties. [01049] In any aspect or embodiment described herein, the linker (L) comprises the following chemical structure:
wherein:
Figure imgf000544_0001
WL1 and WL2 are each independently aryl, heteroaryl, cyclic, heterocyclic, C1-6 alkyl (linear, branched, optionally substituted), C1-C6 alkoxy, (linear, branched, optionally substituted), bicyclic, biaryl, biheteroaryl,or biheterocyclic, each optionally substituted with RQ, each RQ is independently a H, halo, OH, CN, CF3, hydroxyl, nitro, C^CH, C2-6 alkenyl, C2-6 alkynyl, C1-C6 alkyl (linear, branched, optionally substituted), C1-C6 alkoxy (linear, branched, optionally substituted), OC1-3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN, or 2 RQ groups taken together with the atom they are attached to, form a 4-8 membered ring system containing 0-4 heteroatoms;
YL1 is each independently a bond, NRYL1, O, S, NRYL2, CRYL1RYL2, C=O, C=S, SO, SO2, C1- C6 alkyl (linear, branched, optionally substituted) and optionally one or more C atoms are replaced with O; C1-C6 alkoxy (linear, branched, optionally substituted);
QL is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, biheterocyclic, or bicylic, optionally bridged, optionally substituted with 0-6 RQ, each RQ is independently H, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
RYL1, RYL2 are each independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or R1, R2 together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
n is 0-10; and
a dashed line indicates the attachment point to the PTM or ULM moieties.
[01050] In any aspect or embodiment described herein, the L is selected from the group consisting of: 2-(3-(5-(tosyloxy)pentyloxy)propoxy)acetic acid;
2-(3-(3,3-dimethyl-5-(tosyloxy)pentyloxy)propoxy)acetic acid;
2-(3-(3-hydroxy-5-(tosyloxy)pentyloxy)propoxy)acetic acid;
2-(2-(2-(2-(tosyloxy)ethoxy)ethoxy)ethoxy)acetic acid;
2-(2-((2R,3R)-3-(2-(tosyloxy)ethoxy)butan-2-yloxy)ethoxy)acetic acid;
2-(2-((2S,3S)-3-(2-(tosyloxy)ethoxy)butan-2-yloxy)ethoxy)acetic acid;
2-(4-(4-(tosyloxy)butoxy)butoxy)acetic acid;
tert-butyl 2-(3-(4-(tosyloxy)butoxy)propoxy)acetate;
tert-butyl 2-(4-(3-(tosyloxy)propoxy)butoxy)acetate;
tert-butyl 2-(6-(tosyloxy)hexa-2,4-diynyloxy)acetate;
tert-butyl 3-(6-(tosyloxy)hexa-2,4-diynyloxy)propanoate;
tert-butyl 4-(6-(tosyloxy)hexa-2,4-diynyloxy)butanoate;
ethyl 2-(2-(2-aminoethoxy)ethoxy)acetate hydrochloride;
ethyl 2-(5-aminopentyloxy)acetate;
methyl 2-(2-(2-(methylamino)ethoxy)ethoxy)acetate;
ethyl 2-(5-(methylamino)pentyloxy)acetate;
2-(3-(2-(tosyloxy)ethoxy)propoxy)acetic acid;
2-(2-hydroxyethoxy)ethyl 4-methylbenzenesulfonate;
ethyl 2-(2-(2-(tosyloxy)ethoxy)ethoxy)acetate;
ethyl 3-(2-(2-(tosyloxy)ethoxy)ethoxy)propanoate;
ethyl 5-(tosyloxy)pentanoate;
ethyl 3-(2-(tosyloxy)ethoxy)propanoate;
ethyl 2-(5-(tosyloxy)pentyloxy)acetate;
ethyl 3-(5-(tosyloxy)pentyloxy)propanoate;
5-hydroxypentyl 4-methylbenzenesulfonate;
ethyl 2-(5-(tosyloxy)pentyloxy)acetate;
ethyl 2-(3-(tosyloxy)propoxy)acetate;
ethyl 2-(2-(tosyloxy)ethoxy)acetate;
ethyl 2-(4-(2-(tosyloxy)ethoxy)butoxy)acetate;
2-(2-(2-hydroxyethoxy)ethoxy)ethyl 4-methylbenzenesulfonate;
2-((2R,3R)-3-(2-hydroxyethoxy)butan-2-yloxy)ethyl 4-methylbenzenesulfonate; 2-(2-piperazin-1-yl)-ethoxy-acetic acid; and
methyl 6-(4-(2-(2-(tert-butoxy)-2-oxoethoxy)ethyl)piperazin-1-yl)nicotinate.
[01051] In any aspect or embodiment described herein, the compound is a member selected from the group consisting of Examples 1-864 (Tables 2-30), a salt, a polymorph, isotopic derivative, and a prodrug thereof.
[01052] In any aspect or embodiment described herein, the compound is selected from the group consisting of:
Figure imgf000546_0001
Figure imgf000547_0001
Figure imgf000548_0001
Figure imgf000549_0001
Figure imgf000550_0001
Figure imgf000551_0001
[01053] Another aspect of the present disclosure provides an androgen receptor bindingcompound comprising a structure of:
wherein:
Figure imgf000551_0002
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CN, C^CH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
Y1, Y2 are each independently NRY1, O, S;
Y3, Y4, Y5 are each independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2;
Q is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, optionally substituted with 0-6 RQ, each RQ,is independently H, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); R1, R2, Ra, Rb, RY1, RY2 are each independently H, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or R1, R2 together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
W2 is a bond, C1-6 alkyl, alicyclic, heterocyclic, aryl, or heteroaryl, each optionally
substituted by 1, 2 or 3 RW2; and
each RW2 is independently H, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1- 3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN.
[01054] In any aspect or embodiment described herein, the compound is selected from the group consisting of:
trans-2^Chloro^4^[3^amino-2,2,4,4-tetramethylcyclobutoxy]benzonitrile;
cis-2-Chloro-4-[3-amino-2,2,4,4-tetramethylcyclobutoxy]benzonitrile;
trans 6-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyridazine-3- carboxamide;
trans tert-Butyl N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]carbamate; trans 4-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]benzamide; trans 5-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyrazine-2- carboxamide;
trans 2-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyrimidine-5- carboxamide;
4-Methoxy-N-[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]benzamide; trans 1-(2-Hydroxyethyl)-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]- 1H-pyrazole-4-carboxamide;
trans 6-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyridine-3- carboxamide;
trans 4-[(5-Hydroxypentyl)amino]-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]benzamide;
trans tert-Butyl 2-({5-[(4-{[3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]carbamoyl}phenyl)aminopentyl}oxy)acetate;
tert-butyl trans-(3-(3-chloro-4-cyanophenoxy)-2,2-dimethylcyclobutyl)carbamate; and tert-butyl cis-(3-(3-chloro-4-cyanophenoxy)-2,2-dimethylcyclobutyl)carbamate.
[01055] An additional aspect of the present disclosure provides a composition comprising an effective amount of a bifunctional compound of the present disclosure, and a
pharmaceutically acceptable carrier.
[01056] In any aspect or embodiment described herein, the composition further comprises at least one additional bioactive agent.
[01057] In any aspect or embodiment described herein, the bioactive agent is an anti- cancer agent.
[01058] A yet further aspect of the present disclosure provides a therapeutic composition comprising an effective amount of at least two different bifunctional compounds according to the present disclosure.
[01059] Another aspect of the present disclosure provides a method of treating a disease or disorder in a subject comprising administering a composition comprising a pharmaceutically acceptable carrier and an effective amount of a compound of the present disclosure to a subject in need thereof, wherein the compound is effective in treating or ameliorating at least one symptom of the disease or disorder.
[01060] In any aspect or embodiment described herein, the disease or disorder is cancer or Kennedy’s Disease or both.
[01061] In any aspect or embodiment described herein, the cancer is prostate cancer.
[01062] In any aspect or embodiment described herein, the composition further comprises an effective amount of at least one additional anti-cancer agent.

Claims

CLAIMS What Is Claimed Is:
1. A compound having the structure:
[ABM]-[L],
wherein ABM is an androgen receptor (AR) binding moiety, L is a chemical linker moiety, wherein the ABM comprises a structure selected from the group consisting of:
Figure imgf000554_0001
, wherein:
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CN, C^CH, CF3, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
Y1, Y2 are each independently NRY1, O, S;
Y3, Y4, Y5 are each independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2;
Q is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, optionally substituted with 0-6 RQ, each RQ is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); R1, R2, Ra, Rb, RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched,
optionally substituted by 1 or more halo, C1-6 alkoxyl), or R1, R2 together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); W2 is a bond, C1-6 alkyl, alicyclic, heterocyclic, aryl, heteroaryl, bicyclic, biaryl, biheteroaryl, biheterocyclic, each optionally substituted by 1, 2 or 3 RW2; and
each RW2 is independently H, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1- 3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN. 2. The com ound of claim 1 wherein the ABM comprises the structure:
Figure imgf000555_0001
,
wherein:
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, CF3, hydroxyl, nitro, CN, C^CH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
Y3, Y4, Y5 are each independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2;
Q is a 4 membered alicyclic ring with 0-2 heteroatoms, optionally substituted with 0-6 RQ, each RQ is independently H, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl);
W2 is a bond, C1-6 alkyl, alicyclic, heterocyclic, aryl, heteroaryl, bicyclic, biaryl,
biheteroaryl,or biheterocyclic, each optionally substituted by 1,
2 or 3 RW2; and each RW2 is independently H, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1- 3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN.
3. The compound of claim 1 or 2, further comprising an E3 ubiquintin ligase binding moiety (ULM) coupled to the ABM or L or both.
4. The compound of claim 3, wherein ULM comprises a hydroxyl prolyl moiety that binds Von Hippel-Lindau (VHL) E3 ubiquitin ligase.
5. The compound of any of claims 1-4, wherein the ABM comprises the structure:
Figure imgf000556_0001
, wherein:
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CN, CF3, C^CH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
each Y3 is independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2;
Q is a 3-6 alicyclic or aromatic membered ring with 0-4 heteroatoms, optionally substituted with 0-6 RQ, each RQ is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl);
W2 is a bond, C1-6 alkyl, alicyclic, heterocyclic, aryl, or heteroaryl, each optionally
substituted by 1, 2 or 3 RW2; and
each RW2 is independently H, halo, OH, NH2, CN, NRY1RY2, C1-6 alkyl (optionally substituted by 1 or more F), OC1-3alkyl (optionally substituted by 1 or more F).
6. The compound of any of claims 1 or 3-5, wherein the ABM comprises the structure:
Figure imgf000556_0002
ABM-d1
wherein:
W1 is aryl or heteroaryl, each optionally substituted by 1 or more halo, hydroxyl, nitro, CN, CF3,C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo);
each Y3 is independently a bond, O, NRY2, CRY1RY2, or C=O;
each RQ is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
RY1, RY2 are each independently H, OH, or C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl);
W2 is a bond, C1-6 alkyl, aryl, heteroaryl, alicyclic, heterocyclic, each optionally substituted by 1, 2 or 3 RW2; and
each RW2 is independently H, OH, NH2, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1-3alkyl (optionally substituted by 1 or more F).
7. A bifunctional compound comprising the chemical structure: ABM-L-ULM, wherein ABM is an androgen receptor (AR) binding moiety, L is absent (a bond) or a chemical linker, and ULM is an E3 ubiquitin ligase binding moiety,wherein the ABM comprises a structure selected from the group consisting of:
Figure imgf000557_0001
wherein: W1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CN, CF3, C^CH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
Y1, Y2 are each independently NRY1, O, S;
Y3, Y4, Y5 are each independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2;
Q is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, optionally substituted with 0-6 RQ, each RQ is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); R1, R2, Ra, Rb, RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched,
optionally substituted by 1 or more halo, C1-6 alkoxyl), or R1, R2 together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms;
W2 is a bond, C1-6 alkyl, aryl, heteroaryl, alicyclic, heterocyclic, bicyclic, biaryl, biheteroaryl, or biheterocyclic, each optionally substituted by 1, 2 or 3 RW2; and
each RW2 is independently H, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1- 3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN.
8. The bifunctional com ound of claim 7, wherein the ABM comprises the structure:
Figure imgf000558_0001
,
wherein:
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CF3, CN, C^CH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
Y3, Y4, Y5 are each independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2; Q is a 4 membered alicyclic ring with 0-2 heteroatoms, optionally substituted with 0-6 RQ, each RQ,is independently H, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl);
W2 is a bond, C1-6 alkyl, alicyclic, heterocyclic, aryl, heteroaryl, bicyclic, biheterocyclic, biaryl, or biheteroaryl, each optionally substituted by 1, 2 or 3 RW2; and
each RW2 is independently H, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1- 3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN.
9. The bifunctional compound of claim 7, wherein the ABM comprises the structure:
Figure imgf000559_0001
, wherein:
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CN, CF3, C^CH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
each Y3 is independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2;
Q is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, optionally substituted with 0-6 RQ, each RQ,is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); RY1, RY2 are each independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl);
W2 is a bond, C1-6 alkyl, alicyclic, heterocyclic, aryl, heteroaryl, each optionally substituted by 1, 2 or 3 RW2; and each RW2 is independently H, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1- 3alkyl (optionally substituted by 1 or more F), OH, NH2, CN, NRY1RY2.
10. The bifunctional compound of claim 7 or 9, wherein the ABM comprises the structure:
Figure imgf000560_0001
ABM-d1
wherein:
W1 is aryl or heteroaryl, each optionally substituted by 1 or more halo, hydroxyl, nitro, CN, CF3, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo);
each Y3 is independently a bond, O, NRY2, CRY1RY2, or C=O;
each RQ is independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
RY1, RY2 are each independently H, OH, or C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl);
W2 is a bond, C 2
1-6 alkyl, aryl, or heteroaryl, each optionally substituted by 1, 2 or 3 RW ; and each RW2 is independently H, OH, NH2, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1-3alkyl (optionally substituted by 1 or more F).
11. The bifunctional compound of any of claims 7-10, wherein ULM comprises a hydroxyl prolyl moiety that binds Von Hippel-Lindau (VHL) E3 ubiquitin ligase (VLM) comprising the structure ULM-a:
Figure imgf000561_0001
,
wherein:
a dashed line indicates the attachment of at least one ABM, another ULM or VLM (i.e., ULM’ or VLM’), or a chemical linker moiety coupling at least one ABM, a ULM’ or VLM’ to the other end of the linker;
X1, X2 are each independently a bond, O, NRY3, CRY3RY4, C=O, C=S, SO, SO2;
RY3, RY4 are each independently H, inear or branched C1-6 alkyl, optionally substituted by 1 or more halo, optionally substituted C1-6 alkoxyl (e.g., optionally substituted with 0-3 RP groups);
RP is 0, 1, 2, or 3 groups, each independently H, halo, -OH, C1-3alkyl;
W3 is an optionally substituted–T-N(R1aR1b),–T-Aryl, an optionally substituted–T- Heteroaryl, an optionally substituted–T-Heterocycle, an optionally substituted -NR1-T- Aryl, an optionally substituted -NR1-T-Heteroaryl or an optionally substituted -NR1-T- Heterocycle, where T is covalently bonded to X1;
each R1 , R1a , R1b is independently H, a C1-C6 alkyl group (linear, branched, optionally
substituted by 1 or more halo, -OH), RY3C=O, RY3C=S, RY3SO, RY3SO2, N(RY3RY4)C=O, N(RY3RY4)C=S, N(RY3RY4)SO, N(RY3RY4)SO2;
W4 is an optionally substituted -NR1-T-Aryl, an optionally substituted -NR1-T-Heteroaryl group or an optionally substituted -NR1-T-Heterocycle, wherein -NR1 is covalently bonded to X2
; R1 is H or CH3, preferably H; and
T is an optionally substituted–(CH2)n- group, wherein each one of the methylene groups may be optionally substituted with one or two substituents, preferably selected from halogen, a C1-C6 alkyl group (linear, branched, optionally substituted by 1 or more halogen, -OH) or the sidechain of an amino acid as otherwise described herein, preferably methyl, which may be optionally substituted; and n is 0 to 6, often 0, 1, 2, or 3, preferably 0.
12. The bifunctional compound of claim 11, wherein the ULM comprises the structure:
Figure imgf000562_0001
,
wherein:
W3 is optionally substituted aryl, optionally substituted heteroaryl, or
Figure imgf000562_0002
;
each R9 and R10 is independently hydrogen, optionally substituted alkyl, optionally
substituted cycloalkyl, optionally substituted hydroxyalkyl, optionally substituted heteroaryl, or haloalkyl; or R9, R10, and the carbon atom to which they are attached form an optionally substituted cycloalkyl;
R11 is optionally substituted heterocyclic, optionally substituted alkoxy, optionally
or
Figure imgf000562_0003
R12 is H or optionally substituted alkyl;
R13 is H, optionally substituted alkyl, optionally substituted alkylcarbonyl, optionally
substituted (cycloalkyl)alkylcarbonyl, optionally substituted aralkylcarbonyl, optionally substituted arylcarbonyl, optionally substituted (heterocyclyl)carbonyl, or optionally substituted aralkyl;
R14a, R14b, is each independently H, haloalkyl, or optionally substituted alkyl;
W5 is a phenyl or a 5-10 membered heteroaryl, R15 is H, halogen, CN, OH, NO2, NR14aR14b, OR14a, CONR14aR14b, NR14aCOR14b,
SO2NR14aR14b, NR14aSO2R14b, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted haloalkoxy, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted
cycloheteroalkyl,
Figure imgf000563_0001
wherein R17 is H, halo, optionally substituted C3- 6cycloalkyl, optionally substituted C1-6alkyl, optionally substituted C1-6alkenyl, or C1- 6haloalkyl; Xa is S or O;
each R16 is independently halo, optionally substituted alkyl, haloalkyl, hydroxy, optionally substituted alkoxy, or haloalkoxy;
o is 0, 1, 2, 3, or 4;
each R18 is independently halo, optionally substituted alkoxy, cyano, optionally substituted alkyl, haloalkyl, haloalkoxy or a linker; and
p is 0, 1, 2, 3, or 4.
13. The bifunctional compound of any of claims 11 or 12, wherein the ULM comprises the structure:
Figure imgf000563_0002
, wherein:
R9 is H;
R10 is isopropyl, tert-butyl, sec-butyl, cyclopentyl, or cyclohexyl; ;
Figure imgf000564_0001
R13 is H, optionally substituted alkyl, optionally substituted alkylcarbonyl, optionally
substituted (cycloalkyl)alkylcarbonyl, optionally substituted aralkylcarbonyl, optionally substituted arylcarbonyl, optionally substituted (heterocyclyl)carbonyl, or optionally substituted aralkyl;
R14a is H, haloalkyl, methyl, ethyl, isopropyl, cyclopropyl, or C1-C6 alkyl (linear, branched, optionally substituted), each optionally substituted with 1 or more halo, hydroxyl, nitro, CN, C1-C6 alkyl (linear, branched, optionally substituted), or C1-C6 alkoxyl (linear, branched o tionally substituted); and
Figure imgf000564_0002
R15 is wherein R17 is H, halo, optionally substituted C3-6cycloalkyl, optionally substituted C1-6alkyl, optionally substituted C1-6alkenyl, or C1-6haloalkyl; and
Xa is S or O.
14. The bifunctional compound of any of claims 11-13, wherein ULM is selected from the group consisting of:
(2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5- yl)benzyl)pyrrolidine-2-carboxamide;
(2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(thiazol-5- yl)benzyl)pyrrolidine-2-carboxamide;
(2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-((S)-1-(4-(4-methylthiazol-5- yl)phenyl)ethyl)pyrrolidine-2-carboxamide;
(2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(oxazol-5- yl)benzyl)pyrrolidine-2-carboxamide hydrochloride;
(2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methyloxazol-5- yl)benzyl)pyrrolidine-2-carboxamide; (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-N-(4-chlorobenzyl)-4-hydroxypyrrolidine-2- carboxamide hydrochloride;
(2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-N-(4-cyanobenzyl)-4-hydroxypyrrolidine-2- carboxamide hydrochloride;
(2S,4R)-1-((S)-2-amino-3-methylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5- yl)benzyl)pyrrolidine-2-carboxamide hydrochloride;
(2S,4R)-1-((S)-2-amino-3-methylbutanoyl)-4-hydroxy-N-(4-(thiazol-5-yl)benzyl)pyrrolidine- 2-carboxamide hydrochloride;
(2S,4R)-1-((S)-2-amino-3-methylbutanoyl)-4-hydroxy-N-(4-(4-methyloxazol-5- yl)benzyl)pyrrolidine-2-carboxamide hydrochloride;
(2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(1-methyl-1H-pyrazol-5- yl)benzyl)pyrrolidine-2-carboxamide hydrochloride;
(2S,4R)-4-tert-butoxy-N-(2-hydroxy-4-(4-methylthiazol-5-yl)benzyl)-1-((S)-3-methyl-2-(1- oxoisoindolin-2-yl)butanoyl)pyrrolidine-2-carboxamide;
(2S,4R)-4-tert-butoxy-1-((S)-2-(6-fluoro-1-oxoisoindolin-2-yl)-3-methylbutanoyl)-N-(2- hydroxy-4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide;
(2S,4R)-4-tert-butoxy-1-((S)-2-(7-cyano-1-oxoisoindolin-2-yl)-3-methylbutanoyl)-N-(2- hydroxy-4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide; and
(2S,4R)-1-((S)-2-Amino-3,3-dimethylbutanoyl)-4-hydroxy-N-((R)-2-hydroxy-1-(4-(4- methylthiazol-5-yl)phenyl)ethyl)pyrrolidine-2-carboxamide hydrochloride.
15. The bifunctional compound of claim 7, wherein the linker group (L) comprises a chemical structural unit represented by the formula:
-Aq- wherein:
q is an integer greater than 1; and
A is independently selected from the group consisting of a bond, CRL1RL2, O, S, SO, SO2, NRL3, SO2NRL3, SONRL3, CONRL3, NRL3CONRL4, NRL3SO2NRL4, CO, CRL1=CRL2, C^C, SiRL1RL2, P(O)RL1, P(O)ORL1, NRL3C(=NCN)NRL4, NRL3C(=NCN), NRL3C(=CNO2)NRL4, C3-11cycloalkyl optionally substituted with 0-6 RL1 and/or RL2 groups, C3-11heteocyclyl optionally substituted with 0-6 RL1 and/or RL2 groups, aryl optionally substituted with 0-6 RL1 and/or RL2 groups, heteroaryl optionally substituted with 0-6 RL1 and/or RL2 groups; wherein
RL1, RL2, RL3, RL4 and RL5 are each, independently, selected from the group consisting of H, halo, C1-8alkyl, OC1-8alkyl, SC1-8alkyl, NHC1-8alkyl, N(C1-8alkyl)2, C3- 11cycloalkyl, aryl, heteroaryl, C3-11heterocyclyl, OC1-8cycloalkyl, SC1-8cycloalkyl, NHC1-8cycloalkyl, N(C1-8cycloalkyl)2, N(C1-8cycloalkyl)(C1-8alkyl), OH, NH2, SH, SO2C1-8alkyl, P(O)(OC1-8alkyl)(C1-8alkyl), P(O)(OC1-8alkyl)2, CC-C1-8alkyl,
Figure imgf000566_0001
8alkyl)2, Si(OH)3, Si(C1-8alkyl)3, Si(OH)(C1-8alkyl)2, COC1-8alkyl, CO2H, halogen, CN, CF3, CHF2, CH2F, NO2, SF5, SO2NHC1-8alkyl, SO2N(C1-8alkyl)2, SONHC1- 8alkyl, SON(C1-8alkyl)2, CONHC1-8alkyl, CON(C1-8alkyl)2, N(C1- 8alkyl)CONH(C1-8alkyl), N(C1-8alkyl)CON(C1-8alkyl)2, NHCONH(C1-8alkyl), NHCON(C1-8alkyl)2, NHCONH2, N(C1-8alkyl)SO2NH(C1-8alkyl), N(C1-8alkyl) SO2N(C1-8alkyl)2, NH SO2NH(C1-8alkyl), NH SO2N(C1-8alkyl)2, and NH SO2NH2; and
wherein when q is greater than 1, RL1 or RL2 each, independently, can be linked to another A group to form cycloalkyl and/or heterocyclyl moeity that can be further substituted with 0-4 RL5 groups.
16. The compound according to any of claims 7-14, wherein the linker (L) comprises the following chemical structure:
Figure imgf000566_0002
,
wherein:
WL1 and WL2 are each independently a 4-8 membered ring with 0-4 heteroatoms, optionally substituted with RQ, each RQ is independently a H, halo, OH, CN, CF3, NH2, carboxyl, C1-C6 alkyl (linear, branched, optionally substituted), C1-C6 alkoxy (linear, branched, optionally substituted), or 2 RQ groups taken together with the atom they are attached to, form a 4-8 membered ring system containing 0-4 heteroatoms; YL1 is each independently a bond, C1-C6 alkyl (linear, branched, optionally substituted) and optionally one or more C atoms are replaced with O; or C1-C6 alkoxy (linear, branched, optionally substituted);
n is 0-10; and
a dashed line indicates the attachment point to the PTM or ULM moieties.
17. The compound according to any of claims 7-14, wherein the linker (L) comprises the following chemical structure:
Figure imgf000567_0001
,
wherein:
WL1 and WL2 are each independently aryl, heteroaryl, cyclic, heterocyclic, C1-6 alkyl (linear, branched, optionally substituted), C1-C6 alkoxy, (linear, branched, optionally substituted), bicyclic, biaryl, biheteroaryl,or biheterocyclic, each optionally substituted with RQ, each RQ is independently a H, halo, OH, CN, CF3, hydroxyl, nitro, C^CH, C2-6 alkenyl, C2-6 alkynyl, C1-C6 alkyl (linear, branched, optionally substituted), C1-C6 alkoxy (linear, branched, optionally substituted), OC1-3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN, or 2 RQ groups taken together with the atom they are attached to, form a 4-8 membered ring system containing 0-4 heteroatoms;
YL1 is each independently a bond, NRYL1, O, S, NRYL2, CRYL1RYL2, C=O, C=S, SO, SO2, C1- C6 alkyl (linear, branched, optionally substituted) and optionally one or more C atoms are replaced with O; C1-C6 alkoxy (linear, branched, optionally substituted);
QL is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, biheterocyclic, or bicylic, optionally bridged, optionally substituted with 0-6 RQ, each RQ is independently H, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); RYL1, RYL2 are each independently H, OH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or R1, R2 together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
n is 0-10; and
a dashed line indicates the attachment point to the PTM or ULM moieties.
18. The bifunctional compound of any of claims 7-15, wherein the L is selected from the group consisting of:
2-(3-(5-(tosyloxy)pentyloxy)propoxy)acetic acid;
2-(3-(3,3-dimethyl-5-(tosyloxy)pentyloxy)propoxy)acetic acid;
2-(3-(3-hydroxy-5-(tosyloxy)pentyloxy)propoxy)acetic acid;
2-(2-(2-(2-(tosyloxy)ethoxy)ethoxy)ethoxy)acetic acid;
2-(2-((2R,3R)-3-(2-(tosyloxy)ethoxy)butan-2-yloxy)ethoxy)acetic acid;
2-(2-((2S,3S)-3-(2-(tosyloxy)ethoxy)butan-2-yloxy)ethoxy)acetic acid;
2-(4-(4-(tosyloxy)butoxy)butoxy)acetic acid;
tert-butyl 2-(3-(4-(tosyloxy)butoxy)propoxy)acetate;
tert-butyl 2-(4-(3-(tosyloxy)propoxy)butoxy)acetate;
tert-butyl 2-(6-(tosyloxy)hexa-2,4-diynyloxy)acetate;
tert-butyl 3-(6-(tosyloxy)hexa-2,4-diynyloxy)propanoate;
tert-butyl 4-(6-(tosyloxy)hexa-2,4-diynyloxy)butanoate;
ethyl 2-(2-(2-aminoethoxy)ethoxy)acetate hydrochloride;
ethyl 2-(5-aminopentyloxy)acetate;
methyl 2-(2-(2-(methylamino)ethoxy)ethoxy)acetate;
ethyl 2-(5-(methylamino)pentyloxy)acetate;
2-(3-(2-(tosyloxy)ethoxy)propoxy)acetic acid;
2-(2-hydroxyethoxy)ethyl 4-methylbenzenesulfonate;
ethyl 2-(2-(2-(tosyloxy)ethoxy)ethoxy)acetate;
ethyl 3-(2-(2-(tosyloxy)ethoxy)ethoxy)propanoate;
ethyl 5-(tosyloxy)pentanoate;
ethyl 3-(2-(tosyloxy)ethoxy)propanoate;
ethyl 2-(5-(tosyloxy)pentyloxy)acetate; ethyl 3-(5-(tosyloxy)pentyloxy)propanoate;
5-hydroxypentyl 4-methylbenzenesulfonate;
ethyl 2-(5-(tosyloxy)pentyloxy)acetate;
ethyl 2-(3-(tosyloxy)propoxy)acetate;
ethyl 2-(2-(tosyloxy)ethoxy)acetate;
ethyl 2-(4-(2-(tosyloxy)ethoxy)butoxy)acetate;
2-(2-(2-hydroxyethoxy)ethoxy)ethyl 4-methylbenzenesulfonate;
2-((2R,3R)-3-(2-hydroxyethoxy)butan-2-yloxy)ethyl 4-methylbenzenesulfonate;
2-(2-piperazin-1-yl)-ethoxy-acetic acid; and
methyl 6-(4-(2-(2-(tert-butoxy)-2-oxoethoxy)ethyl)piperazin-1-yl)nicotinate.
19. The bifunctional compound of claim 7, wherein the compound is a member selected from the group consisting of Examples 1-864 (Tables 2-30), a salt, a polymorph, isotopic derivative, and a prodrug thereof.
20. The bifunctional compound of claim 19, wherein the compound is selected from the group consisting of:
Figure imgf000570_0001
;
Figure imgf000571_0001
;
Figure imgf000572_0001
Figure imgf000573_0001
;
Figure imgf000574_0001
;
Figure imgf000574_0002
;
Figure imgf000574_0003
; OH
Figure imgf000575_0001
21. An androgen receptor bindingcompound comprising a structure of:
Figure imgf000575_0002
,
wherein:
W1 is aryl or heteroaryl, independently substituted by 1 or more halo, hydroxyl, nitro, CN, C^CH, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), C1-6 alkoxyl (linear, branched, optionally substituted by 1 or more halo), C2-6 alkenyl, C2-6 alkynyl;
Y1, Y2 are each independently NRY1, O, S;
Y3, Y4, Y5 are each independently a bond, O, NRY2, CRY1RY2, C=O, C=S, SO, SO2;
Q is a 3-6 membered alicyclic or aromatic ring with 0-4 heteroatoms, optionally substituted with 0-6 RQ, each RQ,is independently H, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or 2 RQ groups taken together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms); R1, R2, Ra, Rb, RY1, RY2 are each independently H, C1-6 alkyl (linear, branched, optionally substituted by 1 or more halo, C1-6 alkoxyl), or R1, R2 together with the atom they are attached to, form a 3-8 membered ring system containing 0-2 heteroatoms);
W2 is a bond, C1-6 alkyl, alicyclic, heterocyclic, aryl, or heteroaryl, each optionally
substituted by 1, 2 or 3 RW2; and
each RW2 is independently H, halo, C1-6 alkyl (optionally substituted by 1 or more F), OC1- 3alkyl (optionally substituted by 1 or more F), OH, NH2, NRY1RY2, CN.
22. The androgen receptor binding compound of claim 21, wherein the compound is selected from the group consisting of:
trans-2-Chloro-4-[3-amino-2,2,4,4-tetramethylcyclobutoxy]benzonitrile;
cis-2-Chloro-4-[3-amino-2,2,4,4-tetramethylcyclobutoxy]benzonitrile;
trans 6-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyridazine-3- carboxamide;
trans tert-Butyl N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]carbamate; trans 4-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]benzamide; trans 5-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyrazine-2- carboxamide;
trans 2-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyrimidine-5- carboxamide;
4-Methoxy-N-[(1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]benzamide;
trans 1-(2-Hydroxyethyl)-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]- 1H-pyrazole-4-carboxamide;
trans 6-Amino-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl]pyridine-3- carboxamide;
trans 4-[(5-Hydroxypentyl)amino]-N-[3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl]benzamide;
trans tert-Butyl 2-({5-[(4-{[3-(3-chloro-4-cyanophenoxy)-2,2,4,4^
tetramethylcyclobutyl]carbamoyl}phenyl)aminopentyl}oxy)acetate; tert-butyl trans-(3-(3-chloro-4-cyanophenoxy)-2,2-dimethylcyclobutyl)carbamate; and tert-butyl cis-(3-(3-chloro-4-cyanophenoxy)-2,2-dimethylcyclobutyl)carbamate.
23. A composition comprising an effective amount of a bifunctional compound of any of claims 7-19, and a pharmaceutically acceptable carrier.
24. The composition of claim 23, wherein the composition further comprises at least one additional bioactive agent.
25. The composition of claim 24, wherein the bioactive agent is an anti-cancer agent.
26. A therapeutic composition comprising an effective amount of at least two different bifunctional compounds according to any of claims 7-19.
27. A method of treating a disease or disorder in a subject comprising the steps of administering a composition comprising a pharmaceutically acceptable carrier and an effective amount of a compound according to any of claims 7-19 to a subject in need thereof, wherein the compound is effective in treating or ameliorating at least one symptom of the disease or disorder.
28. The method of claim 27, wherein the disease or disorder is cancer or Kennedy’s Disease or both.
29. The method of claim 28, wherein the cancer is prostate cancer.
30. The method of claim 29, wherein the composition further comprises an effective amount of at least one additional anti-cancer agent.
PCT/US2018/044051 2017-07-28 2018-07-27 Compounds and methods for the targeted degradation of androgen receptor WO2019023553A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
CA3069544A CA3069544A1 (en) 2017-07-28 2018-07-27 Compounds and methods for the targeted degradation of androgen receptor
BR112020001825-2A BR112020001825A2 (en) 2017-07-28 2018-07-27 compounds and methods for targeted androgen receptor degradation
JP2020504388A JP2020528918A (en) 2017-07-28 2018-07-27 Compounds and methods for targeted degradation of androgen receptors
CN201880062879.9A CN111212835A (en) 2017-07-28 2018-07-27 Compounds and methods for targeted degradation of androgen receptors
RU2020108515A RU2020108515A (en) 2017-07-28 2018-07-27 COMPOUNDS AND METHODS OF TARGETED DEGRADATION OF THE ANDROGENIC RECEPTOR
KR1020207005895A KR20200035435A (en) 2017-07-28 2018-07-27 Compounds and methods for targeted decomposition of androgen receptors
EP18756023.0A EP3658548A1 (en) 2017-07-28 2018-07-27 Compounds and methods for the targeted degradation of androgen receptor
AU2018306606A AU2018306606B2 (en) 2017-07-28 2018-07-27 Compounds and methods for the targeted degradation of Androgen Receptor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US15/663,273 US20170327469A1 (en) 2015-01-20 2017-07-28 Compounds and methods for the targeted degradation of androgen receptor
US15/663,273 2017-07-28

Publications (1)

Publication Number Publication Date
WO2019023553A1 true WO2019023553A1 (en) 2019-01-31

Family

ID=63245020

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/044051 WO2019023553A1 (en) 2017-07-28 2018-07-27 Compounds and methods for the targeted degradation of androgen receptor

Country Status (9)

Country Link
EP (1) EP3658548A1 (en)
JP (1) JP2020528918A (en)
KR (1) KR20200035435A (en)
CN (1) CN111212835A (en)
AU (1) AU2018306606B2 (en)
BR (1) BR112020001825A2 (en)
CA (1) CA3069544A1 (en)
RU (1) RU2020108515A (en)
WO (1) WO2019023553A1 (en)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110903345A (en) * 2019-11-27 2020-03-24 中国药科大学 Micromolecule degradation agent of targeted androgen receptor, preparation method and application thereof
WO2020132561A1 (en) 2018-12-20 2020-06-25 C4 Therapeutics, Inc. Targeted protein degradation
CN111471054A (en) * 2020-05-16 2020-07-31 长春中医药大学 Small-molecule inhibitor for degrading copper ion transporter Atox1 and CCS and application thereof
WO2020211822A1 (en) * 2019-04-18 2020-10-22 成都海创药业有限公司 A class of bifunctional chimeric heterocyclic compounds for targeted degradation of androgen receptors and use thereof
WO2020214555A1 (en) 2019-04-16 2020-10-22 Northwestern University Bifunctional compounds comprising apcin-a and their use in the treatment of cancer
CN112574278A (en) * 2019-09-29 2021-03-30 江西济民可信集团有限公司 Heterocyclic compound as protein degradation agent and preparation method and medical application thereof
WO2021180160A1 (en) * 2020-03-11 2021-09-16 苏州开拓药业股份有限公司 Bifunctional compound, preparation method therefor, and use thereof
CN113582974A (en) * 2020-04-30 2021-11-02 江西济民可信集团有限公司 Compound as protein degradation agent and preparation method and medical application thereof
KR20220014952A (en) 2020-07-29 2022-02-08 한국화학연구원 Compound for inhibiting or degrading androgen receptor and medical uses thereof
CN114181277A (en) * 2020-09-15 2022-03-15 江苏恒瑞医药股份有限公司 Chimeric compound for targeted degradation of androgen receptor protein, preparation method thereof and application thereof in medicine
WO2022165099A3 (en) * 2021-01-29 2022-10-20 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services A new molecular scaffold for targeting hrpn13
WO2022235945A1 (en) 2021-05-05 2022-11-10 Biogen Ma Inc. Compounds for targeting degradation of bruton's tyrosine kinase
WO2023283610A1 (en) 2021-07-07 2023-01-12 Biogen Ma Inc. Compounds for targeting degradation of irak4 proteins
WO2023283372A1 (en) 2021-07-07 2023-01-12 Biogen Ma Inc. Compounds for targeting degradation of irak4 proteins
KR20230112501A (en) 2022-01-20 2023-07-27 주식회사 유빅스테라퓨틱스 Compound having activity of degrading androgen receptor and medical uses thereof
WO2023152182A1 (en) * 2022-02-08 2023-08-17 Saverna Therapeutics Ag Novel compounds modulating mir-155
US11814367B2 (en) 2021-03-15 2023-11-14 Maze Therapeutics, Inc. Inhibitors of glycogen synthase 1 (GYS1) and methods of use thereof
US11826430B2 (en) 2019-05-14 2023-11-28 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
US11834458B2 (en) 2021-03-23 2023-12-05 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
US11952349B2 (en) 2019-11-13 2024-04-09 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111606868B (en) * 2020-06-23 2023-02-17 温州大学新材料与产业技术研究院 Preparation method of bidentate oxazoline chiral ligand
CN114163444B (en) * 2020-09-11 2023-07-14 江苏恒瑞医药股份有限公司 Chimeric compound for androgen receptor protein targeted degradation, preparation method and medical application thereof
CN114262319B (en) * 2020-12-01 2023-05-05 南昌奥瑞药业有限公司 Bifunctional molecule, preparation method and application thereof
CN114853731A (en) * 2021-02-04 2022-08-05 上海长森药业有限公司 Difunctional MDM2 protein degrading agent, and preparation method, pharmaceutical composition and application thereof
CN115772210A (en) * 2021-09-08 2023-03-10 苏州开拓药业股份有限公司 Amorphous substance, crystal, pharmaceutical composition, preparation method and application of thiohydantoin compound or pharmaceutical salt thereof
WO2023093728A1 (en) * 2021-11-23 2023-06-01 江苏先声药业有限公司 Brm selective degradation agent compound and use thereof
WO2024002206A1 (en) * 2022-06-30 2024-01-04 Anhorn Medicines Co., Ltd. Bifunctional compound and pharmaceutical composition comprising the bifunctional compound, and method for treating androgen receptor related diseases by using the same

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US20140356322A1 (en) 2012-01-12 2014-12-04 Yale University Compounds & Methods for the Enhanced Degradation of Targeted Proteins & Other Polypeptides by an E3 Ubiquitin Ligase
EP2985285A1 (en) * 2014-08-11 2016-02-17 Yale University Estrogen-related receptor alpha based protac compounds and associated methods of use
WO2016118666A1 (en) * 2015-01-20 2016-07-28 Arvinas, Inc. Compounds and methods for the targeted degradation of the androgen receptor

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201311888D0 (en) * 2013-07-03 2013-08-14 Glaxosmithkline Ip Dev Ltd Novel compounds
JP6778114B2 (en) * 2014-04-14 2020-10-28 アルビナス・オペレーションズ・インコーポレイテッドArvinas Operations, Inc. Imid-based proteolysis modulators and related uses

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US20140356322A1 (en) 2012-01-12 2014-12-04 Yale University Compounds & Methods for the Enhanced Degradation of Targeted Proteins & Other Polypeptides by an E3 Ubiquitin Ligase
EP2985285A1 (en) * 2014-08-11 2016-02-17 Yale University Estrogen-related receptor alpha based protac compounds and associated methods of use
WO2016118666A1 (en) * 2015-01-20 2016-07-28 Arvinas, Inc. Compounds and methods for the targeted degradation of the androgen receptor

Non-Patent Citations (23)

* Cited by examiner, † Cited by third party
Title
"Compendium of Organic Synthetic Methods", vol. I-VI, WILEY-LNTERSCIENCE
"Remington: The Science and Practice of Pharmacy", 2003, LIPPINCOTT, WILLIAMS & WILKINS
"Remington's Pharmaceutical Sciences"
A. PEPE, J. MED, CHEM., vol. 56, 2013, pages 8280 - 8297
C. GUO, BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, 2012, pages 2572 - 2578
CHUANGXING GUO ET AL: "Discovery of Aryloxy Tetramethylcyclobutanes as Novel Androgen Receptor Antagonists", JOURNAL OF MEDICINAL CHEMISTRY, vol. 54, no. 21, 10 November 2011 (2011-11-10), pages 7693 - 7704, XP055507875, ISSN: 0022-2623, DOI: 10.1021/jm201059s *
END OCR. REV., vol. 8, no. 1, 1987, pages 1 - 28
ENDOCR. REV, vol. 25, no. 2, 2004, pages 276 - 308
G. F. ALLAN, NUCLEAR RECEPTOR SIGNALING, vol. 1, 2003, pages e009
LIEBERMAN, PHARMACEUTICAL DOSAGE FORMS, vol. 1-3, 1992
LLOYD, THE ART, SCIENCE AND TECHNOLOGY OF PHARMACEUTICAL COMPOUNDING, 1999
M. E. JUNG ET AL., J. A/ED. CHEM., vol. 53, 2010, pages 2779 - 2796
MOL. ENDOCRINOL., vol. 16, no. 10, 2002, pages 2181 - 7
NAT. REV. DRUG DISCOVERY, vol. 12, 2013, pages 823 - 824
P. K. POUTIAINEN, J. MED. CHEM., vol. 55, 2012, pages 6316 - 6327
PHARMACOL. REV., vol. 58, no. 4, 2006, pages 782 - 97
PICKAR, DOSAGE CALCULATIONS, 1999
R. H. BRADBURY, BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, 2011, pages 5442 - 5445
R.C. LAROCK: "Comprehensive Organic Transformations,", WILEY-LNTERSCIENCE
T.W. GREENE: "Protective Groups in Organic Chemistry", 1981, JOHN WILEY & SONS
T.W. GREENE; P.G.M. WUTS: "Protective Groups in Organic Chemistry", 1991, JOHN WILEY & SONS
T.W. GREENE; P.G.M. WUTS: "Protective Groups in Organic Chemistry", 1999, JOHN WILEY & SONS
VITAM. HORM., vol. 55, 1999, pages 309 - 52

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020132561A1 (en) 2018-12-20 2020-06-25 C4 Therapeutics, Inc. Targeted protein degradation
WO2020214555A1 (en) 2019-04-16 2020-10-22 Northwestern University Bifunctional compounds comprising apcin-a and their use in the treatment of cancer
AU2020259946B2 (en) * 2019-04-18 2023-05-18 Hinova Pharmaceuticals Inc. A class of bifunctional chimeric heterocyclic compounds for targeted degradation of androgen receptors and use thereof
WO2020211822A1 (en) * 2019-04-18 2020-10-22 成都海创药业有限公司 A class of bifunctional chimeric heterocyclic compounds for targeted degradation of androgen receptors and use thereof
US11826430B2 (en) 2019-05-14 2023-11-28 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
CN112574278A (en) * 2019-09-29 2021-03-30 江西济民可信集团有限公司 Heterocyclic compound as protein degradation agent and preparation method and medical application thereof
US11952349B2 (en) 2019-11-13 2024-04-09 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
CN110903345A (en) * 2019-11-27 2020-03-24 中国药科大学 Micromolecule degradation agent of targeted androgen receptor, preparation method and application thereof
WO2021180160A1 (en) * 2020-03-11 2021-09-16 苏州开拓药业股份有限公司 Bifunctional compound, preparation method therefor, and use thereof
CN113582974A (en) * 2020-04-30 2021-11-02 江西济民可信集团有限公司 Compound as protein degradation agent and preparation method and medical application thereof
CN113582974B (en) * 2020-04-30 2022-05-17 江西济民可信集团有限公司 Compound as protein degradation agent and preparation method and medical application thereof
CN111471054A (en) * 2020-05-16 2020-07-31 长春中医药大学 Small-molecule inhibitor for degrading copper ion transporter Atox1 and CCS and application thereof
KR20220014952A (en) 2020-07-29 2022-02-08 한국화학연구원 Compound for inhibiting or degrading androgen receptor and medical uses thereof
CN114181277A (en) * 2020-09-15 2022-03-15 江苏恒瑞医药股份有限公司 Chimeric compound for targeted degradation of androgen receptor protein, preparation method thereof and application thereof in medicine
WO2022165099A3 (en) * 2021-01-29 2022-10-20 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services A new molecular scaffold for targeting hrpn13
US11814367B2 (en) 2021-03-15 2023-11-14 Maze Therapeutics, Inc. Inhibitors of glycogen synthase 1 (GYS1) and methods of use thereof
US11834458B2 (en) 2021-03-23 2023-12-05 Nuvation Bio Inc. Anti-cancer nuclear hormone receptor-targeting compounds
WO2022235945A1 (en) 2021-05-05 2022-11-10 Biogen Ma Inc. Compounds for targeting degradation of bruton's tyrosine kinase
WO2023283372A1 (en) 2021-07-07 2023-01-12 Biogen Ma Inc. Compounds for targeting degradation of irak4 proteins
WO2023283610A1 (en) 2021-07-07 2023-01-12 Biogen Ma Inc. Compounds for targeting degradation of irak4 proteins
KR20230112501A (en) 2022-01-20 2023-07-27 주식회사 유빅스테라퓨틱스 Compound having activity of degrading androgen receptor and medical uses thereof
WO2023152182A1 (en) * 2022-02-08 2023-08-17 Saverna Therapeutics Ag Novel compounds modulating mir-155

Also Published As

Publication number Publication date
EP3658548A1 (en) 2020-06-03
RU2020108515A (en) 2021-08-27
CA3069544A1 (en) 2019-01-31
JP2020528918A (en) 2020-10-01
BR112020001825A2 (en) 2020-07-21
KR20200035435A (en) 2020-04-03
AU2018306606A1 (en) 2020-02-06
AU2018306606B2 (en) 2021-04-22
CN111212835A (en) 2020-05-29
RU2020108515A3 (en) 2021-08-27

Similar Documents

Publication Publication Date Title
AU2018306606B2 (en) Compounds and methods for the targeted degradation of Androgen Receptor
US11427548B2 (en) Compounds and methods for the targeted degradation of androgen receptor
US20240059686A1 (en) Compounds and methods for the targeted degradation of the androgen receptor
US20210040044A1 (en) Compounds and methods for the targeted degradation of androgen receptor
US20230084249A1 (en) Compounds and methods for the targeted degradation of androgen receptor and associated methods of use
CN112218859A (en) Modulators of proteolysis and related methods of use
CN113164775A (en) Polycyclic compounds and methods for rapid accelerated targeted degradation of fibrosarcoma polypeptides
EP3302482A1 (en) Tank-binding kinase-1 protacs and associated methods of use
WO2019177902A1 (en) Modulators of btk proteolysis and methods of use
RU2774863C2 (en) Compounds and methods for targeted degradation of androgen receptor

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18756023

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3069544

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2020504388

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112020001825

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2018306606

Country of ref document: AU

Date of ref document: 20180727

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20207005895

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2018756023

Country of ref document: EP

Effective date: 20200228

ENP Entry into the national phase

Ref document number: 112020001825

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20200128