WO2023196512A1 - Propargyl compounds and pharmaceutical compositions that modulate brd4 - Google Patents

Propargyl compounds and pharmaceutical compositions that modulate brd4 Download PDF

Info

Publication number
WO2023196512A1
WO2023196512A1 PCT/US2023/017751 US2023017751W WO2023196512A1 WO 2023196512 A1 WO2023196512 A1 WO 2023196512A1 US 2023017751 W US2023017751 W US 2023017751W WO 2023196512 A1 WO2023196512 A1 WO 2023196512A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
heterocyclyl
aryl
heteroaryl
cycloalkyl
Prior art date
Application number
PCT/US2023/017751
Other languages
French (fr)
Inventor
Simon Bailey
Geoffray LERICHE
Original Assignee
Plexium, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Plexium, Inc. filed Critical Plexium, Inc.
Publication of WO2023196512A1 publication Critical patent/WO2023196512A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms 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
    • C07D215/38Nitrogen atoms
    • C07D215/42Nitrogen atoms attached in position 4
    • C07D215/44Nitrogen atoms attached in position 4 with aryl radicals attached to said nitrogen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/36Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings condensed with carbocyclic rings or ring systems
    • C07D241/38Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings condensed with carbocyclic rings or ring systems with only hydrogen or carbon atoms directly attached to the ring nitrogen atoms
    • 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
    • C07D401/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/02Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings
    • C07D407/12Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • Chromatin is a complex combination of DNA and protein that makes up chromosomes. It is found inside the nuclei of eukaryotic cells and is divided between heterochromatin (condensed) and euchromatin (extended) forms. The major components of chromatin are DNA and proteins. Histones are the chief protein components of chromatin, acting as spools around which DNA winds.
  • chromatin The functions of chromatin are to package DNA into a smaller volume to fit in the cell, to strengthen the DNA to allow mitosis and meiosis, and to serve as a mechanism to control expression and DNA replication.
  • the chromatin structure is controlled by a series of post-translational modifications to histone proteins, notably histones H3 and H4, and most commonly within the “histone tails” which extend beyond the core nucleosome structure. Histone tails tend to be free for protein-protein interaction and are also the portion of the histone most prone to post-translational modification. These modifications include acetylation, methylation, phosphorylation, ubiquitinylation, and SUMOylation.
  • epigenetic marks are written and erased by specific enzymes that place the tags on specific residues within the histone tail, thereby forming an epigenetic code, which is then interpreted by the cell to allow gene specific regulation of chromatin structure and thereby transcription.
  • histones are amongst the most susceptible to post-translational modification. Histone modifications are dynamic, as they can be added or removed in response to specific stimuli, and these modifications direct both structural changes to chromatin and alterations in gene transcription.
  • HATs histone acetyltransferases
  • HDACs histone deacetylases
  • acetylate or de-acetylate specific histone lysine residues Struhl K., Genes Dev., 1989, 12, 5, 599-606.
  • HATs histone acetyltransferases
  • HDACs histone deacetylases
  • acetylate or de-acetylate specific histone lysine residues Struhl K., Genes Dev., 1989, 12, 5, 599-606
  • Bromodomains which are approximately 110 amino acids long, are found in a large number of chromatin-associated proteins and have been identified in approximately 70 human proteins, often adjacent to other protein motifs (Jeanmougin F., et al, Trends Biochem. Sci., 1997, 22, 5,151-153; and Tamkun J. W., et al., Cell, 1992, 7, 3, 561-572).
  • Bromodomain-containing proteins have been implicated in disease processes including cancer, inflammation and viral replication. See, e.g., Prinjha et al, Trends Pharm. Sci., 33(3):146-153 (2012) and Muller et al, Expert Rev., 13(29):1-20 (2011).
  • Cell-type specificity and proper tissue functionality requires the tight control of distinct transcriptional programs that are intimately influenced by their environment. Alterations to this transcriptional homeostasis are directly associated with numerous disease states, most notably cancer, immuno-inflammation, neurological disorders, and metabolic diseases.
  • Bromodomains reside within key chromatin modifying complexes that serve to control distinctive disease-associated transcriptional pathways. This is highlighted by the observation that mutations in bromodomain-containing proteins are linked to cancer, as well as immune and neurologic dysfunction. Moreover, recent findings have demonstrated that small molecule inhibition of the bromodomains of BRD4 may have clinical utility in diverse human diseases, ranging from auto-immunity to cardiac hypertrophy. This is possible because the underlying mechanism resides in transcriptional regulation. Hence, the inhibition of bromodomains across the family creates varied opportunities as novel therapeutic agents in human dysfunction. [0007] Accordingly, the ability to modulate or degrade BRD4 would be a significant advancement in treating cancer and other bromodomain related diseases.
  • the disclosed compounds that bind to or degrade BRD4 are represented by formula I: or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein Q, X 1 , X 2 , Y, R 1 , R 2 , R 3 , R 4 , m, and n are as defined herein.
  • the disclosed compounds that bind to or degrade BRD4 are represented by formula I-1: or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein Q, X 1 , X 2 , R 1 , R 2 , R 3 , R 4 , m, and n are as defined herein.
  • the compounds as described herein degrade BRD4 by 30% or more at 1 ⁇ M concentration.
  • this disclosure provides a pharmaceutical composition comprising a pharmaceutically acceptable excipient and an effective amount of a compound of formula I, formula I-1, or any subformula thereof.
  • this disclosure provides a method for modulating or degrading BRD4, which method comprises contacting BRD4 with an effective amount of a compound of formula I, formula I-1, or any subformula thereof under conditions wherein BRD4 is bound to said compound and modulated or degraded.
  • this disclosure provides a method for modulating or degrading BRD4 in a subject, which method comprises administering to said subject an effective amount of a compound of formula I, formula I-1, or any subformula thereof or a pharmaceutical composition comprising a pharmaceutically acceptable excipient and an effective amount of a compound of formula I, formula I-1, or any subformula thereof under conditions wherein BRD4 is bound to said compound and modulated or degraded.
  • this disclosure provides a method for treating cancer in a subject in need thereof which method comprises administering to said subject an effective amount of a compound of formula I, formula I-1, or any subformula thereof, or a pharmaceutical composition comprising a pharmaceutically acceptable excipient and an effective amount of a compound of formula I, formula I-1, or any subformula thereof.
  • This disclosure provides for compounds, pharmaceutical compositions comprising such compounds, and methods of using such compounds and compositions to treat diseases, disorders, or conditions mediated, at least in part, by BRD4 transcription factors.
  • a wavy line or a dashed line drawn through a line in a structure indicates a specified point of attachment of a group. Unless chemically or structurally required, no directionality or stereochemistry is indicated or implied by the order in which a chemical group is written or named. [0019]
  • the prefix “Cu-v” indicates that the following group has from u to v carbon atoms.
  • C1-6 alkyl indicates that the alkyl group has from 1 to 6 carbon atoms.
  • compositions and methods include the recited elements, but not excluding others.
  • Consisting essentially of when used to define compositions and methods, shall mean excluding other elements of any essential significance to the combination for the stated purpose.
  • composition consisting essentially of the elements as defined herein would not exclude other materials or steps that do not materially affect the basic and novel characteristic(s) of the claimed disclosure.
  • Consisting of shall mean excluding more than trace elements of other ingredients and substantial method steps. Embodiments defined by each of these transition terms are within the scope of this disclosure.
  • Alkyl refers to an unbranched or branched saturated hydrocarbon chain.
  • alkyl has 1 to 20 carbon atoms (i.e., C 1-20 alkyl), 1 to 12 carbon atoms (i.e., C 1-12 alkyl), 1 to 8 carbon atoms (i.e., C1-8 alkyl), 1 to 6 carbon atoms (i.e., C1-6 alkyl), or 1 to 4 carbon atoms (i.e., C1-4 alkyl).
  • alkyl groups include, e.g., methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, pentyl, 2-pentyl, isopentyl, neopentyl, hexyl, 2-hexyl, 3-hexyl, and 3-methylpentyl.
  • butyl includes n-butyl (i.e., -(CH2)3CH3), sec-butyl (i.e., -CH(CH3)CH2CH3), isobutyl (i.e., -CH2CH(CH3)2), and tert-butyl (i.e., -C(CH3)3); and “propyl” includes n-propyl (i.e., -(CH2)2CH3) and isopropyl (i.e., -CH(CH3)2).
  • a divalent group such as a divalent “alkyl” group, a divalent “aryl” group, a divalent heteroaryl group, etc.
  • a divalent group such as a divalent “alkyl” group, a divalent “aryl” group, a divalent heteroaryl group, etc.
  • an “alkylene” group or an “alkylenyl” group for example, methylenyl, ethylenyl, and propylenyl
  • an “arylene” group or an “arylenyl” group for example, phenylenyl or napthylenyl, or quinolinyl for heteroarylene
  • Alkenyl refers to an alkyl group containing at least one (e.g., 1-3, or 1) carbon-carbon double bond and having from 2 to 20 carbon atoms (i.e., C2-20 alkenyl), 2 to 12 carbon atoms (i.e., C2-12 alkenyl), 2 to 8 carbon atoms (i.e., C2-8 alkenyl), 2 to 6 carbon atoms (i.e., C2-6 alkenyl), or 2 to 4 carbon atoms (i.e., C2-4 alkenyl).
  • alkenyl groups include, e.g., ethenyl, propenyl, butadienyl (including 1,2-butadienyl and 1,3-butadienyl).
  • Alkynyl refers to an alkyl group containing at least one (e.g., 1-3, or 1) carbon-carbon triple bond and having from 2 to 20 carbon atoms (i.e., C 2-20 alkynyl), 2 to 12 carbon atoms (i.e., C 2-12 alkynyl), 2 to 8 carbon atoms (i.e., C 2-8 alkynyl), 2 to 6 carbon atoms (i.e., C 2-6 alkynyl), or 2 to 4 carbon atoms (i.e., C 2-4 alkynyl).
  • alkynyl also includes those groups having one triple bond and one double bond.
  • Alkoxy refers to the group “alkyl-O-”. Examples of alkoxy groups include, e.g., methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, and 1,2-dimethylbutoxy.
  • Alkylthio refers to the group “alkyl-S-”.
  • Alkylsulfinyl refers to the group “alkyl-S(O)-”.
  • Alkylsulfonyl refers to the group “alkyl-S(O)2-”. “Alkylsulfonylalkyl” refers to -alkyl-S(O)2-alkyl. [0030] “Acyl” refers to a group -C(O)R y , wherein R y is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein.
  • acyl examples include, e.g., formyl, acetyl, cyclohexylcarbonyl, cyclohexylmethyl- carbonyl, and benzoyl.
  • “Amido” refers to both a “C-amido” group which refers to the group -C(O)NR y R z and an “N-amido” group which refers to the group -NR y C(O)R z , wherein R y and R z are independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein, or R y and R z are taken together to form a cycloalkyl or heterocyclyl; each of which may be unsubstituted or substituted, as defined herein.
  • Amino refers to the group -NR y R z wherein R y and R z are independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein.
  • Amidino refers to -C(NR y )(NR z 2), wherein R y and R z are independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein.
  • Aryl refers to an aromatic carbocyclic group having a single ring (e.g., monocyclic) or multiple rings (e.g., bicyclic or tricyclic) including fused systems.
  • aryl has 6 to 20 ring carbon atoms (i.e., C6-20 aryl), 6 to 12 carbon ring atoms (i.e., C6-12 aryl), or 6 to 10 carbon ring atoms (i.e., C6-10 aryl).
  • aryl groups include, e.g., phenyl, naphthyl, fluorenyl, and anthryl.
  • Aryl does not encompass or overlap in any way with heteroaryl defined below. If one or more aryl groups are fused with a heteroaryl, the resulting ring system is heteroaryl regardless of point of attachment.
  • the resulting ring system is heterocyclyl regardless of point of attachment. If one or more aryl groups are fused with a cycloalkyl, the resulting ring system is cycloalkyl regardless of point of attachment.
  • Carbamoyl refers to both an “O-carbamoyl” group which refers to the group -O-C(O)NR y R z and an “N-carbamoyl” group which refers to the group -NR y C(O)OR z , wherein R y and R z are independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein.
  • Carboxyl ester or “ester” refer to both -OC(O)R x and -C(O)OR x , wherein R x is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein.
  • Cycloalkyl refers to a saturated or partially unsaturated cyclic alkyl group having a single ring or multiple rings including fused, bridged, and spiro ring systems.
  • cycloalkyl includes cycloalkenyl groups (i.e., the cyclic group having at least one double bond) and carbocyclic fused ring systems having at least one sp 3 carbon atom (i.e., at least one non-aromatic ring).
  • cycloalkyl has from 3 to 20 ring carbon atoms (i.e., C3-20 cycloalkyl), 3 to 14 ring carbon atoms (i.e., C3-14 cycloalkyl), 3 to 12 ring carbon atoms (i.e., C3-12 cycloalkyl), 3 to 10 ring carbon atoms (i.e., C3-10 cycloalkyl), 3 to 8 ring carbon atoms (i.e., C3-8 cycloalkyl), or 3 to 6 ring carbon atoms (i.e., C3-6 cycloalkyl).
  • Monocyclic groups include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • Polycyclic groups include, for example, bicyclo[2.2.1]heptanyl, bicyclo[2.2.2]octanyl, adamantyl, norbornyl, decalinyl, 7,7-dimethyl-bicyclo[2.2.1]heptanyl, and the like.
  • cycloalkyl is intended to encompass any non-aromatic ring which may be fused to an aryl ring, regardless of the attachment to the remainder of the molecule.
  • cycloalkyl also includes “spirocycloalkyl” when there are two positions for substitution on the same carbon atom, for example spiro[2.5]octanyl, spiro[4.5]decanyl, or spiro[5.5]undecanyl.
  • “Imino” refers to a group -C(NR y )R z , wherein R y and R z are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein.
  • “Imido” refers to a group -C(O)NR y C(O)R z , wherein R y and R z are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein.
  • “Halogen” or “halo” refers to atoms occupying group VIIA of the periodic table, such as fluoro, chloro, bromo, or iodo.
  • Haloalkyl refers to an unbranched or branched alkyl group as defined above, wherein one or more (e.g., 1 to 6 or 1 to 3) hydrogen atoms are replaced by a halogen.
  • a residue is substituted with more than one halogen, it may be referred to by using a prefix corresponding to the number of halogen moieties attached.
  • Dihaloalkyl and trihaloalkyl refer to alkyl substituted with two (“di”) or three (“tri”) halo groups, which may be, but are not necessarily, the same halogen.
  • haloalkyl examples include, e.g., trifluoromethyl, difluoromethyl, fluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1,2-difluoroethyl, 3-bromo-2-fluoropropyl, 1,2-dibromoethyl, and the like.
  • Haloalkoxy refers to an alkoxy group as defined above, wherein one or more (e.g., 1 to 6 or 1 to 3) hydrogen atoms are replaced by a halogen.
  • Hydroxyalkyl refers to an alkyl group as defined above, wherein one or more (e.g., 1 to 6 or 1 to 3) hydrogen atoms are replaced by a hydroxy group.
  • “Heteroalkyl” refers to an alkyl group in which one or more of the carbon atoms (and any associated hydrogen atoms), excluding any terminal carbon atom(s), are each independently replaced with the same or different heteroatomic group, provided the point of attachment to the remainder of the molecule is through a carbon atom.
  • the term “heteroalkyl” includes unbranched or branched saturated chain having carbon and heteroatoms.
  • 1, 2 or 3 carbon atoms may be independently replaced with the same or different heteroatomic group.
  • Heteroatomic groups include, but are not limited to, -NR y -, -O-, -S-, -S(O)-, -S(O)2-, and the like, wherein R y is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein.
  • heteroalkyl groups include, e.g., ethers (e.g., -CH2OCH3, -CH(CH3)OCH3, -CH2CH2OCH3, -CH2CH2OCH2CH2OCH3, etc.), thioethers (e.g., -CH2SCH3, -CH(CH3)SCH3, -CH2CH2SCH3,-CH2CH2SCH2CH2SCH3, etc.), sulfones (e.g., -CH2S(O)2CH3, -CH(CH3)S(O)2CH3, -CH2CH2S(O)2CH3, -CH2CH2S(O)2CH2OCH3, etc.), and amines (e.g., -CH2NR y CH3, -CH(CH3)NR y CH3, -CH2CH2NR y CH3, -CH2CH2NR y CH2CH2NR y CH3, etc., where R y is hydrogen, alkyl
  • heteroalkyl includes 2 to 10 carbon atoms, 2 to 8 carbon atoms, or 2 to 4 carbon atoms; and 1 to 3 heteroatoms, 1 to 2 heteroatoms, or 1 heteroatom.
  • “Heteroaryl” refers to an aromatic group having a single ring, multiple rings or multiple fused rings, with one or more ring heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • heteroaryl includes 1 to 20 ring carbon atoms (i.e., C1-20 heteroaryl), 3 to 12 ring carbon atoms (i.e., C3-12 heteroaryl), or 3 to 8 carbon ring atoms (i.e., C3-8 heteroaryl), and 1 to 5 ring heteroatoms, 1 to 4 ring heteroatoms, 1 to 3 ring heteroatoms, 1 to 2 ring heteroatoms, or 1 ring heteroatom independently selected from nitrogen, oxygen, and sulfur.
  • ring carbon atoms i.e., C1-20 heteroaryl
  • 3 to 12 ring carbon atoms i.e., C3-12 heteroaryl
  • 3 to 8 carbon ring atoms i.e., C3-8 heteroaryl
  • 1 to 5 ring heteroatoms 1 to 4 ring heteroatoms, 1 to 3 ring heteroatoms, 1 to 2 ring heteroatoms, or 1 ring heteroatom independently selected from nitrogen, oxygen, and sulfur.
  • heteroaryl includes 5-10 membered ring systems, 5-7 membered ring systems, or 5-6 membered ring systems, each independently having 1 to 4 ring heteroatoms, 1 to 3 ring heteroatoms, 1 to 2 ring heteroatoms, or 1 ring heteroatom independently selected from nitrogen, oxygen, and sulfur.
  • heteroaryl groups include, e.g., acridinyl, benzimidazolyl, benzothiazolyl, benzindolyl, benzofuranyl, benzothiazolyl, benzothiadiazolyl, benzonaphthofuranyl, benzoxazolyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[1,2-a]pyridyl, carbazolyl, cinnolinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, isoquinolyl, isoxazolyl, naphthyridinyl, oxadiazolyl, oxazolyl, 1-oxidopyridinyl, 1-oxidopyrimidinyl, 1-oxide
  • fused-heteroaryl rings include, but are not limited to, benzo[d]thiazolyl, quinolinyl, isoquinolinyl, benzo[b]thiophenyl, indazolyl, benzo[d]imidazolyl, pyrazolo[1,5-a]pyridinyl, and imidazo[1,5-a]pyridinyl, where the heteroaryl can be bound via either ring of the fused system. Any aromatic ring, having a single or multiple fused rings, containing at least one heteroatom, is considered a heteroaryl regardless of the attachment to the remainder of the molecule (i.e., through any one of the fused rings).
  • Heteroaryl does not encompass or overlap with aryl as defined above.
  • “Heterocyclyl” – used interchangeably with “heterocycloalkyl”- refers to a saturated or partially unsaturated cyclic alkyl group, with one or more ring heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • the term “heterocyclyl” includes heterocycloalkenyl groups (i.e., the heterocyclyl group having at least one double bond), bridged-heterocyclyl groups, fused-heterocyclyl groups, and spiro-heterocyclyl groups.
  • Any non-aromatic ring containing at least one heteroatom is considered a heterocyclyl, regardless of the attachment (i.e., can be bound through a carbon atom or a heteroatom).
  • the term heterocyclyl is intended to encompass any non-aromatic ring containing at least one heteroatom, which ring may be fused to a cycloalkyl, an aryl, or heteroaryl ring, regardless of the attachment to the remainder of the molecule.
  • heterocyclyl has 2 to 20 ring carbon atoms (i.e., C2-20 heterocyclyl), 2 to 12 ring carbon atoms (i.e., C2-12 heterocyclyl), 2 to 10 ring carbon atoms (i.e., C2-10 heterocyclyl), 2 to 8 ring carbon atoms (i.e., C2-8 heterocyclyl), 3 to 12 ring carbon atoms (i.e., C3-12 heterocyclyl), 3 to 8 ring carbon atoms (i.e., C3-8 heterocyclyl), or 3 to 6 ring carbon atoms (i.e., C3-6 heterocyclyl); having 1 to 5 ring heteroatoms, 1 to 4 ring heteroatoms, 1 to 3 ring heteroatoms, 1 to 2 ring heteroatoms, or 1 ring heteroatom independently selected from nitrogen, sulfur, or oxygen.
  • ring carbon atoms i.e., C2-20 heterocyclyl
  • 2 to 12 ring carbon atoms i.
  • heterocyclyl groups include, e.g., azetidinyl, azepinyl, benzodioxolyl, benzo[b][1,4]dioxepinyl, 1,4- benzodioxanyl, benzopyranyl, benzodioxinyl, benzopyranonyl, benzofuranonyl, dioxolanyl, dihydropyranyl, hydropyranyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, furanonyl, imidazolinyl, imidazolidinyl, indolinyl, indolizinyl, isoindolinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-ox
  • heterocyclyl also includes “spiroheterocyclyl” when there are two positions for substitution on the same carbon atom.
  • spiro-heterocyclyl rings include, e.g., bicyclic and tricyclic ring systems, such as oxabicyclo[2.2.2]octanyl, 2-oxa-7-azaspiro[3.5]nonanyl, 2-oxa-6-azaspiro[3.4]octanyl, and 6-oxa-1-azaspiro[3.3]heptanyl.
  • fused-heterocyclyl rings include, but are not limited to, 1,2,3,4-tetrahydroisoquinolinyl, 4,5,6,7-tetrahydrothieno[2,3- c]pyridinyl, indolinyl, and isoindolinyl, where the heterocyclyl can be bound via either ring of the fused system.
  • “Sulfonyl” refers to the group -S(O)2R y , where R y is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein.
  • Examples of sulfonyl are methylsulfonyl, ethylsulfonyl, phenylsulfonyl, and toluenesulfonyl.
  • “Sulfinyl” refers to the group -S(O)R y , where R y is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein.
  • R y is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein.
  • Examples of sulfinyl are methylsulfinyl, ethylsulfinyl, phenylsulfinyl, and toluenesulfinyl.
  • “Sulfonamido” refers to the groups -SO2NR y R z and -NR y SO2R z , where R y and R z are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein.
  • the terms “optional” or “optionally” means that the subsequently described event or circumstance may or may not occur and that the description includes instances where said event or circumstance occurs and instances in which it does not.
  • the term “unsubstituted or substituted” refers to any one or more (e.g., 1 to 5 or 1 to 3) hydrogen atoms on the designated atom or group may or may not be replaced by a moiety other than hydrogen.
  • substituted used herein means any of the above groups (i.e., alkyl, alkenyl, alkynyl, alkylene, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, aryl, heterocyclyl, heteroaryl, and/or heteroalkyl) wherein at least one (e.g., 1 to 5 or 1 to 3) hydrogen atom is replaced by a bond to a non-hydrogen atom such as, but not limited to alkyl, alkenyl, alkynyl, alkoxy, alkylthio, acyl, amido, amino, amidino, aryl, aralkyl, azido, carbamoy
  • substituted includes any of the above alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl groups in which one or more (e.g., 1 to 5 or 1 to 3) hydrogen atoms are independently replaced with deuterium, halo, cyano, nitro, azido, oxo, alkyl, alkenyl, alkynyl, haloalkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -NR g R h , -NR g C(O)R h , -NR g C(O)NR g R h , -NR g C(O)OR h , -NR g S(O)1-2R h , -C(O)R g , -C(O)OR g , -OC(O)OR g , -
  • substituted also means any of the above groups in which one or more (e.g., 1 to 5 or 1 to 3) hydrogen atoms are replaced with -C(O)R g , -C(O)OR g , -C(O)NR g R h , -CH2SO2R g , or -CH2SO2NR g R h .
  • R g and R h are the same or different and independently hydrogen, alkyl, alkenyl, alkynyl, alkoxy, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, and/or heteroarylalkyl.
  • substituted also means any of the above groups in which one or more (e.g., 1 to 5 or 1 to 3) hydrogen atoms are replaced by a bond to an amino, cyano, hydroxy, imino, nitro, oxo, thioxo, halo, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, and/or heteroarylalkyl, or two of R g and R h are taken together with the atoms to which they are attached to form a heterocyclyl ring unsubstituted or substituted with oxo, halo, or alkyl unsubstituted or substituted with oxo, halo, amino, hydroxy, or alk
  • impermissible substitution patterns e.g., methyl substituted with 5 fluorines or heteroaryl groups having two adjacent oxygen ring atoms. Such impermissible substitution patterns are well known to the skilled artisan.
  • substituted may describe other chemical groups defined herein.
  • the phrase “one or more” refers to one to five. In certain embodiments, as used herein, the phrase “one or more” refers to one to three.
  • Any compound or structure given herein, is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds.
  • isotopically enriched analogs These forms of compounds may also be referred to as “isotopically enriched analogs.” Isotopically labeled compounds have structures depicted herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Examples of isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 31 P, 32 P, 35 S, 18 F, 36 Cl, 123 I, and 125 I, respectively.
  • isotopically labeled compounds of the present disclosure for example those into which radioactive isotopes such as 3 H and 14 C are incorporated.
  • Such isotopically labelled compounds may be useful in metabolic studies, reaction kinetic studies, detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays or in radioactive treatment of patients.
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • the term “isotopically enriched analogs” includes “deuterated analogs” of compounds described herein in which one or more hydrogens is/are replaced by deuterium, such as a hydrogen on a carbon atom.
  • Such compounds exhibit increased resistance to metabolism and are thus useful for increasing the half-life of any compound when administered to a mammal, particularly a human. See, for example, Foster, “Deuterium Isotope Effects in Studies of Drug Metabolism,” Trends Pharmacol. Sci.5(12): 524-527 (1984). Such compounds are synthesized by means well known in the art, for example by employing starting materials in which one or more hydrogens have been replaced by deuterium. [0059] Deuterium labelled or substituted therapeutic compounds of the disclosure may have improved DMPK (drug metabolism and pharmacokinetics) properties, relating to absorption, distribution, metabolism, and excretion (ADME).
  • ADME absorption, distribution, metabolism, and excretion
  • isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life, reduced dosage requirements, and/or an improvement in therapeutic index.
  • An 18 F, 3 H, or 11 C labeled compound may be useful for PET or SPECT or other imaging studies.
  • Isotopically labeled compounds of this disclosure and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent. It is understood that deuterium in this context is regarded as a substituent in a compound described herein.
  • the concentration of such a heavier isotope, specifically deuterium may be defined by an isotopic enrichment factor.
  • any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom.
  • a position is designated specifically as “H” or “hydrogen,” the position is understood to have hydrogen at its natural abundance isotopic composition.
  • any atom specifically designated as a deuterium (D) is meant to represent deuterium.
  • the compounds of this disclosure are capable of forming acid and/or base salts by virtue of the presence of amino, and/or carboxyl groups, or groups similar thereto.
  • “Pharmaceutically acceptable” or “physiologically acceptable” refer to compounds, salts, compositions, dosage forms, and other materials which are useful in preparing a pharmaceutical composition that is suitable for veterinary or human pharmaceutical use.
  • pharmaceutically acceptable salt of a given compound refers to salts that retain the biological effectiveness and properties of the given compound and which are not biologically or otherwise undesirable.
  • physiologically acceptable salts include, for example, salts with inorganic acids, and salts with an organic acid.
  • the free base can be obtained by basifying a solution of the acid salt.
  • an addition salt particularly a pharmaceutically acceptable addition salt, may be produced by dissolving the free base in a suitable organic solvent and treating the solution with an acid, in accordance with conventional procedures for preparing acid addition salts from base compounds.
  • Pharmaceutically acceptable acid addition salts may be prepared from inorganic or organic acids.
  • Salts derived from inorganic acids include, e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Salts derived from organic acids include, e.g., acetic acid, propionic acid, gluconic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluene-sulfonic acid, salicylic acid, and the like.
  • salts derived from inorganic bases include, by way of example only, sodium, potassium, lithium, aluminum, ammonium, calcium, and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, such as alkyl amines (i.e., NH2(alkyl)), dialkyl amines (i.e., HN(alkyl)2), trialkyl amines (i.e., N(alkyl)3), substituted alkyl amines (i.e., NH2(substituted alkyl)), di(substituted alkyl) amines (i.e., HN(substituted alkyl)2), tri(substituted alkyl) amines (i.e., N(substituted alkyl)3), alkenyl amines
  • alkyl amines i.e., NH2(
  • Suitable amines include, by way of example only, isopropylamine, trimethyl amine, diethyl amine, tri(iso-propyl) amine, tri(n-propyl) amine, ethanolamine, 2-dimethylaminoethanol, piperazine, piperidine, morpholine, N-ethylpiperidine, and the like.
  • isopropylamine trimethyl amine, diethyl amine, tri(iso-propyl) amine, tri(n-propyl) amine, ethanolamine, 2-dimethylaminoethanol, piperazine, piperidine, morpholine, N-ethylpiperidine, and the like.
  • Tautomers are in equilibrium with one another.
  • amide containing compounds may exist in equilibrium with imidic acid tautomers.
  • the compounds are understood by one of ordinary skill in the art to comprise both amide and imidic acid tautomers.
  • the amide containing compounds are understood to include their imidic acid tautomers.
  • the imidic acid containing compounds are understood to include their amide tautomers.
  • the compounds, or their pharmaceutically acceptable salts include an asymmetric center and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids.
  • Optically active (+) and (-), (R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and/or fractional crystallization.
  • Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC).
  • a “stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable.
  • the present disclosure contemplates various stereoisomers, or mixtures thereof, and includes “enantiomers,” which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another.
  • “Diastereomers” are stereoisomers that have at least two asymmetric atoms, but which are not mirror-images of each other.
  • Prodrugs means any compound which releases an active parent drug according to a structure described herein in vivo when such prodrug is administered to a mammalian subject. Prodrugs of a compound described herein are prepared by modifying functional groups present in the compound described herein in such a way that the modifications may be cleaved in vivo to release the parent compound.
  • Prodrugs may be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds.
  • Prodrugs include compounds described herein wherein a hydroxy, amino, carboxyl, or sulfhydryl group in a compound described herein is bonded to any group that may be cleaved in vivo to regenerate the free hydroxy, amino, or sulfhydryl group, respectively.
  • prodrugs include, but are not limited to esters (e.g., acetate, formate, and benzoate derivatives), amides, guanidines, carbamates (e.g., N,N-dimethylaminocarbonyl) of hydroxy functional groups in compounds described herein, and the like.
  • esters e.g., acetate, formate, and benzoate derivatives
  • amides e.g., acetate, formate, and benzoate derivatives
  • carbamates e.g., N,N-dimethylaminocarbonyl
  • Preparation, selection, and use of prodrugs is discussed in T. Higuchi and V. Stella, “Pro-drugs as Novel Delivery Systems,” Vol. 14 of the A.C.S. Symposium Series; “Design of Prodrugs,” ed. H. Bundgaard, Elsevier, 1985; and in Bioreversible Carriers in Drug Design, ed. Edward B.
  • a BRD4 “modulator” may bind to BRD4, but not degrade BRD4.
  • a BRD4 “degrader” binds to BRD4 and also degrades BRD4.
  • this disclosure provides a compound of formula I: or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein: m is an integer from 0 to 4; n is an integer from 0 to 4; Y is CH(R 4 ), NR 5 , or O; each of X 1 and X 2 is independently CH(R 4 ) or NR 5 ; Q is H, -CH 3 , -CH 2 OH, -CH(CH 3 )N(R 13 ) 2 , -CH 2 N(R 13 ) 2 , -CH 2 N(R 13 )C(O)R 1 , or -CH 2 CH 2 N(R 13 ) 2 ; R 1 is C 1 -C 6 alkyl, C 3- C 10 cycloalkyl, aryl, heteroaryl, or heterocyclyl, wherein each C 1 -C 6 alkyl, C 3- C 10 cycloalkyl, ary
  • this disclosure provides a compound of formula I as described above, provided that the compound at 1 ⁇ M concentration degrades BRD4 by 30% or more.
  • this disclosure provides a compound of formula IA: or a pharmaceutically a cceptable salt, solvate, stereoisomer, or tautomer thereof, wherein: m is an integer from 0 to 4; n is an integer from 0 to 4; Y is CH(R 4 ), NR 5 , or O; each of X 1 and X 2 is independently CH(R 4 ) or NR 5 ; each R 2 is independently selected from halo, -N(R 13 ) 2 , -OR 14 , C 1 -C 6 alkyl, aryl, cyano, C 3- C 10 cycloalkyl, heteroaryl, heterocyclyl, guanidino, and nitro, wherein each C 1 -C 6 alkyl, aryl, C 3- C 10
  • the compound of formula I is represented by formula IB:
  • the compound of formula IB is represented by formula IC: or a pharmaceutically accep table salt, solvate, stereoisomer, or tautomer thereof, wherein R 2 , R 3 , R 4 , m, and n are as defined herein.
  • the compound of formula IC is represented by formula ID: or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein R 2 , R 3 , R 4 , and n are as defined herein.
  • this disclosure provides a compound of formula I, IA, IB, IC, or ID, wherein each R 4 independently is C 1 -C 6 alkyl.
  • this disclosure provides a compound of formula ID, wherein R 4 is C1-C6 alkyl and n is 0.
  • the compound of formula ID is represented by formula IE: NH or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein R 2 is as defined herein.
  • this disclosure provides a compound of formula I, IA, IB, or IC, wherein each R 2 independently is H, halo, C 1 -C 6 alkyl, phenyl substituted with C 1 -C 6 alkoxy, pyridyl, or 1-methylpyrazol-5-yl.
  • this disclosure provides a compound of formula ID or IE, wherein each R 2 independently is H, halo, C 1 -C 6 alkyl, phenyl substituted with C 1 -C 6 alkoxy, pyridyl, or 1-methylpyrazol-5-yl. [0080] In some embodiments, this disclosure provides a compound selected from Table 1 or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof.
  • this disclosure provides a compound selected from Table 2, or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof.
  • Compounds of Table 2 degrade BRD4 by less than 30% at 1 ⁇ M concentration.
  • this disclosure provides a compound of formula I-1: or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein: m is an integer from 0 to 4; n is an integer from 0 to 4; each of X 1 and X 2 is independently CH(R 4 ) or NR 5 ; Q is H, -CH 2 OH, -CH 2 N(R 13 ) 2 , -CH 2 CH 2 N(R 13 ) 2 , -CH 2 N(R 13 )C(O)R 1 , or heterocyclyl; R 1 is C1-C6 alkyl, C3-C10 cycloalkyl, aryl, heteroaryl, or heterocyclyl, wherein each C1-C6 alkyl, C 3- C 10 cycloalkyl, aryl, heteroaryl, or heterocyclyl is independently unsubstituted or substituted with one to five Z 1a ; each of
  • this disclosure provides a compound of formula I-1 as described above, provided that the compound at 1 ⁇ M concentration degrades BRD4 by 30% or more. [0084] In some embodiments, this disclosure provides a compound of formula IA-1:
  • each of X 1 and X 2 is independently CH(R 4 ) or NR 5 ; each of R 2 and R 3 is independently selected from halo, -N(R 13 )2, -OR 14 , C1-C6 alkyl, aryl, cyano, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, guanidino, and nitro, wherein each C1-C6 alkyl, aryl, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, and guanidino is independently unsubstituted or substituted with one to five Z 1a ; each R 4 is independently H, halo, cyano, hydroxy, -SH, -N(R 13 )2, -NO2, -SF
  • the compound of formula I-1 is represented by formula IB-1: [0086] or a pharmaceutically ac ceptable salt, solvate, stereoisomer, or tautomer thereof, wherein R 2 , R 3 , R 4 , m, and n are as defined herein. [0087] In some embodiments, the compound of formula IB-1 is represented by formula IC-1:
  • this disclosure provides a compound of formula I-1, IA-1, IB-1, or IC-1, wherein each R 4 independently is C1-C6 alkyl. In some embodiments, this disclosure provides a compound of formula I-1, IA-1, IB-1, or IC-1, wherein each R 4 independently is methyl. In some embodiments, this disclosure provides a compound of formula I-1, IA-1, IB-1, or IC-1, wherein each R 4 independently is hydrogen.
  • this disclosure provides a compound of formula I-1, IA-1, IB-1, or IC-1, wherein one R 4 is methyl and the other R 4 is hydrogen. In some embodiments, this disclosure provides a compound of formula IB-1, wherein R 4 is C1-C6 alkyl and n is 0. [0089] In some embodiments, this disclosure provides a compound of formula I-1, IA-1, or IB-1, wherein each R 2 independently is H, halo, C1-C6 alkyl, phenyl substituted with C1-C6 alkoxy, pyridyl, or 1-methylpyrazol-5-yl. [0090] In some embodiments, this disclosure provides a compound selected from Table 1A or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof. Table 1A Compound # Structure Name
  • this disclosure provides a compound selected from Table 2A, or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof.
  • Compounds of Table 2A degrade BRD4 by less than 30% at 1 ⁇ M concentration.
  • Table 2A Compound # Structure Name
  • X 1 , X 2 , R 1 , R 2 , R 3 , R 4 , Y, Q, m, and n are as defined throughout the specification.
  • X is a suitable Suzuki cross-coupling partner for a boronic ester (including, but not limited to, Br, Cl, I, triflate, and the like).
  • the first step is an Ullmann N-Arylation reaction, wherein at least a stoichiometric equivalent of a suitable amine, compound 1, is combined with a suitably functionalized aryl halide, compound 2, in an inert diluent such as tetrahydrofuran, DMF, DMSO, and the like, typically in the presence of a copper(I) catalyst (e.g. CuI) and a suitable base such as cesium carbonate, potassium carbonate, and the like.
  • a copper(I) catalyst e.g. CuI
  • a suitable base such as cesium carbonate, potassium carbonate, and the like.
  • the ester, compound 3 may be converted to a primary amide (Journal of Medicinal Chemistry, 2005, vol.48, # 25, p.8045 - 8054), wherein at least a stoichiometric equivalent of ammonia or ammonium hydroxide is combined with compound 3, in an inert diluent, such as MeOH, EtOH, THF, and the like.
  • the reaction is typically maintained at from 60° to 100°C until it is substantially complete.
  • the reaction is typically maintained at from 20° to 60°C for a period of time sufficient for substantial completion of the reaction, as evidenced by e.g., thin layer chromatography.
  • conventional workup of the reaction solution can be followed by isolation / purification processes, such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 5.
  • the isopropylcarbonyl group is illustrative only and other conventional amino protecting groups, such as benzyl, t-butoxycarbonyl (Boc), benzyloxycarbonyl (Cbz), p-nitrobenzyloxycarbonyl, and the like could be used.
  • compound 5 is cyclized under conventional reaction conditions (Journal of Medicinal Chemistry, 2014, vol.57, # 19, p.8111 – 8131) well known in the art, including the use of sodium tetrahydroborate, magnesium chloride, and lithium tert-butoxide.
  • the reaction is typically conducted in an inert solvent such as ethanol, THF, toluene, N,N-dimethylformamide, and the like.
  • the reaction is typically conducted at from about -10o to about 20o C for a period of time sufficient for substantial completion of the reaction, as evidenced by e.g., thin layer chromatography.
  • reaction completion Upon reaction completion, conventional workup of the reaction solution can be followed by isolation / purification processes, such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 6.
  • isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 6.
  • HPLC high performance liquid chromatography
  • at least a stoichiometric amount of a suitably substituted carboxylic acid or carboxylic halide compound is combined with compound 6 under conventional amidation reaction conditions well known in the art, including the use of N,N-dicyclohexylcarbodiimide (DCC) as an activation agent for the carboxyl group.
  • DCC N,N-dicyclohexylcarbodiimide
  • Other activation agents are well known in the art.
  • the reaction is typically conducted in an inert solvent, such as chloroform, methylene chloride, toluene, N,N- dimethylformamide, and the like.
  • the reaction is typically conducted at from about 0o to about 30o C for a period of time sufficient for substantial completion of the reaction, as evidenced by e.g., thin layer chromatography.
  • conventional workup of the reaction solution can be followed by isolation / purification processes, such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 7.
  • the isopropylcarbonyl protecting group is removed by conventional conditions, such as aluminum trichloride in dichloromethane, to provide for compound 8.
  • the isopropylcarbonyl group is illustrative only and other conventional amino protecting groups, such as benzyl, t-butoxycarbonyl (Boc), benzyloxycarbonyl (Cbz), p-nitrobenzyloxycarbonyl, and the like could be used.
  • Boc benzyl, t-butoxycarbonyl
  • Cbz benzyloxycarbonyl
  • p-nitrobenzyloxycarbonyl and the like
  • conventional workup of the reaction solution can be followed by isolation / purification processes, such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 8.
  • a conventional Suzuki coupling reaction wherein at least a stoichiometric equivalent of a suitable boronic acid is combined with compound 10, in an inert diluent, such as tetrahydrofuran, dioxane, toluene, dimethoxyethane, and the like, typically in the presence of a palladium catalyst (e.g, palladium diacetate) and a suitable base, such as diisopropylethylamine, triethylamine, pyridine, potassium carbonate, and the like.
  • a palladium catalyst e.g, palladium diacetate
  • a suitable base such as diisopropylethylamine, triethylamine, pyridine, potassium carbonate, and the like.
  • reaction is typically maintained at from 10° to 65°C until it is substantially complete.
  • Conventional workup of the reaction solution can be followed by isolation / purification processes, such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 11.
  • HPLC high performance liquid chromatography
  • at least a stoichiometric amount of compound 11 is combined with a terminal alkyne under conventional Sonogashira coupling conditions, in an inert diluent such as tetrahydrofuran, dioxane, DMSO, DMF, and the like, typically in the presence of a palladium catalyst (e.g.
  • reaction is typically conducted at from about 30o to about 120o C for a period of time sufficient for substantial completion of the reaction, as evidenced by e.g., thin layer chromatography.
  • suitable base such as diisopropylethylamine, triethylamine, pyridine, potassium carbonate, and the like.
  • the reaction is typically conducted at from about 30o to about 120o C for a period of time sufficient for substantial completion of the reaction, as evidenced by e.g., thin layer chromatography.
  • conventional workup of the reaction solution can be followed by isolation / purification processes, such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compounds of formula I.
  • Scheme 1A illustrates a general method for preparing compounds of formula I-1.
  • substituents and variables X 1 , X 2 , R 1 , R 2 , and m are as defined throughout the specification.
  • X is a suitable Suzuki cross-coupling partner for a boronic ester (including, but not limited to, Br, Cl, I, triflate, and the like).
  • the first step is a conventional SNAR reaction wherein at least a stoichiometric equivalent of compound 1A, is combined with compound 2A, in an inert diluent such as tetrahydrofuran, dioxane, DMSO, DMF, and the like, typically in the presence of a suitable base such as diisopropylethylamine, triethylamine, pyridine, potassium carbonate, and the like.
  • the reaction is typically maintained at from 25 °C to 100 °C until it is substantially complete.
  • reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 3A.
  • compound 3A is reduced under conventional nitro reduction reaction conditions well known in the art including the use Fe in the presence of ammonium chloride.
  • Other nitro reducing reagents are well known in the art.
  • the reaction is typically conducted in an inert solvent such as EtOH, ethyl acetate, toluene, and the like.
  • the reaction is typically conducted from about 20 oC to about 60 oC for a period of time sufficient for substantial completion of the reaction as evidenced by e.g., thin layer chromatography.
  • reaction solution Upon reaction completion, conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 4A.
  • the amine is activated as a tosyl sulfonamide. At least a stoichiometric amount of tosyl chloride is added to compound 4A, in an inert diluent such as THF, MeCN, toluene and the like in the presence of a suitable base such as triethylamine, diisopropylethylamine, pyridine and the like.
  • the reaction is typically maintained at from 0 °C to 30 °C until it is substantially complete.
  • reaction is typically maintained at from 0 °C to 30 °C until it is substantially complete.
  • Conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 6A.
  • HPLC high performance liquid chromatography
  • at least a stoichiometric amount of a suitably carboxylic acid compound is combined with compound 6A under conventional amidation reaction conditions (Angewandte Chemie- International Edition, 2020, 59, 3028-3032) well known in the art, including the use of N,N- dicyclohexylcarbodiimide (DCC), as an activation agent for the carboxyl group.
  • DCC N,N- dicyclohexylcarbodiimide
  • Other activation agents are well known in the art.
  • the reaction is typically conducted in an inert solvent such as chloroform, methylene chloride, toluene, N,N-dimethylformamide, and the like.
  • the reaction is typically conducted at from about 0 oC to about 30 oC for a period of time sufficient for substantial completion of the reaction as evidenced by e.g., thin layer chromatography.
  • conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 7A.
  • the tosyl (Ts) protecting group is removed by conventional conditions to provide for compound 8A.
  • reaction completion conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like.
  • HPLC high performance liquid chromatography
  • the amine 8A is protected with t-butoxycarbonyl (BOC) group with.
  • BOC t-butoxycarbonyl
  • At least a stoichiometric amount of a BOC anhydride is combined with compound 8A in an inert diluent such as dichloromethane, dichloroethane, THF, and the like.
  • the reaction is typically maintained at from about 20 °C to about 50 °C until it is substantially complete.
  • a conventional Suzuki coupling reaction wherein at least a stoichiometric equivalent of compound 10A, is combined with a suitable aryl halide, in an inert diluent such as tetrahydrofuran, dioxane, toluene, dimethoxyethane, and the like, typically in the presence of a palladium catalyst (e.g, palladium diacetate) and a suitable base such as diisopropylethylamine, triethylamine, pyridine, potassium carbonate, and the like.
  • a palladium catalyst e.g, palladium diacetate
  • a suitable base such as diisopropylethylamine, triethylamine, pyridine, potassium carbonate, and the like.
  • the t-butoxycarbonyl (BOC) protecting group is removed by conventional conditions.
  • the BOC group is illustrative only and other conventional amino blocking groups such as benzyl, 9-fluorenylmethoxycarbonyl (Fmoc), benzyloxycarbonyl (Cbz), p-nitrobenzyloxycarbonyl and the like.
  • a suitable base such as diisopropylethylamine, triethylamine, pyridine, potassium carbonate, and the like.
  • the reaction is typically maintained at from 80 °C to 120 °C until it is substantially complete.
  • Conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 13A.
  • At least a stoichiometric amount of compound 13A is combined with a terminal alkyne under conventional Sonogashira coupling conditions, in an inert diluent such as tetrahydrofuran, dioxane, DMSO, DMF, and the like, typically in the presence of a palladium catalyst (e.g, Bis(triphenylphosphine)palladium(II) dichloride), a copper(I) co-catalyst (e.g. CuI), and a suitable base such as diisopropylethylamine, triethylamine, pyridine, potassium carbonate, and the like.
  • a palladium catalyst e.g, Bis(triphenylphosphine)palladium(II) dichloride
  • a copper(I) co-catalyst e.g. CuI
  • a suitable base such as diisopropylethylamine, triethylamine,
  • reaction is typically conducted at from about 30 oC to about 120 oC for a period of time sufficient for substantial completion of the reaction as evidenced by e.g., thin layer chromatography.
  • conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 14A.
  • Other variants of compound 12A are commercially available or can be prepared by conventional reaction conditions well known in the art.
  • Other starting materials used herein are either well known in the art, commercially available, or can be prepared by conventional synthetic methods.
  • the compounds and compositions described herein are useful in methods for treating a BRD4 dependent disease or disorder or a disease or disorder that is mediated, at least in part by, BRD4.
  • the methods comprise administering to a subject suffering from a BRD4 dependent disease or disorder an effective amount of a compound, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof or a pharmaceutical composition comprising said compound, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof as described herein.
  • a compound, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof or a pharmaceutical composition comprising said compound, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof as described herein for use in treating an BRD4 dependent disease or disorder.
  • the method relates a compound, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof or a pharmaceutical composition comprising said compound, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof as described herein for use in manufacture of a medicament for reducing BRD4 protein levels where reduction of such protein levels treats or ameliorates the diseases or disorder.
  • the method relates a compound, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof or a pharmaceutical composition comprising said compound, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof as described herein for use as described herein, wherein the BRD4 degradation at 1 ⁇ M concentration of the compounds described herein is at least 50% or at least 70%.
  • the BRD4 degradation is measured by the assay described in the biological example.
  • BRD4 dependent diseases or disorders such as liposarcoma, glioblastoma, bladder cancer, adrenocortical cancer, multiple myeloma, colorectal cancer, non-small cell lung cancer, Human Papilloma Virus-associated cervical, oropharyngeal, penis, anal, thyroid, or vaginal cancer or Epstein-Barr Virus-associated nasopharyngeal carcinoma, gastric cancer, rectal cancer, thyroid cancer, Hodgkin lymphoma or diffuse large B-cell lymphoma.
  • the cancer may be selected from prostate cancer, breast carcinoma, lymphomas, leukemia, myeloma, bladder carcinoma, colon cancer, cutaneous melanoma, hepatocellular carcinoma, endometrial cancer, ovarian cancer, cervical cancer, lung cancer, renal cancer, glioblastoma multiform, glioma, thyroid cancer, parathyroid tumor, nasopharyngeal cancer, tongue cancer, pancreatic cancer, esophageal cancer, cholangiocarcinoma, gastric cancer, soft tissue sarcomas, rhabdomyosarcoma (RMS), synovial sarcoma, osteosarcoma, rhabdoid cancers, cancer for which the immune response is deficient, an immunogenic cancer, and Ewing’s sarcoma.
  • the BRD4-dependent disease or disorder is a disease or disorder is selected from non-small cell lung cancer (NSCLC), melanoma, triple-negative breast cancer (TNBC), nasopharyngeal cancer (NPC), microsatellite stable colorectal cancer (mssCRC), thymoma, carcinoid, and gastrointestinal stromal tumor (GIST).
  • NSCLC non-small cell lung cancer
  • TNBC triple-negative breast cancer
  • NPC nasopharyngeal cancer
  • mssCRC microsatellite stable colorectal cancer
  • thymoma thymoma
  • carcinoid gastrointestinal stromal tumor
  • the cancer is selected from non-small cell lung cancer (NSCLC), melanoma, triple-negative breast cancer (TNBC), nasopharyngeal cancer (NPC), microsatellite stable colorectal cancer (mssCRC), thymoma, carcinoid, acute myelogenous leukemia, and gastrointestinal stromal tumor (GIST).
  • the BRD4-dependent disease or disorder is a disease or disorder is selected from non-small cell lung cancer (NSCLC), melanoma, triple- negative breast cancer (TNBC), nasopharyngeal cancer (NPC), and microsatellite stable colorectal cancer (mssCRC).
  • the compounds of the disclosure can be administered in effective amounts to treat or prevent a disorder and/or prevent the development thereof in subjects.
  • methods of using the compounds of the present application comprise administering to a subject in need thereof an effective amount of a compound as described herein.
  • compounds as described herein are useful in the treatment of proliferative disorders (e.g., cancer, benign neoplasms, inflammatory disease, and autoimmune diseases).
  • levels of cell proteins of interest e.g., pathogenic and oncogenic proteins are modulated, or their expression is inhibited or the proteins are modulated or degraded by contacting said cells with a compound or composition, as described herein.
  • the compounds are useful in treating cancer.
  • methods for the treatment of cancer comprising administering an effective amount of compound or composition, as described herein, to a subject in need thereof.
  • a method for the treatment of cancer comprising administering an effective amount of a compound, or a pharmaceutical composition comprising a compound as described herein to a subject in need thereof, in such amounts and for such time as is necessary to achieve the desired result.
  • the compounds of present application are administered orally.
  • the compounds and compositions, according to the method of the present application are administered orally to a subject using any amount and any route of administration effective for killing or inhibiting the growth of tumor cells.
  • the expression “amount effective to kill or inhibit the growth of tumor cells,” as used herein, refers to a sufficient amount of agent to kill or inhibit the growth of tumor cells.
  • the method involves the administration of a therapeutically effective amount of the compound or a pharmaceutically acceptable salt thereof to a subject (including, but not limited to a human or other mammal in need of it.
  • the compounds or compositions described herein are useful for the treatment of cancer (including, but not limited to, glioblastoma, retinoblastoma, breast cancer, cervical cancer, colon and rectal cancer, leukemia, lymphoma, lung cancer (including, but not limited to small cell lung cancer), melanoma and/or skin cancer, multiple myeloma, non-Hodgkin's lymphoma, ovarian cancer, pancreatic cancer, prostate cancer and gastric cancer, bladder cancer, uterine cancer, kidney cancer, testicular cancer, stomach cancer, brain cancer, liver cancer, or esophageal cancer).
  • cancer including, but not limited to, glioblastoma, retinoblastoma, breast cancer, cervical cancer, colon and rectal cancer, leukemia, lymphoma, lung cancer (including, but not limited to small cell lung cancer), melanoma and/or skin cancer, multiple myeloma, non-Hodgkin's lymphoma, ova
  • the compounds or compositions described herein are useful in the treatment of cancers and other proliferative disorders, including, but not limited to breast cancer, cervical cancer, colon and rectal cancer, leukemia, lung cancer, melanoma, multiple myeloma, non-Hodgkin's lymphoma, ovarian cancer, pancreatic cancer, prostate cancer, and gastric cancer.
  • compounds or compositions described herein are active against solid tumors.
  • the present application provides pharmaceutically acceptable derivatives of the compounds, and methods of treating a subject using these compounds, pharmaceutical compositions thereof, or either of these in combination with one or more additional therapeutic agents.
  • Another aspect of the application relates to a method of treating or lessening the severity of a disease or condition associated with a proliferation disorder in a patient, said method comprising a step of administering to said patient, a compound of Formula I or Formula I-1, or a composition comprising said compound.
  • the compounds and compositions, according to the method of the present application may be administered using any amount and any route of administration effective for the treatment of cancer and/or disorders associated with cell hyperproliferation.
  • the present application provides methods for the treatment of a proliferative disorder in a subject in need thereof by administering to a subject in need of such treatment, a therapeutically effective amount of a compound of the present application, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof.
  • the proliferative disorder can be cancer or a precancerous condition.
  • the present application further provides the use of a compound of the present application, or a pharmaceutically acceptable salt, salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof, for the preparation of a medicament useful for the treatment of a proliferative disorder.
  • the present application also provides methods of protecting against a proliferative disorder in a subject in need thereof by administering a therapeutically effective amount of compound of the present application, or a pharmaceutically acceptable salt, salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof, to a subject in need of such treatment.
  • the proliferative disorder can be cancer or a precancerous condition.
  • the present application also provides the use of compound of the present application, or a pharmaceutically acceptable salt, salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof, for the preparation of a medicament useful for the prevention of a proliferative disorder.
  • proliferative disorder refers to conditions in which unregulated or abnormal growth, or both, of cells can lead to the development of an unwanted condition or disease, which may or may not be cancerous.
  • exemplary proliferative disorders of the application encompass a variety of conditions wherein cell division is deregulated.
  • Exemplary proliferative disorders include, but are not limited to, neoplasms, benign tumors, malignant tumors, uterine fibroids, pre-cancerous conditions, in situ tumors, encapsulated tumors, metastatic tumors, liquid tumors, solid tumors, immunological tumors, hematological tumors, cancers, carcinomas, leukemias, lymphomas, sarcomas, and rapidly dividing cells.
  • a proliferative disorder includes a precancer or a precancerous condition.
  • a proliferative disorder includes cancer.
  • the methods provided herein are used to treat or alleviate a symptom of cancer.
  • cancer includes solid tumors, as well as, hematologic tumors and/or malignancies.
  • precancer cell or “precancerous cell” is a cell manifesting a proliferative disorder that is a precancer or a precancerous condition.
  • cancer cell or “cancerous cell” is a cell manifesting a proliferative disorder that is a cancer. Any reproducible means of measurement may be used to identify cancer cells or precancerous cells. Cancer cells or precancerous cells can be identified by histological typing or grading of a tissue sample (e.g., a biopsy sample). Cancer cells or precancerous cells can be identified through the use of appropriate molecular markers.
  • non-cancerous conditions or disorders include, but are not limited to, rheumatoid arthritis; inflammation; autoimmune disease; lymphoproliferative conditions; acromegaly; rheumatoid spondylitis; osteoarthritis; gout, other arthritic conditions; sepsis; septic shock; endotoxic shock; gram- negative sepsis; toxic shock syndrome; asthma; adult respiratory distress syndrome; chronic obstructive pulmonary disease; chronic pulmonary inflammation; inflammatory bowel disease; Crohn's disease; psoriasis; eczema; ulcerative colitis; pancreatic fibrosis; hepatic fibrosis; acute and chronic renal disease; irritable bowel syndrome; pyresis; restenosis; cerebral malaria; stroke and ischemic injury; neural trauma; Alzheimer's disease; Huntington's disease; Parkinson's disease; acute and chronic pain; allergic rhinitis; allergic conjunctivitis; chronic
  • Exemplary cancers include, but are not limited to, adrenocortical carcinoma, AIDS-related cancers, AIDS-related lymphoma, anal cancer, anorectal cancer, cancer of the anal canal, appendix cancer, childhood cerebellar astrocytoma, childhood cerebral astrocytoma, basal cell carcinoma, skin cancer (non-melanoma), biliary cancer, extrahepatic bile duct cancer, intrahepatic bile duct cancer, bladder cancer, urinary bladder cancer, bone and joint cancer, osteosarcoma and malignant fibrous histiocytoma, brain cancer, brain tumor, brain stem glioma, cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumors, visual pathway and hypothalamic glioma, breast cancer, bronchial adeno
  • a “proliferative disorder of the hematologic system” is a proliferative disorder involving cells of the hematologic system.
  • a proliferative disorder of the hematologic system can include lymphoma, leukemia, myeloid neoplasms, mast cell neoplasms, myelodysplasia, benign monoclonal gammopathy, lymphomatoid granulomatosis, lymphomatoid papulosis, polycythemia vera, chronic myelocytic leukemia, agnogenic myeloid metaplasia, and essential thrombocythemia.
  • a proliferative disorder of the hematologic system can include hyperplasia, dysplasia, and metaplasia of cells of the hematologic system.
  • the compositions of the present application may be used to treat a cancer selected from the group consisting of a hematologic cancer of the present application or a hematologic proliferative disorder of the present application.
  • a hematologic cancer of the present application can include multiple myeloma, lymphoma (including Hodgkin's lymphoma, non-Hodgkin's lymphoma, childhood lymphomas, and lymphomas of lymphocytic and cutaneous origin), leukemia (including childhood leukemia, hairy-cell leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, chronic myelogenous leukemia, and mast cell leukemia), myeloid neoplasms and mast cell neoplasms.
  • lymphoma including Hodgkin's lymphoma, non-Hodgkin's lymphoma, childhood lymphomas, and lymphomas of lymphocytic and cutaneous origin
  • leukemia including childhood leukemia, hairy-cell leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, chronic
  • a “proliferative disorder of the lung” is a proliferative disorder involving cells of the lung.
  • Proliferative disorders of the lung can include all forms of proliferative disorders affecting lung cells.
  • Proliferative disorders of the lung can include lung cancer, a precancer or precancerous condition of the lung, benign growths or lesions of the lung, and malignant growths or lesions of the lung, and metastatic lesions in tissue and organs in the body other than the lung.
  • the compositions of the present application may be used to treat lung cancer or proliferative disorders of the lung.
  • Lung cancer can include all forms of cancer of the lung.
  • Lung cancer can include malignant lung neoplasms, carcinoma in situ, typical carcinoid tumors, and atypical carcinoid tumors.
  • Lung cancer can include small cell lung cancer (“SCLC”), non-small cell lung cancer (“NSCLC”), squamous cell carcinoma, adenocarcinoma, small cell carcinoma, large cell carcinoma, adenosquamous cell carcinoma, and mesothelioma.
  • Lung cancer can include “scar carcinoma”, bronchioalveolar carcinoma, giant cell carcinoma, spindle cell carcinoma, and large cell neuroendocrine carcinoma.
  • Lung cancer can include lung neoplasms having histologic and ultrastructual heterogeneity (e.g., mixed cell types).
  • Proliferative disorders of the lung can include all forms of proliferative disorders affecting lung cells.
  • Proliferative disorders of the lung can include lung cancer, precancerous conditions of the lung.
  • Proliferative disorders of the lung can include hyperplasia, metaplasia, and dysplasia of the lung.
  • Proliferative disorders of the lung can include asbestos-induced hyperplasia, squamous metaplasia, and benign reactive mesothelial metaplasia.
  • Proliferative disorders of the lung can include replacement of columnar epithelium with stratified squamous epithelium, and mucosal dysplasia.
  • Prior lung diseases that may predispose individuals to development of proliferative disorders of the lung can include chronic interstitial lung disease, necrotizing pulmonary disease, scleroderma, rheumatoid disease, sarcoidosis, interstitial pneumonitis, tuberculosis, repeated pneumonias, idiopathic pulmonary fibrosis, granulomata, asbestosis, fibrosing alveolitis, and Hodgkin's disease.
  • a “proliferative disorder of the colon” is a proliferative disorder involving cells of the colon.
  • the proliferative disorder of the colon is colon cancer.
  • compositions of the present application may be used to treat colon cancer or proliferative disorders of the colon.
  • Colon cancer can include all forms of cancer of the colon.
  • Colon cancer can include sporadic and hereditary colon cancers.
  • Colon cancer can include malignant colon neoplasms, carcinoma in situ, typical carcinoid tumors, and atypical carcinoid tumors.
  • Colon cancer can include adenocarcinoma, squamous cell carcinoma, and adenosquamous cell carcinoma.
  • Colon cancer can be associated with a hereditary syndrome selected from the group consisting of hereditary nonpolyposis colorectal cancer, familial adenomatous polyposis, Gardner's syndrome, Peutz-Jeghers syndrome, Turcot's syndrome and juvenile polyposis.
  • Colon cancer can be caused by a hereditary syndrome selected from the group consisting of hereditary nonpolyposis colorectal cancer, familial adenomatous polyposis, Gardner's syndrome, Koz- Jeghers syndrome, Turcot's syndrome and juvenile polyposis.
  • Proliferative disorders of the colon can include all forms of proliferative disorders affecting colon cells.
  • Proliferative disorders of the colon can include colon cancer, precancerous conditions of the colon, adenomatous polyps of the colon and metachronous lesions of the colon.
  • a proliferative disorder of the colon can include adenoma.
  • Proliferative disorders of the colon can be characterized by hyperplasia, metaplasia, and dysplasia of the colon.
  • Prior colon diseases that may predispose individuals to development of proliferative disorders of the colon can include prior colon cancer.
  • Current disease that may predispose individuals to development of proliferative disorders of the colon can include Crohn's disease and ulcerative colitis.
  • a proliferative disorder of the colon can be associated with a mutation in a gene selected from the group consisting of p53, ras, FAP and DCC.
  • An individual can have an elevated risk of developing a proliferative disorder of the colon due to the presence of a mutation in a gene selected from the group consisting of p53, ras, FAP and DCC.
  • a “proliferative disorder of the pancreas” is a proliferative disorder involving cells of the pancreas. Proliferative disorders of the pancreas can include all forms of proliferative disorders affecting pancreatic cells.
  • Proliferative disorders of the pancreas can include pancreas cancer, a precancer or precancerous condition of the pancreas, hyperplasia of the pancreas, and dysplasia of the pancreas, benign growths or lesions of the pancreas, and malignant growths or lesions of the pancreas, and metastatic lesions in tissue and organs in the body other than the pancreas.
  • Pancreatic cancer includes all forms of cancer of the pancreas.
  • Pancreatic cancer can include ductal adenocarcinoma, adenosquamous carcinoma, pleomorphic giant cell carcinoma, mucinous adenocarcinoma, osteoclast-like giant cell carcinoma, mucinous cystadenocarcinoma, acinar carcinoma, unclassified large cell carcinoma, small cell carcinoma, pancreatoblastoma, papillary neoplasm, mucinous cystadenoma, papillary cystic neoplasm, and serous cystadenoma.
  • Pancreatic cancer can also include pancreatic neoplasms having histologic and ultrastructual heterogeneity (e.g., mixed cell types).
  • a “proliferative disorder of the prostate” is a proliferative disorder involving cells of the prostate.
  • Proliferative disorders of the prostate can include all forms of proliferative disorders affecting prostate cells.
  • Proliferative disorders of the prostate can include prostate cancer, a precancer or precancerous condition of the prostate, benign growths or lesions of the prostate, and malignant growths or lesions of the prostate, and metastatic lesions in tissue and organs in the body other than the prostate.
  • Proliferative disorders of the prostate can include hyperplasia, metaplasia, and dysplasia of the prostate.
  • a “proliferative disorder of the skin” is a proliferative disorder involving cells of the skin.
  • Proliferative disorders of the skin can include all forms of proliferative disorders affecting skin cells.
  • Proliferative disorders of the skin can include a precancer or precancerous condition of the skin, benign growths or lesions of the skin, melanoma, malignant melanoma and other malignant growths or lesions of the skin, and metastatic lesions in tissue and organs in the body other than the skin.
  • Proliferative disorders of the skin can include hyperplasia, metaplasia, and dysplasia of the skin.
  • a “proliferative disorder of the ovary” is a proliferative disorder involving cells of the ovary.
  • Proliferative disorders of the ovary can include all forms of proliferative disorders affecting cells of the ovary.
  • Proliferative disorders of the ovary can include a precancer or precancerous condition of the ovary, benign growths or lesions of the ovary, ovarian cancer, malignant growths or lesions of the ovary, and metastatic lesions in tissue and organs in the body other than the ovary.
  • Proliferative disorders of the skin can include hyperplasia, metaplasia, and dysplasia of cells of the ovary.
  • a “proliferative disorder of the breast” is a proliferative disorder involving cells of the breast.
  • Proliferative disorders of the breast can include all forms of proliferative disorders affecting breast cells.
  • Proliferative disorders of the breast can include breast cancer, a precancer or precancerous condition of the breast, benign growths or lesions of the breast, and malignant growths or lesions of the breast, and metastatic lesions in tissue and organs in the body other than the breast.
  • Proliferative disorders of the breast can include hyperplasia, metaplasia, and dysplasia of the breast.
  • a cancer that is to be treated can be staged according to the American Joint Committee on Cancer (AJCC) TNM classification system, where the tumor (T) has been assigned a stage of TX, T1, T1mic, T1a, T1b, T1c, T2, T3, T4, T4a, T4b, T4c, or T4d; and where the regional lymph nodes (N) have been assigned a stage of NX, N0, N1, N2, N2a, N2b, N3, N3a, N3b, or N3c; and where distant metastasis (M) can be assigned a stage of MX, M0, or M1.
  • AJCC American Joint Committee on Cancer
  • a cancer that is to be treated can be staged according to an American Joint Committee on Cancer (AJCC) classification as Stage I, Stage IIA, Stage IIB, Stage IIIA, Stage IIIB, Stage IIIC, or Stage IV.
  • AJCC American Joint Committee on Cancer
  • a cancer that is to be treated can be assigned a grade according to an AJCC classification as Grade GX (e.g., grade cannot be assessed), Grade 1, Grade 2, Grade 3 or Grade 4.
  • a cancer that is to be treated can be staged according to an AJCC pathologic classification (pN) of pNX, pN0, PN0 (I-), PN0 (I+), PN0 (mol-), PN0 (mol+), PN1, PN1(mi), PN1a, PN1b, PN1c, pN2, pN2a, pN2b, pN3, pN3a, pN3b, or pN3c.
  • pN AJCC pathologic classification
  • a cancer that is to be treated can include a tumor that has been determined to be less than or equal to about 2 centimeters in diameter.
  • a cancer that is to be treated can include a tumor that has been determined to be from about 2 to about 5 centimeters in diameter.
  • a cancer that is to be treated can include a tumor that has been determined to be greater than or equal to about 3 centimeters in diameter.
  • a cancer that is to be treated can include a tumor that has been determined to be greater than 5 centimeters in diameter.
  • a cancer that is to be treated can be classified by microscopic appearance as well differentiated, moderately differentiated, poorly differentiated, or undifferentiated.
  • a cancer that is to be treated can be classified by microscopic appearance with respect to mitosis count (e.g., amount of cell division) or nuclear pleiomorphism (e.g., change in cells).
  • a cancer that is to be treated can be classified by microscopic appearance as being associated with areas of necrosis (e.g., areas of dying or degenerating cells).
  • a cancer that is to be treated can be classified as having an abnormal karyotype, having an abnormal number of chromosomes, or having one or more chromosomes that are abnormal in appearance.
  • a cancer that is to be treated can be classified as being aneuploid, triploid, tetraploid, or as having an altered ploidy.
  • a cancer that is to be treated can be classified as having a chromosomal translocation, or a deletion or duplication of an entire chromosome, or a region of deletion, duplication or amplification of a portion of a chromosome.
  • a cancer that is to be treated can be evaluated by DNA cytometry, flow cytometry, or image cytometry.
  • a cancer that is to be treated can be typed as having 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of cells in the synthesis stage of cell division (e.g., in S phase of cell division).
  • a cancer that is to be treated can be typed as having a low S-phase fraction or a high S-phase fraction.
  • a “normal cell” is a cell that cannot be classified as part of a “proliferative disorder”. A normal cell lacks unregulated or abnormal growth, or both, that can lead to the development of an unwanted condition or disease.
  • a normal cell possesses normally functioning cell cycle checkpoint control mechanisms.
  • One skilled in the art may refer to general reference texts for detailed descriptions of known techniques discussed herein or equivalent techniques. These texts include Ausubel et al., Current Protocols in Molecular Biology, John Wiley and Sons, Inc. (2005); Sambrook et al., Molecular Cloning, A Laboratory Manual (3rd edition), Cold Spring Harbor Press, Cold Spring Harbor, N.Y.
  • compounds of the application are useful in the treatment of proliferative disorders (e.g., cancer, benign neoplasms, inflammatory disease, and autoimmune diseases).
  • proliferative disorders e.g., cancer, benign neoplasms, inflammatory disease, and autoimmune diseases.
  • levels of cell proteins of interest e.g., pathogenic and oncogenic proteins are modulated, or their growth is inhibited by contacting said cells with a compound or composition, as described herein.
  • the compounds are useful in treating cancer.
  • the method involves the administration of a therapeutically effective amount of the compound or a pharmaceutically acceptable derivative thereof to a subject (including, but not limited to a human or animal) in need of it.
  • the compounds are useful for the treatment of cancer (including, but not limited to, glioblastoma, retinoblastoma, breast cancer, cervical cancer, colon and rectal cancer, leukemia, lymphoma, lung cancer (including, but not limited to small cell lung cancer), melanoma and/or skin cancer, multiple myeloma, non-Hodgkin's lymphoma, ovarian cancer, pancreatic cancer, prostate cancer and gastric cancer, bladder cancer, uterine cancer, kidney cancer, testicular cancer, stomach cancer, brain cancer, liver cancer, or esophageal cancer).
  • cancer including, but not limited to, glioblastoma, retinoblastoma, breast cancer, cervical cancer, colon and rectal cancer, leukemia, lymphoma, lung cancer (including, but not limited to small cell lung cancer), melanoma and/or skin cancer, multiple myeloma, non-Hodgkin's lymphoma, ovarian cancer, pancreatic
  • the anticancer agents are useful in the treatment of cancers and other proliferative disorders, including, but not limited to breast cancer, cervical cancer, colon and rectal cancer, leukemia, lung cancer, melanoma, multiple myeloma, non-Hodgkin's lymphoma, ovarian cancer, pancreatic cancer, prostate cancer, and gastric cancer.
  • the anticancer agents are active against solid tumors.
  • the present application provides pharmaceutically acceptable derivatives of the compounds, and methods of treating a subject using these compounds, pharmaceutical compositions thereof, or either of these in combination with one or more additional therapeutic agents.
  • therapies or anticancer agents that may be used in combination with the compounds disclosed herein including surgery, radiotherapy, endocrine therapy, biologic response modifiers (interferons, interleukins, and tumor necrosis factor (TNF), to name a few), hyperthermia and cryotherapy, agents to attenuate any adverse effects (e.g., antiemetics), and other approved chemotherapeutic drugs, including, but not limited to, alkylating drugs (mechlorethamine, chlorambucil, Cyclophosphamide, Melphalan, Ifosfamide), antimetabolites (Methotrexate), purine antagonists and pyrimidine antagonists (6-Mercaptopurine, 5-Fluorouracil, Cytarabile, Gemcitabine), spindle poisons (Vinblastine, Vincristine, Vinorelbine, Paclitaxel), podophyllotoxins (Etoposide, Irinotecan, Topotecan), antibiotics (Doxorubi
  • compositions comprising the compounds disclosed herein further comprise one or more additional therapeutically active ingredients (e.g., chemotherapeutic and/or palliative).
  • additional therapeutically active ingredients e.g., chemotherapeutic and/or palliative.
  • the term “palliative” refers to treatment that is focused on the relief of symptoms of a disease and/or side effects of a therapeutic regimen, but is not curative.
  • palliative treatment encompasses painkillers, antinausea medications and anti-sickness drugs.
  • chemotherapy, radiotherapy and surgery can all be used palliatively (that is, to reduce symptoms without going for cure; e.g., for shrinking tumors and reducing pressure, bleeding, pain and other symptoms of cancer).
  • Administration Pharmaceutical Compositions [0162] Administration of the disclosed compounds and pharmaceutical compositions can be accomplished via any mode of administration for therapeutic agents.
  • compositions can be in solid, semi-solid or liquid dosage form, such as, for example, injectables, tablets, suppositories, pills, time- release capsules, elixirs, tinctures, emulsions, syrups, powders, liquids, suspensions, or the like, sometimes in unit dosages and consistent with conventional pharmaceutical practices.
  • injectables tablets, suppositories, pills, time- release capsules, elixirs, tinctures, emulsions, syrups, powders, liquids, suspensions, or the like, sometimes in unit dosages and consistent with conventional pharmaceutical practices.
  • intravenous both bolus and infusion
  • intraperitoneal subcutaneous or intramuscular form
  • Illustrative pharmaceutical compositions are tablets and gelatin capsules comprising a compound of the disclosure and a pharmaceutically acceptable carrier, such as a) a diluent, e.g., purified water, triglyceride oils, such as hydrogenated or partially hydrogenated vegetable oil, or mixtures thereof, com oil, olive oil, sunflower oil, safflower oil, fish oils, such as EPA or DHA, or their esters or triglycerides or mixtures thereof, omega-3 fatty acids or derivatives thereof, lactose, dextrose, sucrose, mannitol, sorbitol, cellulose, sodium, saccharin, glucose and/or glycine; b) a lubricant, e.g., silica, talcum, stearic acid, its magnesium or calcium salt, sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and/or polyethylene glycol; for example,
  • Liquid, particularly injectable, compositions can, for example, be prepared by dissolution, dispersion, etc.
  • the disclosed compound is dissolved in or mixed with a pharmaceutically acceptable solvent such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form an injectable isotonic solution or suspension.
  • a pharmaceutically acceptable solvent such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like
  • Proteins such as albumin, chylomicron particles, or serum proteins can be used to solubilize the disclosed compounds.
  • the disclosed compounds can be also formulated as a suppository that can be prepared from fatty emulsions or suspensions; using polyalkylene glycols such as propylene glycol, as the carrier.
  • the disclosed compounds can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles.
  • Liposomes can be formed from a variety of phospholipids, containing cholesterol, stearylamine or phosphatidylcholines.
  • a film of lipid components is hydrated with an aqueous solution of drug to a form lipid layer encapsulating the drug, as described in U.S. Pat. No.5,262,564, which is hereby incorporated by reference in its entirety.
  • Disclosed compounds can also be delivered by the use of monoclonal antibodies as individual carriers to which the disclosed compounds are coupled.
  • the disclosed compounds can also be coupled with soluble polymers as targetable drug carriers.
  • Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspanamidephenol, or polyethyleneoxidepolylysine substituted with palmitoyl residues.
  • the disclosed compounds can be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and cross-linked or amphipathic block copolymers of hydrogels.
  • a drug for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and cross-linked or amphipathic block copolymers of hydrogels.
  • disclosed compounds are not covalently bound to a polymer, e.g., a polycarboxylic acid polymer, or a polyacrylate.
  • Parental injectable administration is generally used for subcutaneous, intramuscular or intravenous injections and infusions.
  • Injectables can be prepared in conventional forms, either as liquid solutions or suspensions or solid forms suitable for dissolving in liquid prior to injection.
  • Another aspect of the disclosure is directed to pharmaceutical compositions comprising a compound of Formula I, I-1, IA, or IA-1 and a pharmaceutically acceptable carrier.
  • Another aspect of the disclosure is directed to pharmaceutical compositions comprising a compound of Formula I, I-1, IA, or IA-1, or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof and a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier may further include an excipient, diluent, or surfactant.
  • compositions can be prepared according to conventional mixing, granulating or coating methods, respectively, and the present pharmaceutical compositions can contain from about 0.1% to about 99%, from about 5% to about 90%, or from about 1% to about 20% of the disclosed compound by weight or volume.
  • the disclosure provides a kit comprising two or more separate pharmaceutical compositions, at least one of which contains a compound of the present disclosure.
  • the kit comprises means for separately retaining said compositions, such as a container, divided bottle, or divided foil packet.
  • An example of such a kit is a blister pack, as typically used for the packaging of tablets, capsules and the like.
  • the kit of the disclosure may be used for administering different dosage forms, for example, oral and parenteral, for administering the separate compositions at different dosage intervals, or for titrating the separate compositions against one another.
  • the kit of the disclosure typically comprises directions for administration.
  • Pharmaceutical dosage forms of a compound of this disclosure may be manufactured by any of the methods well-known in the art, such as, for example, by conventional mixing, sieving, dissolving, melting, granulating, dragee-making, tableting, suspending, extruding, spray-drying, levigating, emulsifying, (nano-/micro-) encapsulating, entrapping, or lyophilization processes.
  • compositions of this disclosure can include one or more physiologically acceptable inactive ingredients that facilitate processing of active molecules into preparations for pharmaceutical use.
  • the compositions are comprised of, in general, a compound of this disclosure in combination with at least one pharmaceutically acceptable excipient.
  • Acceptable excipients are non- toxic, aid administration, and do not adversely affect the therapeutic benefit of the claimed compounds.
  • excipient may be any solid, liquid, semi-solid or, in the case of an aerosol composition, gaseous excipient that is generally available to one of skill in the art.
  • Solid pharmaceutical excipients include starch, cellulose, talc, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, magnesium stearate, sodium stearate, glycerol monostearate, sodium chloride, dried skim milk and the like.
  • Liquid and semi-solid excipients may be selected from glycerol, propylene glycol, water, ethanol and various oils, including those of petroleum, animal, vegetable or synthetic origin, e.g., peanut oil, soybean oil, mineral oil, sesame oil, etc.
  • liquid carriers, particularly for injectable solutions include water, saline, aqueous dextrose, and glycols.
  • compositions of this disclosure may, if desired, be presented in a pack or dispenser device containing one or more unit dosage forms containing the active ingredient.
  • a pack or device may, for example, comprise metal or plastic foil, such as a blister pack, or glass, and rubber stoppers such as in vials.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • compositions comprising a compound of this disclosure that can be formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • the amount of the compound in a formulation can vary within the full range employed by those skilled in the art. Typically, the formulation will contain, on a weight percent (wt %) basis, from about 0.01-99.99 wt % of a compound of this disclosure based on the total formulation, with the balance being one or more suitable pharmaceutical excipients. In one embodiment, the compound is present at a level of about 1-80 wt %. Representative pharmaceutical formulations are described below. Formulation Examples [0181] The following are representative pharmaceutical formulations containing a compound of this disclosure.
  • Formulation Example 1 Tablet formulation [0182] The following ingredients are mixed intimately and pressed into single scored tablets.
  • Quantity per Formulation Example 2 Capsule formulation [0183] The following ingredients are mixed intimately and loaded into a hard-shell gelatin capsule
  • Quantity per Formulation Example 3 Suspension formulation [0184] The following ingredients are mixed to form a suspension for oral administration.
  • Formulation Example 4 Injectable formulation [0185] The following ingredients are mixed to form an injectable formulation.
  • a suppository of total weight 2.5 g is prepared by mixing the compound of this disclosure with Witepsol® H-15 (triglycerides of saturated vegetable fatty acid; Riches-Nelson, Inc., New York), and has the following composition: Ingredient Amount Dosing [0187] The dosage regimen utilizing the disclosed compound is selected in accordance with a variety of factors including type, species, age, weight, sex, and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal or hepatic function of the patient; and the particular disclosed compound employed.
  • Effective dosage amounts of the disclosed compounds when used for the indicated effects, range from about 0.5 mg to about 5000 mg of the disclosed compound as needed to treat the condition.
  • Compositions for in vivo or in vitro use can contain about 0.5, 5, 20, 50, 75, 100, 150, 250, 500, 750, 1000, 1250, 2500, 3500, or 5000 mg of the disclosed compound, or, in a range of from one amount to another amount in the list of doses.
  • the compositions are in the form of a tablet that can be scored.
  • Acetyl chloride (1.2 g, 14.9 mmol, 1.1 mL, 1.1 eq) was added dropwise at 0 °C. The resulting mixture was stirred at 25 °C for 20 hr. The solvents were evaporated in vacuo, and the residue diluted with water (20 mL), then extracted with ethyl acetate (40 mL). The organic phase was separated, washed with brine (20 mL), dried over Na 2 SO 4 , filtered and the solvents were evaporated in vacuo to give 1-acetyl-2,3-dihydroquinolin-4(1H)-one, which was used in the next step without further purification.
  • reaction mixture was quenched by slow addition 0.5 N hydrochloric acid (20 mL) and diluted with H 2 O (50 mL), then extracted with ethyl acetate (50 mL). The combined organic layers were washed with brine (10 mL), dried over Na 2 SO 4 , filtered and the solvents were evaporated in vacuo. The residue was purified by column chromatography (SiO 2 , 0 to 50% ethyl acetate in petroleum ether) to give (R)-N-((R)-1-acetyl-1,2,3,4-tetrahydroquinolin- 4-yl)-2-methylpropane-2-sulfinamide.
  • Step 3 [0196] To a solution of (R)-N-((R)-1-acetyl-1,2,3,4-tetrahydroquinolin-4-yl)-2-methylpropane-2- sulfinamide (1.2 g, 4.3 mmol, 1.0 eq) in dioxane (13 mL) was added HCl/dioxane (6.4 mL) dropwise at 0 °C. The mixture was stirred at 25 °C for 3 hr. The reaction mixture was quenched by addition of triethylamine (20 mL) in portions, and then diluted with H2O (50 mL) and extracted with ethyl acetate (50 mL).
  • Step 4 [0197] To a solution of (R)-1-(4-amino-3,4-dihydroquinolin-1(2H)-yl)ethan-1-one (50.0 mg, 262.8 ⁇ mol, 1.0 eq) and [4-[3-(1,3-dioxoisoindolin-2-yl)prop-1-ynyl]phenyl]boronic acid (96.2 mg, 315.3 ⁇ mol, 1.2 eq) in MeCN (4 mL), was added pyridine (62.3 mg, 788.4 ⁇ mol, 63.6 ⁇ L, 3.0 eq) and Cu(OAc) 2 (95.4 mg, 525.6 ⁇ mol, 2.0 eq).
  • Step 5 [0198] To a solution of (R)-2-(3-(4-((1-acetyl-1,2,3,4-tetrahydroquinolin-4-yl)amino)phenyl)prop-2-yn- 1-yl)isoindoline-1,3-dione (50.0 mg, 111.2 ⁇ mol, 1.0 eq) in EtOH (0.3 mL) was added hydrazine hydrate (139.2 mg, 2.7 mmol, 135.1 ⁇ L, 25.0 eq). The mixture was stirred at 25 °C for 2 hr. The reaction mixture was partitioned between H 2 O (3 mL) and DCM (3 mL).
  • Step 4 [0202] To a mixture of (S)-3-((4-bromophenyl)amino)butanamide (12 g, 46.67 mmol) in ethyl acetate (100 mL) was added isopropyl carbonochloridate (7.44 g, 60.67 mmol, 8.42 mL) at -10 °C. Lithium tert- butoxide (8.97 g, 112.01 mmol, 10.10 mL) in tetrahydrofuran (150 mL) was added dropwise at -10 °C to the mixture. The reaction was stirred 0 °C for 0.5 h.
  • Step 5 To a mixture of isopropyl (S)-(3-((4-bromophenyl)amino)butanoyl)carbamate (12 g, 34.96 mmol) in ethanol (120 mL) was added NaBH 4 (992.06 mg, 26.22 mmol) at -20 °C. MgCl 2 (3.66 g, 38.46 mmol) in water (12 mL) was added dropwise at -20 °C. The mixture was stirred at 0 °C for 1 h.
  • Step 6 [0204] To a solution of isopropyl ((2S,4R)-6-bromo-2-methyl-1,2,3,4-tetrahydroquinolin-4-yl)carbamate (8.6 g, 26.28 mmol) and pyridine (8.32 g, 105.13 mmol, 8.49 mL) in dichloromethane (90 mL), was added acetyl chloride (6.19 g, 78.85 mmol, 5.63 mL) dropwise under nitrogen at 20 °C. The mixture was stirred for 1 h. The mixture was poured into saturated NaHCO3 solution (200 mL) and extracted with dichloromethane (3x100 mL).
  • Step 7 A solution of aluminum trichloride (10.11 g, 75.83 mmol) in dichloromethane (60 mL) was added dropwise to isopropyl ((2S,4R)-1-acetyl-6-bromo-2-methyl-1,2,3,4-tetrahydroquinolin-4-yl)carbamate (7 g, 18.96 mmol) in dichloromethane (20 mL) at 0 °C under nitrogen. The mixture was stirred at 0 °C for 0.5 h. Triethylamine (23.02 g, 227.49 mmol, 31.66 mL) in methanol (8 mL) was added at 0 °C.
  • Step 8 To a solution of 1-((2S,4R)-4-amino-6-bromo-2-methyl-3,4-dihydroquinolin-1(2H)-yl)ethan-1- one (1 g, 3.5 mmol, 1.0 eq) in ACN (10 mL), was added (4-(3-((tert-butoxycarbonyl)amino)prop-1-yn-1- yl)phenyl)boronic acid (1.9 g, 7.1 mmol, 2.0 eq), Cu(OAc)2 (641.4 mg, 3.5 mmol, 1.0 eq), and pyridine (558.7 mg, 7.1 mmol, 570.1 ⁇ L, 2.0 eq).
  • Step 9 [0207] To a solution of tert-butyl (3-(4-(((2S,4R)-1-acetyl-6-bromo-2-methyl-1,2,3,4-tetrahydroquinolin- 4-yl)amino)phenyl)prop-2-yn-1-yl)carbamate (200 mg, 390.3 ⁇ mol, 1.0 eq) in DCM (3 mL), was added 2,6-dimethylpyridine (250.9 mg, 2.3 mmol, 272.7 ⁇ L, 6.0 eq) at 25 °C.
  • Step 2 [0209] To a solution of tert-butyl (3-(4-(((2R,4S)-1-acetyl-2,6-dimethyl-1,2,3,4-tetrahydroquinolin-4- yl)amino)phenyl)prop-2-yn-1-yl)carbamate (20 mg, 44.69 ⁇ mol, 1 eq) in CH2Cl2 (0.5 mL), was added TMSOTf (59.59 mg, 268.12 ⁇ mol, 48.45 ⁇ L, 6 eq) and 2,6-dimethylpyridine (19.15 mg, 178.74 ⁇ mol, 20.82 ⁇ L, 4 eq).
  • reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (2x10 mL). The combined organic layers were washed with brine (3x10 mL), dried over Na2SO4, filtered, and the solvents evaporated in vacuo.
  • Step 3 To a mixture of AlCl3 (173 mg, 1.30 mmol, 63.80 ⁇ L, 4 eq) in DCM (1 mL) at 0 °C was added a solution of isopropyl ((2S,4R)-1-acetyl-6-fluoro-2-methyl-1,2,3,4-tetrahydroquinolin-4-yl)carbamate (100 mg, 324.31 ⁇ mol, 1 eq) in DCM (1 mL) at 0 °C. The mixture was stirred at 0 °C for 30 minutes. Triethylamine (2 mL) was slowly added to the reaction at 0 °C.
  • Step 4 [0213] A solution of 1-((2S,4R)-4-amino-6-fluoro-2-methyl-3,4-dihydroquinolin-1(2H)-yl)ethan-1-one (70 mg, 314.95 ⁇ mol, 1 eq), [4-[3-(tert-butoxycarbonylamino)prop-1-ynyl]phenyl]boronic acid (173.29 mg, 629.90 ⁇ mol, 2 eq), Cu(OAc) 2 (57.20 mg, 314.95 ⁇ mol, 1 eq), and pyridine (49.82 mg, 629.90 ⁇ mol, 50.84 ⁇ L, 2 eq) in ACN (1 mL) was stirred at 20 °C for 12 h.
  • Step 5 [0214] To a solution of tert-butyl (3-(4-(((2S,4R)-1-acetyl-6-fluoro-2-methyl-1,2,3,4-tetrahydroquinolin- 4-yl)amino)phenyl)prop-2-yn-1-yl)carbamate (30 mg, 66.44 ⁇ mol, 1 eq) and lutidine (28.48 mg, 265.76 ⁇ mol, 30.95 ⁇ L, 4 eq) in DCM (1 mL), was added trimethylsilyl trifluoromethanesulfonate (44.30 mg, 199.32 ⁇ mol, 36.02 ⁇ L, 3 eq) dropwise at 20 °C, and the mixture was stirred for 2 h.
  • Step 2 To a solution of 1-((2S,4R)-6-bromo-4-((4-chlorophenyl)amino)-2-methyl-3,4-dihydroquinolin- 1(2H)-yl)ethan-1-one (50.0 mg, 127.0 ⁇ mol, 1.0 eq) in THF (0.8 mL) and H 2 O (0.2 mL), was added 1- methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (39.6 mg, 190.5 ⁇ mol, 1.5 eq), K 3 PO 4 (80.8 mg, 381.0 ⁇ mol, 3.0 eq), and XPhos-Pd-G 2 (10.0 mg, 12.7 ⁇ mol, 0.1 eq).
  • Step 3 [02 7] .5 m otage m crowave reactor was c arge wt -[( S, )- -[( -c orop eny)am no]- - methyl-6-(1-methyl-1H-pyrazol-5-yl)-1,2,3,4-tetrahydroquinolin-1-yl]ethan-1-one (52.0 mg, 132 ⁇ mol), prop-2-yn-1-amine (25.3 ⁇ L, 3 eq., 395 ⁇ mol), cesium carbonate (257 mg, 6 eq., 790 ⁇ mol), degassed ACN (3 mL), and Pd(Cy*Phine) 2 Cl 2 (14.0 mg, 132 ⁇ mol).
  • the microwave tube was then sealed under an argon atmosphere and heated at 90 °C under microwave irradiation for 2 hours.
  • the solvents were evaporated in vacuo and the resulting residue was dissolved in a mixture of DCM and DMA (2 mL).
  • the insoluble solid was filtered and the solvents were evaporated in vacuo.
  • the resulting residue was purified by prep-HPLC to give 1-((2S,4R)-4-((4-(3-aminoprop-1-yn-1-yl)phenyl)amino)-2-methyl-6-(1-methyl- 1H-pyrazol-5-yl)-3,4-dihydroquinolin-1(2H)-yl)ethan-1-one.
  • Step 2 [0220] To a solution of 1-((2S)-4-(1-(4-bromophenyl)ethyl)-2-methyl-3,4-dihydroquinoxalin-1(2H)- yl)ethan-1-one (50 mg, 139 ⁇ mol, 1 eq) and prop-2-yn-1-amine (15.33 mg, 278 ⁇ mol, 17.83 ⁇ L, 2 eq) in DMSO (2 mL) was added CuI (2.65 mg, 13.92 ⁇ mol, 0.1 eq), Pd(PPh3)2Cl2 (9.77 mg, 13.92 ⁇ mol, 0.1 eq) and Cs2CO3 (136.04 mg, 417.53 ⁇ mol, 3 eq).
  • Step 3 A mixture of (S)-2-((2-amino-4-bromophenyl)amino)propan-1-ol (93 g, 379.41 mmol) in pyridine (1 L) was added 4-methylbenzene-1-sulfonyl chloride (72.33 g, 379.41 mmol) dropwise at 0 °C. The mixture was stirred at 0°C for 2 h. The mixture was poured into water (2 L) and extracted with ethyl acetate (3 ⁇ 1 L). The organic phase was washed with brine (1.5 L), dried over Na2SO4 and concentrated to give a residue.
  • 4-methylbenzene-1-sulfonyl chloride 72.33 g, 379.41 mmol
  • Step 4 To a mixture of (S)-N-(5-bromo-2-((1-hydroxypropan-2-yl)amino)phenyl)-4- methylbenzenesulfonamide (140 g, 350.61 mmol) and PPh 3 (110.35 g, 420.73 mmol) in THF (2.5 L) was added DIAD (77.99 g, 385.67 mmol, 74.99 mL) dropwise at 0°C. The mixture was stirred at 0°C for 1.5 h. The mixture was concentrated under pressure to give a residue.
  • Step 5 To a mixture of (S)-7-bromo-3-methyl-1-tosyl-1,2,3,4-tetrahydroquinoxaline (133 g, 348.82 mmol) and pyridine (41.39 g, 523.23 mmol, 42.23 mL) in DCM (3 L) at 0°C was added acetyl chloride (32.86 g, 418.58 mmol, 29.87 mL) dropwise. The mixture was stirred at 0°C for 1.5 h.
  • Step 7 [0229] To a solution o (S)- -(6- romo- -met y-3, - y roqu noxa n- ( )-y )ethan-1-one (2 g, 7.43 mmol, 1 eq) and DMAP (90.79 mg, 743.12 ⁇ mol, 0.1 eq) in DCE (30 mL) was added Boc 2 O (2.43 g, 11.15 mmol, 2.56 mL, 1.5 eq) dropwise. The mixture was stirred at 50°C for 1 h. The reaction mixture was concentrated under reduced pressure to give a residue.
  • Step 8 [0230] A mixture of tert-butyl (S)-4-acetyl-7-bromo-3-methyl-3,4-dihydroquinoxaline-1(2H)- carboxylate (1.5 g, 4.06 mmol, 1 eq), BPD (3.09 g, 12.19 mmol, 3 eq), Pd(dppf)Cl 2 (297.24 mg, 406.23 ⁇ mol, 0.1 eq) and KOAc (1.20 g, 12.19 mmol, 3 eq) in DMSO (15 mL) was heated at 80°C for 12 h under a N 2 atmosphere.
  • reaction mixture was diluted with water (100 mL) and extracted with ethyl acetate (2 ⁇ 50 mL). The combined organic layers were washed with brine (4 ⁇ 100 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue.
  • the residue was purified by column chromatography (SiO 2 , 20 to 35% ethyl acetate in petroleum ether) to give tert-butyl (S)-4-acetyl-3- methyl-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,4-dihydroquinoxaline-1(2H)-carboxylate.
  • Step 9 [0231] To a mixture of KOtBu (424.51 mg, 3.78 mmol, 1.05 eq) in MeOH (15 mL) was added tert-butyl (S)-4-acetyl-3-methyl-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,4-dihydroquinoxaline-1(2H)- carboxylate (1.5 g, 3.60 mmol, 1 eq) at 20 ° C. After stirring for 15 min, the reaction mixture was cooled to 0 ° C and AgOTf (2.78 g, 10.81 mmol, 3 eq) was added.
  • Step 10 To a solution of tert-butyl (S)-4-acetyl-7-fluoro-3-methyl-3,4-dihydroquinoxaline-1(2H)- carboxylate (0.46 g, 1.49 mmol, 1 eq) in DCM (10 mL) was added TFA (1.70 g, 14.92 mmol, 1.10 mL, 10 eq) dropwise. The mixture was stirred for 12 h. The reaction mixture was concentrated under reduced pressure to give (S)-1-(6-fluoro-2-methyl-3,4-dihydroquinoxalin-1(2H)-yl)ethan-1-one as a TFA salt, which was used into the next step without purification.
  • Step 11 I [0233] A so lutio of (S) 1 (6 fluo o 2 ethyl 3,4 dihyd oqui o ali 1(2H) yl)etha 1 o e (400 mg, 1.24 mmol, 1 eq, TFA salt), KI (20.61 mg, 124.13 ⁇ mol, 0.1 eq), DIEA (802.12 mg, 6.21 mmol, 1.08 mL, 5 eq) and 1-(bromomethyl)-4-iodo-benzene (1.11 g, 3.72 mmol, 3 eq) in DMF (4 mL) was stirred at 50°C for 1 h.
  • Step 12 [0234] To a solution of (S)-1-(6-fluoro-4-(4-iodobenzyl)-2-methyl-3,4-dihydroquinoxalin-1(2H)- yl)ethan-1-one (20 mg, 47.14 ⁇ mol, 1 eq) and prop-2-yn-1-amine (7.79 mg, 141.43 ⁇ mol, 9.06 ⁇ L, 3 eq) in ACN (1 mL) was added Pd(PPh3)2Cl2 (3.31 mg, 4.71 ⁇ mol, 0.1 eq), Cs2CO3 (46.08 mg, 141.43 ⁇ mol, 3 eq) and CuI (897.82 ⁇ g, 4.71 ⁇ mol, 0.1 eq) under a N2 atmosphere.
  • Table 3A Compound Biological Examples Target engagement assay
  • the ability of a compound to bind to BRD4 is assessed using HEK-293T cells and the NanoBRET Target Engagement Intracellular BET BRD Assay from Promega (Cat. No. N2131). Assays were run according to manufacturer’s specifications and luminescent readout was recorded using a CLARIOstar Plus instrument (BMG Labtech). Resulting BRET ratios were plotted and IC50s determined by non-linear regression analyses (GraphPad Prism).
  • BRD4 degradation assay [0237] BRD4 degradation was monitored by immunofluorescence in HEK-293T cells.
  • 96-well plates black, clear-bottom
  • test compounds were added in a 10-point dilution series (typically 30 ⁇ M to 100 pM) using a TECAN D300e Digital Dispenser, and plates were subsequently incubated for 24 hours at 37 °C.
  • Media was carefully removed, and cells were fixed in PBS + 2.5% formalin (50 ⁇ L) for 20 minutes at 37 °C. Following formalin fixation, cells were washed once with PBS and methanol was added (30 ⁇ L). Plates were wrapped in parafilm and incubated at -20 °C for 1 hour to overnight.
  • DAPI (1 ⁇ M final) plus secondary antibody (Southern Biotech 4030-30 anti-Rabbit IgG Alexa Fluor 488; diluted 1:2000 in PBS + 1X fish gelatin, 0.1% Triton X-100) were added at 40 ⁇ L/well, and plates were incubated at room temperature for 2 hours, covered with foil. Cells were washed (3x) with PBS + 0.1% tween-20, followed by one wash with PBS, and the addition of 100 ⁇ L of PBS for imaging. Images were acquired using the ImageXpress Pico system (Molecular Devices). Cell Reporter Xpress software was utilized to segment cells and determine fluorescence intensities, which were used to construct dose- response curves and calculation of degradation DC 50 s (GraphPad Prism). Results for certain compounds are reported in Table 4 and 4A below. Table 4 Table 4A

Abstract

Disclosed are compounds of formula I, formula I-1, or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof that are useful for modulating or degrading bromodomain (e.g., BRD4).

Description

PROPARGYL COMPOUNDS AND PHARMACEUTICAL COMPOSITIONS THAT MODULATE BRD4 CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims the benefit under 35 U.S.C. § 119(e) of United States Provisional Application Serial Number 63/329,308 filed April 8, 2022, and United States Provisional Application Serial Number 63/329,310, filed April 8, 2022, the contents of which are hereby incorporated by reference in their entirety. Field [0002] This disclosure provides for compounds and salts thereof that are useful for modulating or degrading bromodomain (e.g., BRD4). The compounds disclosed herein bind to and, in some cases, degrade BET/BRD4. Also disclosed are pharmaceutical compositions comprising the compounds, or a salt (e.g., a pharmaceutically acceptable salt) thereof, and methods of using such compounds and their salts in the treatment of various bromodomain-mediated diseases or disorders. State of the Art [0003] Chromatin is a complex combination of DNA and protein that makes up chromosomes. It is found inside the nuclei of eukaryotic cells and is divided between heterochromatin (condensed) and euchromatin (extended) forms. The major components of chromatin are DNA and proteins. Histones are the chief protein components of chromatin, acting as spools around which DNA winds. The functions of chromatin are to package DNA into a smaller volume to fit in the cell, to strengthen the DNA to allow mitosis and meiosis, and to serve as a mechanism to control expression and DNA replication. The chromatin structure is controlled by a series of post-translational modifications to histone proteins, notably histones H3 and H4, and most commonly within the “histone tails” which extend beyond the core nucleosome structure. Histone tails tend to be free for protein-protein interaction and are also the portion of the histone most prone to post-translational modification. These modifications include acetylation, methylation, phosphorylation, ubiquitinylation, and SUMOylation. These epigenetic marks are written and erased by specific enzymes that place the tags on specific residues within the histone tail, thereby forming an epigenetic code, which is then interpreted by the cell to allow gene specific regulation of chromatin structure and thereby transcription. [0004] Of all classes of proteins, histones are amongst the most susceptible to post-translational modification. Histone modifications are dynamic, as they can be added or removed in response to specific stimuli, and these modifications direct both structural changes to chromatin and alterations in gene transcription. Distinct classes of enzymes, namely histone acetyltransferases (HATs) and histone deacetylases (HDACs), acetylate or de-acetylate specific histone lysine residues (Struhl K., Genes Dev., 1989, 12, 5, 599-606). [0005] Bromodomains, which are approximately 110 amino acids long, are found in a large number of chromatin-associated proteins and have been identified in approximately 70 human proteins, often adjacent to other protein motifs (Jeanmougin F., et al, Trends Biochem. Sci., 1997, 22, 5,151-153; and Tamkun J. W., et al., Cell, 1992, 7, 3, 561-572). Interactions between bromodomains and modified histones may be an important mechanism underlying chromatin structural changes and gene regulation. Bromodomain-containing proteins have been implicated in disease processes including cancer, inflammation and viral replication. See, e.g., Prinjha et al, Trends Pharm. Sci., 33(3):146-153 (2012) and Muller et al, Expert Rev., 13(29):1-20 (2011). [0006] Cell-type specificity and proper tissue functionality requires the tight control of distinct transcriptional programs that are intimately influenced by their environment. Alterations to this transcriptional homeostasis are directly associated with numerous disease states, most notably cancer, immuno-inflammation, neurological disorders, and metabolic diseases. Bromodomains reside within key chromatin modifying complexes that serve to control distinctive disease-associated transcriptional pathways. This is highlighted by the observation that mutations in bromodomain-containing proteins are linked to cancer, as well as immune and neurologic dysfunction. Moreover, recent findings have demonstrated that small molecule inhibition of the bromodomains of BRD4 may have clinical utility in diverse human diseases, ranging from auto-immunity to cardiac hypertrophy. This is possible because the underlying mechanism resides in transcriptional regulation. Hence, the inhibition of bromodomains across the family creates varied opportunities as novel therapeutic agents in human dysfunction. [0007] Accordingly, the ability to modulate or degrade BRD4 would be a significant advancement in treating cancer and other bromodomain related diseases. Summary [0008] Disclosed are compounds as well as pharmaceutical compositions comprising said compounds and methods of using said compound that modulate the activity of or which degrade BRD4. Such compounds are useful in treating diseases mediated, at least in part, by dysfunction of BRD4 including cancers. [0009] In one embodiment, the disclosed compounds that bind to or degrade BRD4 are represented by formula I:
Figure imgf000004_0001
or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein Q, X1, X2, Y, R1, R2, R3, R4, m, and n are as defined herein. [0010] In one embodiment, the disclosed compounds that bind to or degrade BRD4 are represented by formula I-1:
Figure imgf000004_0002
or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein Q, X1, X2, R1, R2, R3, R4, m, and n are as defined herein. [0011] In one embodiment, the compounds as described herein degrade BRD4 by 30% or more at 1 µM concentration. [0012] In one embodiment, this disclosure provides a pharmaceutical composition comprising a pharmaceutically acceptable excipient and an effective amount of a compound of formula I, formula I-1, or any subformula thereof. [0013] In one embodiment, this disclosure provides a method for modulating or degrading BRD4, which method comprises contacting BRD4 with an effective amount of a compound of formula I, formula I-1, or any subformula thereof under conditions wherein BRD4 is bound to said compound and modulated or degraded. [0014] In one embodiment, this disclosure provides a method for modulating or degrading BRD4 in a subject, which method comprises administering to said subject an effective amount of a compound of formula I, formula I-1, or any subformula thereof or a pharmaceutical composition comprising a pharmaceutically acceptable excipient and an effective amount of a compound of formula I, formula I-1, or any subformula thereof under conditions wherein BRD4 is bound to said compound and modulated or degraded. [0015] In one embodiment, this disclosure provides a method for treating cancer in a subject in need thereof which method comprises administering to said subject an effective amount of a compound of formula I, formula I-1, or any subformula thereof, or a pharmaceutical composition comprising a pharmaceutically acceptable excipient and an effective amount of a compound of formula I, formula I-1, or any subformula thereof. Detailed Description [0016] This disclosure provides for compounds, pharmaceutical compositions comprising such compounds, and methods of using such compounds and compositions to treat diseases, disorders, or conditions mediated, at least in part, by BRD4 transcription factors. However, prior to providing a detailed description of the disclosure, the following terms will first be defined. If not defined, terms used herein have their generally accepted scientific meaning. [0017] The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the disclosure. As used herein, the singular forms “a”, “an” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. [0018] A dash (“-”) that is not between two letters or symbols is used to indicate a point of attachment for a substituent. For example, -C(O)NH2 is attached through the carbon atom. A dash at the front or end of a chemical group is a matter of convenience; chemical groups may be depicted with or without one or more dashes without losing their ordinary meaning. A wavy line or a dashed line drawn through a line in a structure indicates a specified point of attachment of a group. Unless chemically or structurally required, no directionality or stereochemistry is indicated or implied by the order in which a chemical group is written or named. [0019] The prefix “Cu-v” indicates that the following group has from u to v carbon atoms. For example, “C1-6 alkyl” indicates that the alkyl group has from 1 to 6 carbon atoms. [0020] The term “about” when used before a numerical designation, e.g., temperature, time, amount, concentration, and such other, including a range, indicates approximations which may vary by ( + ) or ( - ) 10%, 5%, 1%, or any subrange or subvalue there between. In one embodiment, the term “about” when used with regard to a dose amount means that the dose may vary by +/- 10%. [0021] “Comprising” or “comprises” is intended to mean that the compositions and methods include the recited elements, but not excluding others. [0022] “Consisting essentially of” when used to define compositions and methods, shall mean excluding other elements of any essential significance to the combination for the stated purpose. Thus, a composition consisting essentially of the elements as defined herein would not exclude other materials or steps that do not materially affect the basic and novel characteristic(s) of the claimed disclosure. [0023] “Consisting of” shall mean excluding more than trace elements of other ingredients and substantial method steps. Embodiments defined by each of these transition terms are within the scope of this disclosure. [0024] “Alkyl” refers to an unbranched or branched saturated hydrocarbon chain. As used herein, alkyl has 1 to 20 carbon atoms (i.e., C1-20 alkyl), 1 to 12 carbon atoms (i.e., C1-12 alkyl), 1 to 8 carbon atoms (i.e., C1-8 alkyl), 1 to 6 carbon atoms (i.e., C1-6 alkyl), or 1 to 4 carbon atoms (i.e., C1-4 alkyl). Examples of alkyl groups include, e.g., methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, pentyl, 2-pentyl, isopentyl, neopentyl, hexyl, 2-hexyl, 3-hexyl, and 3-methylpentyl. When an alkyl residue having a specific number of carbons is named by chemical name or identified by molecular formula, all positional isomers having that number of carbons may be encompassed; thus, for example, “butyl” includes n-butyl (i.e., -(CH2)3CH3), sec-butyl (i.e., -CH(CH3)CH2CH3), isobutyl (i.e., -CH2CH(CH3)2), and tert-butyl (i.e., -C(CH3)3); and “propyl” includes n-propyl (i.e., -(CH2)2CH3) and isopropyl (i.e., -CH(CH3)2). [0025] Certain commonly used alternative chemical names may be used. For example, a divalent group such as a divalent “alkyl” group, a divalent “aryl” group, a divalent heteroaryl group, etc., may also be referred to as an “alkylene” group or an “alkylenyl” group (for example, methylenyl, ethylenyl, and propylenyl), an “arylene” group or an “arylenyl” group (for example, phenylenyl or napthylenyl, or quinolinyl for heteroarylene), respectively. Also, unless indicated explicitly otherwise, where combinations of groups are referred to herein as one moiety, e.g., arylalkyl or aralkyl, the last mentioned group contains the atom by which the moiety is attached to the rest of the molecule. [0026] “Alkenyl” refers to an alkyl group containing at least one (e.g., 1-3, or 1) carbon-carbon double bond and having from 2 to 20 carbon atoms (i.e., C2-20 alkenyl), 2 to 12 carbon atoms (i.e., C2-12 alkenyl), 2 to 8 carbon atoms (i.e., C2-8 alkenyl), 2 to 6 carbon atoms (i.e., C2-6 alkenyl), or 2 to 4 carbon atoms (i.e., C2-4 alkenyl). Examples of alkenyl groups include, e.g., ethenyl, propenyl, butadienyl (including 1,2-butadienyl and 1,3-butadienyl). [0027] “Alkynyl” refers to an alkyl group containing at least one (e.g., 1-3, or 1) carbon-carbon triple bond and having from 2 to 20 carbon atoms (i.e., C2-20 alkynyl), 2 to 12 carbon atoms (i.e., C2-12 alkynyl), 2 to 8 carbon atoms (i.e., C2-8 alkynyl), 2 to 6 carbon atoms (i.e., C2-6 alkynyl), or 2 to 4 carbon atoms (i.e., C2-4 alkynyl). The term “alkynyl” also includes those groups having one triple bond and one double bond. [0028] “Alkoxy” refers to the group “alkyl-O-”. Examples of alkoxy groups include, e.g., methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, and 1,2-dimethylbutoxy. [0029] “Alkylthio” refers to the group “alkyl-S-”. “Alkylsulfinyl” refers to the group “alkyl-S(O)-”. “Alkylsulfonyl” refers to the group “alkyl-S(O)2-”. “Alkylsulfonylalkyl” refers to -alkyl-S(O)2-alkyl. [0030] “Acyl” refers to a group -C(O)Ry, wherein Ry is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein. Examples of acyl include, e.g., formyl, acetyl, cyclohexylcarbonyl, cyclohexylmethyl- carbonyl, and benzoyl. [0031] “Amido” refers to both a “C-amido” group which refers to the group -C(O)NRyRz and an “N-amido” group which refers to the group -NRyC(O)Rz, wherein Ry and Rz are independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein, or Ry and Rz are taken together to form a cycloalkyl or heterocyclyl; each of which may be unsubstituted or substituted, as defined herein. [0032] “Amino” refers to the group -NRyRz wherein Ry and Rz are independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein. [0033] “Amidino” refers to -C(NRy)(NRz2), wherein Ry and Rz are independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein. [0034] “Aryl” refers to an aromatic carbocyclic group having a single ring (e.g., monocyclic) or multiple rings (e.g., bicyclic or tricyclic) including fused systems. As used herein, aryl has 6 to 20 ring carbon atoms (i.e., C6-20 aryl), 6 to 12 carbon ring atoms (i.e., C6-12 aryl), or 6 to 10 carbon ring atoms (i.e., C6-10 aryl). Examples of aryl groups include, e.g., phenyl, naphthyl, fluorenyl, and anthryl. Aryl, however, does not encompass or overlap in any way with heteroaryl defined below. If one or more aryl groups are fused with a heteroaryl, the resulting ring system is heteroaryl regardless of point of attachment. If one or more aryl groups are fused with a heterocyclyl, the resulting ring system is heterocyclyl regardless of point of attachment. If one or more aryl groups are fused with a cycloalkyl, the resulting ring system is cycloalkyl regardless of point of attachment. [0035] “Carbamoyl” refers to both an “O-carbamoyl” group which refers to the group -O-C(O)NRyRz and an “N-carbamoyl” group which refers to the group -NRyC(O)ORz, wherein Ry and Rz are independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein. [0036] “Carboxyl ester” or “ester” refer to both -OC(O)Rx and -C(O)ORx, wherein Rx is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein. [0037] “Cycloalkyl” refers to a saturated or partially unsaturated cyclic alkyl group having a single ring or multiple rings including fused, bridged, and spiro ring systems. The term “cycloalkyl” includes cycloalkenyl groups (i.e., the cyclic group having at least one double bond) and carbocyclic fused ring systems having at least one sp3 carbon atom (i.e., at least one non-aromatic ring). As used herein, cycloalkyl has from 3 to 20 ring carbon atoms (i.e., C3-20 cycloalkyl), 3 to 14 ring carbon atoms (i.e., C3-14 cycloalkyl), 3 to 12 ring carbon atoms (i.e., C3-12 cycloalkyl), 3 to 10 ring carbon atoms (i.e., C3-10 cycloalkyl), 3 to 8 ring carbon atoms (i.e., C3-8 cycloalkyl), or 3 to 6 ring carbon atoms (i.e., C3-6 cycloalkyl). Monocyclic groups include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Polycyclic groups include, for example, bicyclo[2.2.1]heptanyl, bicyclo[2.2.2]octanyl, adamantyl, norbornyl, decalinyl, 7,7-dimethyl-bicyclo[2.2.1]heptanyl, and the like. Further, the term cycloalkyl is intended to encompass any non-aromatic ring which may be fused to an aryl ring, regardless of the attachment to the remainder of the molecule. Still further, cycloalkyl also includes “spirocycloalkyl” when there are two positions for substitution on the same carbon atom, for example spiro[2.5]octanyl, spiro[4.5]decanyl, or spiro[5.5]undecanyl. [0038] “Imino” refers to a group -C(NRy)Rz, wherein Ry and Rz are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein. [0039] “Imido” refers to a group -C(O)NRyC(O)Rz, wherein Ry and Rz are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein. [0040] “Halogen” or “halo” refers to atoms occupying group VIIA of the periodic table, such as fluoro, chloro, bromo, or iodo. [0041] “Haloalkyl” refers to an unbranched or branched alkyl group as defined above, wherein one or more (e.g., 1 to 6 or 1 to 3) hydrogen atoms are replaced by a halogen. For example, where a residue is substituted with more than one halogen, it may be referred to by using a prefix corresponding to the number of halogen moieties attached. Dihaloalkyl and trihaloalkyl refer to alkyl substituted with two (“di”) or three (“tri”) halo groups, which may be, but are not necessarily, the same halogen. Examples of haloalkyl include, e.g., trifluoromethyl, difluoromethyl, fluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1,2-difluoroethyl, 3-bromo-2-fluoropropyl, 1,2-dibromoethyl, and the like. [0042] “Haloalkoxy” refers to an alkoxy group as defined above, wherein one or more (e.g., 1 to 6 or 1 to 3) hydrogen atoms are replaced by a halogen. [0043] “Hydroxyalkyl” refers to an alkyl group as defined above, wherein one or more (e.g., 1 to 6 or 1 to 3) hydrogen atoms are replaced by a hydroxy group. [0044] “Heteroalkyl” refers to an alkyl group in which one or more of the carbon atoms (and any associated hydrogen atoms), excluding any terminal carbon atom(s), are each independently replaced with the same or different heteroatomic group, provided the point of attachment to the remainder of the molecule is through a carbon atom. The term “heteroalkyl” includes unbranched or branched saturated chain having carbon and heteroatoms. By way of example, 1, 2 or 3 carbon atoms may be independently replaced with the same or different heteroatomic group. Heteroatomic groups include, but are not limited to, -NRy-, -O-, -S-, -S(O)-, -S(O)2-, and the like, wherein Ry is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein. Examples of heteroalkyl groups include, e.g., ethers (e.g., -CH2OCH3, -CH(CH3)OCH3, -CH2CH2OCH3, -CH2CH2OCH2CH2OCH3, etc.), thioethers (e.g., -CH2SCH3, -CH(CH3)SCH3, -CH2CH2SCH3,-CH2CH2SCH2CH2SCH3, etc.), sulfones (e.g., -CH2S(O)2CH3, -CH(CH3)S(O)2CH3, -CH2CH2S(O)2CH3, -CH2CH2S(O)2CH2CH2OCH3, etc.), and amines (e.g., -CH2NRyCH3, -CH(CH3)NRyCH3, -CH2CH2NRyCH3, -CH2CH2NRyCH2CH2NRyCH3, etc., where Ry is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein). As used herein, heteroalkyl includes 2 to 10 carbon atoms, 2 to 8 carbon atoms, or 2 to 4 carbon atoms; and 1 to 3 heteroatoms, 1 to 2 heteroatoms, or 1 heteroatom. [0045] “Heteroaryl” refers to an aromatic group having a single ring, multiple rings or multiple fused rings, with one or more ring heteroatoms independently selected from nitrogen, oxygen, and sulfur. As used herein, heteroaryl includes 1 to 20 ring carbon atoms (i.e., C1-20 heteroaryl), 3 to 12 ring carbon atoms (i.e., C3-12 heteroaryl), or 3 to 8 carbon ring atoms (i.e., C3-8 heteroaryl), and 1 to 5 ring heteroatoms, 1 to 4 ring heteroatoms, 1 to 3 ring heteroatoms, 1 to 2 ring heteroatoms, or 1 ring heteroatom independently selected from nitrogen, oxygen, and sulfur. In certain instances, heteroaryl includes 5-10 membered ring systems, 5-7 membered ring systems, or 5-6 membered ring systems, each independently having 1 to 4 ring heteroatoms, 1 to 3 ring heteroatoms, 1 to 2 ring heteroatoms, or 1 ring heteroatom independently selected from nitrogen, oxygen, and sulfur. Examples of heteroaryl groups include, e.g., acridinyl, benzimidazolyl, benzothiazolyl, benzindolyl, benzofuranyl, benzothiazolyl, benzothiadiazolyl, benzonaphthofuranyl, benzoxazolyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[1,2-a]pyridyl, carbazolyl, cinnolinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, isoquinolyl, isoxazolyl, naphthyridinyl, oxadiazolyl, oxazolyl, 1-oxidopyridinyl, 1-oxidopyrimidinyl, 1-oxidopyrazinyl, 1-oxidopyridazinyl, phenazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinazolinyl, quinoxalinyl, quinolinyl, quinuclidinyl, isoquinolinyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, and triazinyl. Examples of the fused-heteroaryl rings include, but are not limited to, benzo[d]thiazolyl, quinolinyl, isoquinolinyl, benzo[b]thiophenyl, indazolyl, benzo[d]imidazolyl, pyrazolo[1,5-a]pyridinyl, and imidazo[1,5-a]pyridinyl, where the heteroaryl can be bound via either ring of the fused system. Any aromatic ring, having a single or multiple fused rings, containing at least one heteroatom, is considered a heteroaryl regardless of the attachment to the remainder of the molecule (i.e., through any one of the fused rings). Heteroaryl does not encompass or overlap with aryl as defined above. [0046] “Heterocyclyl” – used interchangeably with “heterocycloalkyl”- refers to a saturated or partially unsaturated cyclic alkyl group, with one or more ring heteroatoms independently selected from nitrogen, oxygen, and sulfur. The term “heterocyclyl” includes heterocycloalkenyl groups (i.e., the heterocyclyl group having at least one double bond), bridged-heterocyclyl groups, fused-heterocyclyl groups, and spiro-heterocyclyl groups. A heterocyclyl may be a single ring or multiple rings wherein the multiple rings may be fused, bridged, or spiro, and may comprise one or more (e.g., 1 to 3) oxo (=O) or N-oxide (-O-) moieties. Any non-aromatic ring containing at least one heteroatom is considered a heterocyclyl, regardless of the attachment (i.e., can be bound through a carbon atom or a heteroatom). Further, the term heterocyclyl is intended to encompass any non-aromatic ring containing at least one heteroatom, which ring may be fused to a cycloalkyl, an aryl, or heteroaryl ring, regardless of the attachment to the remainder of the molecule. As used herein, heterocyclyl has 2 to 20 ring carbon atoms (i.e., C2-20 heterocyclyl), 2 to 12 ring carbon atoms (i.e., C2-12 heterocyclyl), 2 to 10 ring carbon atoms (i.e., C2-10 heterocyclyl), 2 to 8 ring carbon atoms (i.e., C2-8 heterocyclyl), 3 to 12 ring carbon atoms (i.e., C3-12 heterocyclyl), 3 to 8 ring carbon atoms (i.e., C3-8 heterocyclyl), or 3 to 6 ring carbon atoms (i.e., C3-6 heterocyclyl); having 1 to 5 ring heteroatoms, 1 to 4 ring heteroatoms, 1 to 3 ring heteroatoms, 1 to 2 ring heteroatoms, or 1 ring heteroatom independently selected from nitrogen, sulfur, or oxygen. Examples of heterocyclyl groups include, e.g., azetidinyl, azepinyl, benzodioxolyl, benzo[b][1,4]dioxepinyl, 1,4- benzodioxanyl, benzopyranyl, benzodioxinyl, benzopyranonyl, benzofuranonyl, dioxolanyl, dihydropyranyl, hydropyranyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, furanonyl, imidazolinyl, imidazolidinyl, indolinyl, indolizinyl, isoindolinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, oxiranyl, oxetanyl, phenothiazinyl, phenoxazinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, tetrahydropyranyl, trithianyl, tetrahydroquinolinyl, thiophenyl (i.e., thienyl), thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl. The term “heterocyclyl” also includes “spiroheterocyclyl” when there are two positions for substitution on the same carbon atom. Examples of the spiro-heterocyclyl rings include, e.g., bicyclic and tricyclic ring systems, such as oxabicyclo[2.2.2]octanyl, 2-oxa-7-azaspiro[3.5]nonanyl, 2-oxa-6-azaspiro[3.4]octanyl, and 6-oxa-1-azaspiro[3.3]heptanyl. Examples of the fused-heterocyclyl rings include, but are not limited to, 1,2,3,4-tetrahydroisoquinolinyl, 4,5,6,7-tetrahydrothieno[2,3- c]pyridinyl, indolinyl, and isoindolinyl, where the heterocyclyl can be bound via either ring of the fused system. In some embodiments, the heterocycloalkyl may be substituted with oxo group(s) on a heteroatom (e.g., S=O, S(=O)2). [0047] “Oxime” refers to the group -CRy(=NOH) wherein Ry is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein. [0048] “Oxo” refers to the moiety =O. [0049] “Sulfonyl” refers to the group -S(O)2Ry, where Ry is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein. Examples of sulfonyl are methylsulfonyl, ethylsulfonyl, phenylsulfonyl, and toluenesulfonyl. [0050] “Sulfinyl” refers to the group -S(O)Ry, where Ry is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein. Examples of sulfinyl are methylsulfinyl, ethylsulfinyl, phenylsulfinyl, and toluenesulfinyl. [0051] “Sulfonamido” refers to the groups -SO2NRyRz and -NRySO2Rz, where Ry and Rz are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroalkyl, or heteroaryl; each of which may be unsubstituted or substituted, as defined herein. [0052] The terms “optional” or “optionally” means that the subsequently described event or circumstance may or may not occur and that the description includes instances where said event or circumstance occurs and instances in which it does not. Also, the term “unsubstituted or substituted” refers to any one or more (e.g., 1 to 5 or 1 to 3) hydrogen atoms on the designated atom or group may or may not be replaced by a moiety other than hydrogen. [0053] The term “substituted” used herein means any of the above groups (i.e., alkyl, alkenyl, alkynyl, alkylene, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, aryl, heterocyclyl, heteroaryl, and/or heteroalkyl) wherein at least one (e.g., 1 to 5 or 1 to 3) hydrogen atom is replaced by a bond to a non-hydrogen atom such as, but not limited to alkyl, alkenyl, alkynyl, alkoxy, alkylthio, acyl, amido, amino, amidino, aryl, aralkyl, azido, carbamoyl, carboxyl, carboxyl ester, cyano, cycloalkyl, cycloalkylalkyl, guanadino, halo, haloalkyl, haloalkoxy, hydroxyalkyl, heteroalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, -NHNH2, =NNH2, imino, imido, hydroxy, oxo, oxime, nitro, sulfonyl, sulfinyl, alkylsulfonyl, alkylsulfinyl, thiocyanate, -S(O)OH, -S(O)2OH, sulfonamido, thiol, thioxo, N-oxide, or -Si(Ry)3, wherein each Ry is independently hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, cycloalkyl, aryl, heteroaryl, or heterocyclyl. [0054] In certain embodiments, “substituted” includes any of the above alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl groups in which one or more (e.g., 1 to 5 or 1 to 3) hydrogen atoms are independently replaced with deuterium, halo, cyano, nitro, azido, oxo, alkyl, alkenyl, alkynyl, haloalkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -NRgRh, -NRgC(O)Rh, -NRgC(O)NRgRh, -NRgC(O)ORh, -NRgS(O)1-2Rh, -C(O)Rg, -C(O)ORg, -OC(O)ORg, -OC(O)Rg, -C(O)NRgRh, -OC(O)NRgRh, -ORg, -SRg, -S(O)Rg, -S(O)2Rg, -OS(O)1-2Rg, -S(O)1-2ORg, -NRgS(O)1-2NRgRh, =NSO2Rg, =NORg, -S(O)1-2NRgRh, -SF5, -SCF3, or -OCF3. In certain embodiments, “substituted” also means any of the above groups in which one or more (e.g., 1 to 5 or 1 to 3) hydrogen atoms are replaced with -C(O)Rg, -C(O)ORg, -C(O)NRgRh, -CH2SO2Rg, or -CH2SO2NRgRh. In the foregoing, Rg and Rh are the same or different and independently hydrogen, alkyl, alkenyl, alkynyl, alkoxy, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, and/or heteroarylalkyl. In certain embodiments, “substituted” also means any of the above groups in which one or more (e.g., 1 to 5 or 1 to 3) hydrogen atoms are replaced by a bond to an amino, cyano, hydroxy, imino, nitro, oxo, thioxo, halo, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, and/or heteroarylalkyl, or two of Rg and Rh are taken together with the atoms to which they are attached to form a heterocyclyl ring unsubstituted or substituted with oxo, halo, or alkyl unsubstituted or substituted with oxo, halo, amino, hydroxy, or alkoxy. [0055] Polymers or similar indefinite structures arrived at by defining substituents with further substituents appended ad infinitum (e.g., a substituted aryl having a substituted alkyl which is itself substituted with a substituted aryl group, which is further substituted by a substituted heteroalkyl group, etc.) are not intended for inclusion herein. Unless otherwise noted, the maximum number of serial substitutions in compounds described herein is three. For example, serial substitutions of substituted aryl groups with two other substituted aryl groups are limited to ((substituted aryl)substituted aryl) substituted aryl. Similarly, the above definitions are not intended to include impermissible substitution patterns (e.g., methyl substituted with 5 fluorines or heteroaryl groups having two adjacent oxygen ring atoms). Such impermissible substitution patterns are well known to the skilled artisan. When used to modify a chemical group, the term “substituted” may describe other chemical groups defined herein. [0056] In certain embodiments, as used herein, the phrase “one or more” refers to one to five. In certain embodiments, as used herein, the phrase “one or more” refers to one to three. [0057] Any compound or structure given herein, is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds. These forms of compounds may also be referred to as “isotopically enriched analogs.” Isotopically labeled compounds have structures depicted herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Examples of isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2H, 3H, 11C, 13C, 14C, 13N, 15N, 15O, 17O, 18O, 31P, 32P, 35S, 18F, 36Cl, 123I, and 125I, respectively. Various isotopically labeled compounds of the present disclosure, for example those into which radioactive isotopes such as 3H and 14C are incorporated. Such isotopically labelled compounds may be useful in metabolic studies, reaction kinetic studies, detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays or in radioactive treatment of patients. [0058] The term “isotopically enriched analogs” includes “deuterated analogs” of compounds described herein in which one or more hydrogens is/are replaced by deuterium, such as a hydrogen on a carbon atom. Such compounds exhibit increased resistance to metabolism and are thus useful for increasing the half-life of any compound when administered to a mammal, particularly a human. See, for example, Foster, “Deuterium Isotope Effects in Studies of Drug Metabolism,” Trends Pharmacol. Sci.5(12): 524-527 (1984). Such compounds are synthesized by means well known in the art, for example by employing starting materials in which one or more hydrogens have been replaced by deuterium. [0059] Deuterium labelled or substituted therapeutic compounds of the disclosure may have improved DMPK (drug metabolism and pharmacokinetics) properties, relating to absorption, distribution, metabolism, and excretion (ADME). Substitution with heavier isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life, reduced dosage requirements, and/or an improvement in therapeutic index. An 18F, 3H, or 11C labeled compound may be useful for PET or SPECT or other imaging studies. Isotopically labeled compounds of this disclosure and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent. It is understood that deuterium in this context is regarded as a substituent in a compound described herein. [0060] The concentration of such a heavier isotope, specifically deuterium, may be defined by an isotopic enrichment factor. In the compounds of this disclosure any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom. Unless otherwise stated, when a position is designated specifically as “H” or “hydrogen,” the position is understood to have hydrogen at its natural abundance isotopic composition. Accordingly, in the compounds of this disclosure any atom specifically designated as a deuterium (D) is meant to represent deuterium. [0061] In many cases, the compounds of this disclosure are capable of forming acid and/or base salts by virtue of the presence of amino, and/or carboxyl groups, or groups similar thereto. [0062] Provided are also or a pharmaceutically acceptable salt, isotopically enriched analog, deuterated analog, stereoisomer, mixture of stereoisomers, and prodrugs of the compounds described herein. “Pharmaceutically acceptable” or “physiologically acceptable” refer to compounds, salts, compositions, dosage forms, and other materials which are useful in preparing a pharmaceutical composition that is suitable for veterinary or human pharmaceutical use. [0063] The term “pharmaceutically acceptable salt” of a given compound refers to salts that retain the biological effectiveness and properties of the given compound and which are not biologically or otherwise undesirable. “Pharmaceutically acceptable salts” or “physiologically acceptable salts” include, for example, salts with inorganic acids, and salts with an organic acid. In addition, if the compounds described herein are obtained as an acid addition salt, the free base can be obtained by basifying a solution of the acid salt. Conversely, if the product is a free base, an addition salt, particularly a pharmaceutically acceptable addition salt, may be produced by dissolving the free base in a suitable organic solvent and treating the solution with an acid, in accordance with conventional procedures for preparing acid addition salts from base compounds. Those skilled in the art will recognize various synthetic methodologies that may be used to prepare nontoxic pharmaceutically acceptable addition salts. Pharmaceutically acceptable acid addition salts may be prepared from inorganic or organic acids. Salts derived from inorganic acids include, e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Salts derived from organic acids include, e.g., acetic acid, propionic acid, gluconic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluene-sulfonic acid, salicylic acid, and the like. Likewise, pharmaceutically acceptable base addition salts can be prepared from inorganic or organic bases. Salts derived from inorganic bases include, by way of example only, sodium, potassium, lithium, aluminum, ammonium, calcium, and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, such as alkyl amines (i.e., NH2(alkyl)), dialkyl amines (i.e., HN(alkyl)2), trialkyl amines (i.e., N(alkyl)3), substituted alkyl amines (i.e., NH2(substituted alkyl)), di(substituted alkyl) amines (i.e., HN(substituted alkyl)2), tri(substituted alkyl) amines (i.e., N(substituted alkyl)3), alkenyl amines (i.e., NH2(alkenyl)), dialkenyl amines (i.e., HN(alkenyl)2), trialkenyl amines (i.e., N(alkenyl)3), substituted alkenyl amines (i.e., NH2(substituted alkenyl)), di(substituted alkenyl) amines (i.e., HN(substituted alkenyl)2), tri(substituted alkenyl) amines (i.e., N(substituted alkenyl)3, mono-, di- or tri- cycloalkyl amines (i.e., NH2(cycloalkyl), HN(cycloalkyl)2, N(cycloalkyl)3), mono-, di- or tri- arylamines (i.e., NH2(aryl), HN(aryl)2, N(aryl)3), or mixed amines, etc. Specific examples of suitable amines include, by way of example only, isopropylamine, trimethyl amine, diethyl amine, tri(iso-propyl) amine, tri(n-propyl) amine, ethanolamine, 2-dimethylaminoethanol, piperazine, piperidine, morpholine, N-ethylpiperidine, and the like. [0064] Some of the compounds exist as tautomers. Tautomers are in equilibrium with one another. For example, amide containing compounds may exist in equilibrium with imidic acid tautomers. Regardless of which tautomer is shown and regardless of the nature of the equilibrium among tautomers, the compounds are understood by one of ordinary skill in the art to comprise both amide and imidic acid tautomers. Thus, the amide containing compounds are understood to include their imidic acid tautomers. Likewise, the imidic acid containing compounds are understood to include their amide tautomers. [0065] The compounds, or their pharmaceutically acceptable salts include an asymmetric center and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids. The present disclosure is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (-), (R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and/or fractional crystallization. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. [0066] A “stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present disclosure contemplates various stereoisomers, or mixtures thereof, and includes “enantiomers,” which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another. [0067] “Diastereomers” are stereoisomers that have at least two asymmetric atoms, but which are not mirror-images of each other. [0068] Relative centers of the compounds as depicted herein are indicated graphically using the “thick bond” style (bold or parallel lines) and absolute stereochemistry is depicted using wedge bonds (bold or parallel lines). [0069] “Prodrugs” means any compound which releases an active parent drug according to a structure described herein in vivo when such prodrug is administered to a mammalian subject. Prodrugs of a compound described herein are prepared by modifying functional groups present in the compound described herein in such a way that the modifications may be cleaved in vivo to release the parent compound. Prodrugs may be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds. Prodrugs include compounds described herein wherein a hydroxy, amino, carboxyl, or sulfhydryl group in a compound described herein is bonded to any group that may be cleaved in vivo to regenerate the free hydroxy, amino, or sulfhydryl group, respectively. Examples of prodrugs include, but are not limited to esters (e.g., acetate, formate, and benzoate derivatives), amides, guanidines, carbamates (e.g., N,N-dimethylaminocarbonyl) of hydroxy functional groups in compounds described herein, and the like. Preparation, selection, and use of prodrugs is discussed in T. Higuchi and V. Stella, “Pro-drugs as Novel Delivery Systems,” Vol. 14 of the A.C.S. Symposium Series; “Design of Prodrugs,” ed. H. Bundgaard, Elsevier, 1985; and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, each of which are hereby incorporated by reference in their entirety. [0070] As used herein, a BRD4 “modulator” may bind to BRD4, but not degrade BRD4. As used herein, a BRD4 “degrader” binds to BRD4 and also degrades BRD4. Compounds [0071] In one embodiment, this disclosure provides a compound of formula I:
Figure imgf000016_0001
or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein: m is an integer from 0 to 4; n is an integer from 0 to 4; Y is CH(R4), NR5, or O; each of X1 and X2 is independently CH(R4) or NR5; Q is H, -CH3, -CH2OH, -CH(CH3)N(R13)2, -CH2N(R13)2, -CH2N(R13)C(O)R1, or -CH2CH2N(R13)2; R1 is C1-C6 alkyl, C3-C10 cycloalkyl, aryl, heteroaryl, or heterocyclyl, wherein each C1-C6 alkyl, C3-C10 cycloalkyl, aryl, heteroaryl, or heterocyclyl is independently unsubstituted or substituted with one to five Z1a; each of R2 and R3 is independently selected from halo, -N(R13)2, -OR14, C1-C6 alkyl, aryl, cyano, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, guanidino, and nitro, wherein each C1-C6 alkyl, aryl, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, and guanidino is independently unsubstituted or substituted with one to five Z1a; each R4 is independently H, halo, cyano, hydroxy, -SH, -N(R13)2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl, wherein each of C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each R5 is independently H, -C(O)R13, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl, wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each Z1a is independently halo, cyano, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -N(R13)2, -OR13, -C(O)R13, -C(O)OR13, -S(O)0-2R13, -NR13S(O)1-2-R13, -S(O)1-2N(R13)2, -NR13S(O)1-2N(R13)2, -NR13C(O)N(R13)2, -C(O)N(R13)2, -NR13C(O)R13, -OC(O)N(R13)2, or -NR13C(O)OR13; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl is independently unsubstituted or substituted with one to five Z1b; each R13 is independently H, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R13 is independently unsubstituted or substituted with one to five Z1b; each R14 is independently H, C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R14 is independently unsubstituted or substituted with one to five Z1b; each Z1b is independently halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -L-C1-6 alkyl, -L-C2-6 alkenyl, -L-C2-6 alkynyl, -L-C1-6 haloalkyl, -L-C3-10 cycloalkyl, -L-heterocyclyl, -L-aryl, or -L-heteroaryl; each L is independently -O-, -NH-, -S-, -S(O)-, -S(O)2-, -N(C1-C6 alkyl)-, -N(C2-C6 alkenyl), -N(C2-C6 alkynyl)-, -N(C1-C6 haloalkyl)-, -N(C3-10 cycloalkyl)-, -N(heterocyclyl)-, -N(aryl)-, -N(heteroaryl)-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)N(C1-C6 alkyl)-, -C(O)N(C2-C6 alkenyl)-, -C(O)N(C2-C6 alkynyl)-, -C(O)N(C1-6 haloalkyl)-, -C(O)N(C3-10 cycloalkyl)-, -C(O)N(heterocyclyl)-, -C(O)N(aryl)-, -C(O)N(heteroaryl)-, -NHC(O)-, -NHC(O)O-, -NHC(O)NH-, -NHS(O)-, or -S(O)2NH-; and wherein each C1-C6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, and heteroaryl of Z1b and L is further independently unsubstituted or substituted with one to five halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl. [0072] In some embodiments, this disclosure provides a compound of formula I as described above, provided that the compound at 1 µM concentration degrades BRD4 by 30% or more. [0073] In some embodiments, this disclosure provides a compound of formula IA: or a pharmaceutically a
Figure imgf000018_0001
cceptable salt, solvate, stereoisomer, or tautomer thereof, wherein: m is an integer from 0 to 4; n is an integer from 0 to 4; Y is CH(R4), NR5, or O; each of X1 and X2 is independently CH(R4) or NR5; each R2 is independently selected from halo, -N(R13)2, -OR14, C1-C6 alkyl, aryl, cyano, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, guanidino, and nitro, wherein each C1-C6 alkyl, aryl, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, and guanidino is independently unsubstituted or substituted with one to five Z1a, provided that R2 is not imidazolyl, isooxazolyl, pyrazol-4-yl, or pyrimidin-5-yl; each R3 is independently selected from halo, -N(R13)2, -OR14, C1-C6 alkyl, aryl, cyano, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, guanidino, and nitro, wherein each C1-C6 alkyl, aryl, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, and guanidino is independently unsubstituted or substituted with one to five Z1a; each R4 is independently H, halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl, wherein each of C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each R5 is independently H, -C(O)R13, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl, wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each Z1a is independently halo, cyano, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -N(R13)2, -OR13, -C(O)R13, -C(O)OR13, -S(O)0-2R13, -NR13S(O)1-2-R13, -S(O)1-2N(R13)2, -NR13S(O)1-2N(R13)2, -NR13C(O)N(R13)2, -C(O)N(R13)2, -NR13C(O)R13, -OC(O)N(R13)2, or -NR13C(O)OR13; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl is independently unsubstituted or substituted with one to five Z1b; each R13 is independently H, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R13 is independently unsubstituted or substituted with one to five Z1b; each R14 is independently H, C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R14 is independently unsubstituted or substituted with one to five Z1b; each Z1b is independently halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -L-C1-6 alkyl, -L-C2-6 alkenyl, -L-C2-6 alkynyl, -L-C1-6 haloalkyl, -L-C3-10 cycloalkyl, -L-heterocyclyl, -L-aryl, or -L-heteroaryl; each L is independently -O-, -NH-, -S-, -S(O)-, -S(O)2-, -N(C1-C6 alkyl)-, -N(C2-C6 alkenyl), -N(C2-C6 alkynyl)-, -N(C1-C6 haloalkyl)-, -N(C3-10 cycloalkyl)-, -N(heterocyclyl)-, -N(aryl)-, -N(heteroaryl)-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)N(C1-C6 alkyl)-, -C(O)N(C2-C6 alkenyl)-, -C(O)N(C2-C6 alkynyl)-, -C(O)N(C1-6 haloalkyl)-, -C(O)N(C3-10 cycloalkyl)-, -C(O)N(heterocyclyl)-, -C(O)N(aryl)-, -C(O)N(heteroaryl)-, -NHC(O)-, -NHC(O)O-, -NHC(O)NH-, -NHS(O)-, or -S(O)2NH-; and wherein each C1-C6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, and heteroaryl of Z1b and L is further independently unsubstituted or substituted with one to five halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl, provided that the following compounds are excluded: , , ,
Figure imgf000020_0001
Figure imgf000021_0001
[0074] In some embodiments, the compound of formula I is represented by formula IB:
Figure imgf000022_0001
or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein Y, R2, R3, R4, m, and n are as defined herein. [0075] In some embodiments, the compound of formula IB is represented by formula IC: or a pharmaceutically accep
Figure imgf000022_0002
table salt, solvate, stereoisomer, or tautomer thereof, wherein R2, R3, R4, m, and n are as defined herein. [0076] In some embodiments, the compound of formula IC is represented by formula ID:
Figure imgf000022_0003
or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein R2, R3, R4, and n are as defined herein. [0077] In some embodiments, this disclosure provides a compound of formula I, IA, IB, IC, or ID, wherein each R4 independently is C1-C6 alkyl. In some embodiments, this disclosure provides a compound of formula ID, wherein R4 is C1-C6 alkyl and n is 0. [0078] In some embodiments, the compound of formula ID is represented by formula IE: NH
Figure imgf000023_0001
or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein R2 is as defined herein. [0079] In some embodiments, this disclosure provides a compound of formula I, IA, IB, or IC, wherein each R2 independently is H, halo, C1-C6 alkyl, phenyl substituted with C1-C6 alkoxy, pyridyl, or 1-methylpyrazol-5-yl. In some embodiments, this disclosure provides a compound of formula ID or IE, wherein each R2 independently is H, halo, C1-C6 alkyl, phenyl substituted with C1-C6 alkoxy, pyridyl, or 1-methylpyrazol-5-yl. [0080] In some embodiments, this disclosure provides a compound selected from Table 1 or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof.
Table 1.
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0002
[0081] In some embodiments, this disclosure provides a compound selected from Table 2, or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof. Compounds of Table 2 degrade BRD4 by less than 30% at 1 µM concentration. Table 2 C d
Figure imgf000026_0001
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
[0082] In one embodiment, this disclosure provides a compound of formula I-1:
Figure imgf000037_0001
or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein: m is an integer from 0 to 4; n is an integer from 0 to 4; each of X1 and X2 is independently CH(R4) or NR5; Q is H, -CH2OH, -CH2N(R13)2, -CH2CH2N(R13)2, -CH2N(R13)C(O)R1, or heterocyclyl; R1 is C1-C6 alkyl, C3-C10 cycloalkyl, aryl, heteroaryl, or heterocyclyl, wherein each C1-C6 alkyl, C3-C10 cycloalkyl, aryl, heteroaryl, or heterocyclyl is independently unsubstituted or substituted with one to five Z1a; each of R2 and R3 is independently selected from halo, -N(R13)2, -OR14, C1-C6 alkyl, aryl, cyano, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, guanidino, and nitro, wherein each C1-C6 alkyl, aryl, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, and guanidino is independently unsubstituted or substituted with one to five Z1a; each R4 is independently H, halo, cyano, hydroxy, -SH, -N(R13)2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl, wherein each of C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each R5 is independently H, -C(O)R13, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl, wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each Z1a is independently halo, cyano, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -N(R13)2, -OR13, -C(O)R13, -C(O)OR13, -S(O)0-2R13, -NR13S(O)1-2-R13, -S(O)1-2N(R13)2, -NR13S(O)1-2N(R13)2, -NR13C(O)N(R13)2, -C(O)N(R13)2, -NR13C(O)R13, -OC(O)N(R13)2, or -NR13C(O)OR13; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl is independently unsubstituted or substituted with one to five Z1b; each R13 is independently H, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R13 is independently unsubstituted or substituted with one to five Z1b; each R14 is independently H, C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R14 is independently unsubstituted or substituted with one to five Z1b; each Z1b is independently halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -L-C1-6 alkyl, -L-C2-6 alkenyl, -L-C2-6 alkynyl, -L-C1-6 haloalkyl, -L-C3-10 cycloalkyl, -L-heterocyclyl, -L-aryl, or -L-heteroaryl; each L is independently -O-, -NH-, -S-, -S(O)-, -S(O)2-, -N(C1-C6 alkyl)-, -N(C2-C6 alkenyl), -N(C2-C6 alkynyl)-, -N(C1-C6 haloalkyl)-, -N(C3-10 cycloalkyl)-, -N(heterocyclyl)-, -N(aryl)-, -N(heteroaryl)-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)N(C1-C6 alkyl)-, -C(O)N(C2-C6 alkenyl)-, -C(O)N(C2-C6 alkynyl)-, -C(O)N(C1-6 haloalkyl)-, -C(O)N(C3-10 cycloalkyl)-, -C(O)N(heterocyclyl)-, -C(O)N(aryl)-, -C(O)N(heteroaryl)-, -NHC(O)-, -NHC(O)O-, -NHC(O)NH-, -NHS(O)-, or -S(O)2NH-; and wherein each C1-C6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, and heteroaryl of Z1b and L is further independently unsubstituted or substituted with one to five halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl. [0083] In some embodiments, this disclosure provides a compound of formula I-1 as described above, provided that the compound at 1 µM concentration degrades BRD4 by 30% or more. [0084] In some embodiments, this disclosure provides a compound of formula IA-1:
or a pharmaceutically acc
Figure imgf000039_0001
eptable salt, solvate, stereoisomer, or tautomer thereof, wherein: m is an integer from 0 to 4; n is an integer from 0 to 4; each of X1 and X2 is independently CH(R4) or NR5; each of R2 and R3 is independently selected from halo, -N(R13)2, -OR14, C1-C6 alkyl, aryl, cyano, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, guanidino, and nitro, wherein each C1-C6 alkyl, aryl, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, and guanidino is independently unsubstituted or substituted with one to five Z1a; each R4 is independently H, halo, cyano, hydroxy, -SH, -N(R13)2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl, wherein each of C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each R5 is independently H, -C(O)R13, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl, wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each Z1a is independently halo, cyano, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -N(R13)2, -OR13, -C(O)R13, -C(O)OR13, -S(O)0-2R13, -NR13S(O)1-2-R13, -S(O)1-2N(R13)2, -NR13S(O)1-2N(R13)2, -NR13C(O)N(R13)2, -C(O)N(R13)2, -NR13C(O)R13, -OC(O)N(R13)2, or -NR13C(O)OR13; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl is independently unsubstituted or substituted with one to five Z1b; each R13 is independently H, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R13 is independently unsubstituted or substituted with one to five Z1b; each R14 is independently H, C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R14 is independently unsubstituted or substituted with one to five Z1b; each Z1b is independently halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -L-C1-6 alkyl, -L-C2-6 alkenyl, -L-C2-6 alkynyl, -L-C1-6 haloalkyl, -L-C3-10 cycloalkyl, -L-heterocyclyl, -L-aryl, or -L-heteroaryl; each L is independently -O-, -NH-, -S-, -S(O)-, -S(O)2-, -N(C1-C6 alkyl)-, -N(C2-C6 alkenyl), -N(C2-C6 alkynyl)-, -N(C1-C6 haloalkyl)-, -N(C3-10 cycloalkyl)-, -N(heterocyclyl)-, -N(aryl)-, -N(heteroaryl)-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)N(C1-C6 alkyl)-, -C(O)N(C2-C6 alkenyl)-, -C(O)N(C2-C6 alkynyl)-, -C(O)N(C1-6 haloalkyl)-, -C(O)N(C3-10 cycloalkyl)-, -C(O)N(heterocyclyl)-, -C(O)N(aryl)-, -C(O)N(heteroaryl)-, -NHC(O)-, -NHC(O)O-, -NHC(O)NH-, -NHS(O)-, or -S(O)2NH-; and wherein each C1-C6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, and heteroaryl of Z1b and L is further independently unsubstituted or substituted with one to five halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl, provided that the following compounds are excluded: ,
Figure imgf000040_0001
Figure imgf000041_0001
, .
Figure imgf000042_0001
[0085] In some embodiments, the compound of formula I-1 is represented by formula IB-1: [0086] or a pharmaceutically ac
Figure imgf000042_0002
ceptable salt, solvate, stereoisomer, or tautomer thereof, wherein R2, R3, R4, m, and n are as defined herein. [0087] In some embodiments, the compound of formula IB-1 is represented by formula IC-1:
Figure imgf000043_0001
or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein R4 is as defined herein. [0088] In some embodiments, this disclosure provides a compound of formula I-1, IA-1, IB-1, or IC-1, wherein each R4 independently is C1-C6 alkyl. In some embodiments, this disclosure provides a compound of formula I-1, IA-1, IB-1, or IC-1, wherein each R4 independently is methyl. In some embodiments, this disclosure provides a compound of formula I-1, IA-1, IB-1, or IC-1, wherein each R4 independently is hydrogen. In some embodiments, this disclosure provides a compound of formula I-1, IA-1, IB-1, or IC-1, wherein one R4 is methyl and the other R4 is hydrogen. In some embodiments, this disclosure provides a compound of formula IB-1, wherein R4 is C1-C6 alkyl and n is 0. [0089] In some embodiments, this disclosure provides a compound of formula I-1, IA-1, or IB-1, wherein each R2 independently is H, halo, C1-C6 alkyl, phenyl substituted with C1-C6 alkoxy, pyridyl, or 1-methylpyrazol-5-yl. [0090] In some embodiments, this disclosure provides a compound selected from Table 1A or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof. Table 1A Compound # Structure Name
Figure imgf000043_0002
Figure imgf000044_0001
[0091] In some embodiments, this disclosure provides a compound selected from Table 2A, or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof. Compounds of Table 2A degrade BRD4 by less than 30% at 1 µM concentration. Table 2A Compound # Structure Name
Figure imgf000044_0002
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
General Synthetic Methods [0092] The compounds described herein can be prepared from readily available starting materials using the following general methods and procedures. It will be appreciated that where typical process conditions (e.g., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures. [0093] Additionally, as will be apparent to those skilled in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions. Suitable protecting groups for various functional groups as well as suitable conditions for protecting and deprotecting particular functional groups are well known in the art. For example, numerous protecting groups are described in T. W. Greene and P. G. M. Wuts, Protecting Groups in Organic Synthesis, Third Edition, Wiley, New York, 1999, and references cited therein. [0094] The starting materials for the following reactions are generally known compounds or can be prepared by known procedures or obvious modifications thereof. For example, many of the starting materials are available from commercial suppliers such as Sigma Aldrich (St. Louis, Missouri, USA), Bachem (Torrance, California, USA), Emka-Chemce (St. Louis, Missouri, USA). Others may be prepared by procedures, or obvious modifications thereof, described in standard reference texts such as Fieser and Fieser’s Reagents for Organic Synthesis, Volumes 1-15 (John Wiley, and Sons, 2016), Rodd’s Chemistry of Carbon Compounds, Volumes 1-5, and Supplementals (Elsevier Science Publishers, 2001), Organic Reactions, Volumes 1-40 (John Wiley, and Sons, 2019), March’s Advanced Organic Chemistry, (John Wiley, and Sons, 8th Edition, 2019), and Larock’s Comprehensive Organic Transformations (VCH Publishers Inc., 1989). Synthesis of Representative Compounds [0095] The general synthesis of the compounds described herein is set forth in the reaction schemes below. Scheme 1 illustrates a general method for preparing compounds of formula I. In Scheme 1, substituents and variables X1, X2, R1, R2, R3, R4, Y, Q, m, and n are as defined throughout the specification. X is a suitable Suzuki cross-coupling partner for a boronic ester (including, but not limited to, Br, Cl, I, triflate, and the like).
Figure imgf000049_0001
[0096] As to the reaction in Scheme 1, the first step is an Ullmann N-Arylation reaction, wherein at least a stoichiometric equivalent of a suitable amine, compound 1, is combined with a suitably functionalized aryl halide, compound 2, in an inert diluent such as tetrahydrofuran, DMF, DMSO, and the like, typically in the presence of a copper(I) catalyst (e.g. CuI) and a suitable base such as cesium carbonate, potassium carbonate, and the like. The reaction is typically maintained at from 80° to 120°C until it is substantially complete. Conventional workup of the reaction solution can be followed by isolation / purification processes, such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 3. [0097] In the next step, the ester, compound 3, may be converted to a primary amide (Journal of Medicinal Chemistry, 2005, vol.48, # 25, p.8045 - 8054), wherein at least a stoichiometric equivalent of ammonia or ammonium hydroxide is combined with compound 3, in an inert diluent, such as MeOH, EtOH, THF, and the like. The reaction is typically maintained at from 60° to 100°C until it is substantially complete. Conventional workup of the reaction solution can be followed by isolation / purification processes, such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 4. [0098] In the next step, the free amino group of compound 4 is protected (Journal of general chemistry of the USSR, 1966, vol.36, # 8, p. 1448 – 1450), wherein at least a stoichiometric equivalent of isopropyl carbonochloridate is combined with compound 4, typically in the presence of a suitable base such as cesium carbonate, potassium carbonate, lithium tert-butoxide, and the like, in an inert diluent such as DMF, THF, dioxane, and the like. The reaction is typically maintained at from 20° to 60°C for a period of time sufficient for substantial completion of the reaction, as evidenced by e.g., thin layer chromatography. Upon reaction completion, conventional workup of the reaction solution can be followed by isolation / purification processes, such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 5. The isopropylcarbonyl group is illustrative only and other conventional amino protecting groups, such as benzyl, t-butoxycarbonyl (Boc), benzyloxycarbonyl (Cbz), p-nitrobenzyloxycarbonyl, and the like could be used. [0099] In the next step, compound 5 is cyclized under conventional reaction conditions (Journal of Medicinal Chemistry, 2014, vol.57, # 19, p.8111 – 8131) well known in the art, including the use of sodium tetrahydroborate, magnesium chloride, and lithium tert-butoxide. The reaction is typically conducted in an inert solvent such as ethanol, THF, toluene, N,N-dimethylformamide, and the like. The reaction is typically conducted at from about -10º to about 20º C for a period of time sufficient for substantial completion of the reaction, as evidenced by e.g., thin layer chromatography. Upon reaction completion, conventional workup of the reaction solution can be followed by isolation / purification processes, such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 6. [0100] In the next step, at least a stoichiometric amount of a suitably substituted carboxylic acid or carboxylic halide compound is combined with compound 6 under conventional amidation reaction conditions well known in the art, including the use of N,N-dicyclohexylcarbodiimide (DCC) as an activation agent for the carboxyl group. Other activation agents are well known in the art. The reaction is typically conducted in an inert solvent, such as chloroform, methylene chloride, toluene, N,N- dimethylformamide, and the like. The reaction is typically conducted at from about 0º to about 30º C for a period of time sufficient for substantial completion of the reaction, as evidenced by e.g., thin layer chromatography. Upon reaction completion, conventional workup of the reaction solution can be followed by isolation / purification processes, such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 7. [0101] In the next step, the isopropylcarbonyl protecting group is removed by conventional conditions, such as aluminum trichloride in dichloromethane, to provide for compound 8. The isopropylcarbonyl group is illustrative only and other conventional amino protecting groups, such as benzyl, t-butoxycarbonyl (Boc), benzyloxycarbonyl (Cbz), p-nitrobenzyloxycarbonyl, and the like could be used. Upon reaction completion, conventional workup of the reaction solution can be followed by isolation / purification processes, such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 8. [0102] In the next step, a Chan-Evans-Lam reaction (Tetrahedron Lett.39 (19): 2933–293), wherein at least a stoichiometric amount of a suitably substituted aryl boronic acid, compound 9, is combined with compound 8 in an inert diluent such as acetonitrile, dichloromethane, toluene, and the like, typically in the presence of a copper(II) catalyst (e.g. Cu(OAc)2) and a suitable base, such as pyridine, triethylamine, and the like. The reaction is typically maintained at from 20° to 80°C until it is substantially complete. Conventional workup of the reaction solution can be followed by isolation / purification processes, such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 10. [0103] In the next step, a conventional Suzuki coupling reaction, wherein at least a stoichiometric equivalent of a suitable boronic acid is combined with compound 10, in an inert diluent, such as tetrahydrofuran, dioxane, toluene, dimethoxyethane, and the like, typically in the presence of a palladium catalyst (e.g, palladium diacetate) and a suitable base, such as diisopropylethylamine, triethylamine, pyridine, potassium carbonate, and the like. The reaction is typically maintained at from 10° to 65°C until it is substantially complete. Conventional workup of the reaction solution can be followed by isolation / purification processes, such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 11. [0104] In the final step, at least a stoichiometric amount of compound 11 is combined with a terminal alkyne under conventional Sonogashira coupling conditions, in an inert diluent such as tetrahydrofuran, dioxane, DMSO, DMF, and the like, typically in the presence of a palladium catalyst (e.g. bis(triphenylphosphine)palladium (II) dichloride), a copper(I) co-catalyst (e.g. CuI), and a suitable base, such as diisopropylethylamine, triethylamine, pyridine, potassium carbonate, and the like. The reaction is typically conducted at from about 30º to about 120º C for a period of time sufficient for substantial completion of the reaction, as evidenced by e.g., thin layer chromatography. Upon reaction completion, conventional workup of the reaction solution can be followed by isolation / purification processes, such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compounds of formula I. [0105] Other starting materials used herein are either well known in the art, commercially available, or can be prepared by conventional synthetic methods. [0106] Scheme 1A illustrates a general method for preparing compounds of formula I-1. In Scheme 1A, substituents and variables X1, X2, R1, R2, and m are as defined throughout the specification. X is a suitable Suzuki cross-coupling partner for a boronic ester (including, but not limited to, Br, Cl, I, triflate, and the like).
Figure imgf000052_0001
[0107] As to the reaction in Scheme 1A, the first step is a conventional SNAR reaction wherein at least a stoichiometric equivalent of compound 1A, is combined with compound 2A, in an inert diluent such as tetrahydrofuran, dioxane, DMSO, DMF, and the like, typically in the presence of a suitable base such as diisopropylethylamine, triethylamine, pyridine, potassium carbonate, and the like. The reaction is typically maintained at from 25 °C to 100 °C until it is substantially complete. Conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 3A. [0108] In the next step, compound 3A, is reduced under conventional nitro reduction reaction conditions well known in the art including the use Fe in the presence of ammonium chloride. Other nitro reducing reagents are well known in the art. The reaction is typically conducted in an inert solvent such as EtOH, ethyl acetate, toluene, and the like. The reaction is typically conducted from about 20 ºC to about 60 ºC for a period of time sufficient for substantial completion of the reaction as evidenced by e.g., thin layer chromatography. Upon reaction completion, conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 4A. [0109] In the next step, the amine is activated as a tosyl sulfonamide. At least a stoichiometric amount of tosyl chloride is added to compound 4A, in an inert diluent such as THF, MeCN, toluene and the like in the presence of a suitable base such as triethylamine, diisopropylethylamine, pyridine and the like. The reaction is typically maintained at from 0 °C to 30 °C until it is substantially complete. Conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 5A. [0110] In the next step, a conventional Mitsunobu reaction, wherein at least a stoichiometric equivalent of 5A, under conventional reaction conditions [Hughes, D.L. (2004). The Mitsunobu Reaction, Organic Reactions, (Ed.).] well known in the art, including the use diethylazodicarboxylate and triphenylphosphine. The reaction is typically conducted in an inert solvent such as acetonitrile, THF, toluene, and the like. The reaction is typically maintained at from 0 °C to 30 °C until it is substantially complete. Conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 6A. [0111] In the next step, at least a stoichiometric amount of a suitably carboxylic acid compound, is combined with compound 6A under conventional amidation reaction conditions (Angewandte Chemie- International Edition, 2020, 59, 3028-3032) well known in the art, including the use of N,N- dicyclohexylcarbodiimide (DCC), as an activation agent for the carboxyl group. Other activation agents are well known in the art. The reaction is typically conducted in an inert solvent such as chloroform, methylene chloride, toluene, N,N-dimethylformamide, and the like. The reaction is typically conducted at from about 0 ºC to about 30 ºC for a period of time sufficient for substantial completion of the reaction as evidenced by e.g., thin layer chromatography. Upon reaction completion, conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 7A. [0112] In the next step, the tosyl (Ts) protecting group is removed by conventional conditions to provide for compound 8A. Upon reaction completion, conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like. [0113] In the next step, the amine 8A is protected with t-butoxycarbonyl (BOC) group with. At least a stoichiometric amount of a BOC anhydride is combined with compound 8A in an inert diluent such as dichloromethane, dichloroethane, THF, and the like. The reaction is typically maintained at from about 20 °C to about 50 °C until it is substantially complete. Conventional workup of the reaction solution can be followed by isolation/purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 9A. The BOC group is illustrative only and other conventional amino blocking groups such as benzyl, 9-fluorenylmethoxycarbonyl (Fmoc), benzyloxycarbonyl (Cbz), p-nitrobenzyloxycarbonyl and the like could be used. [0114] In the next step, a Miyuaura borylation reaction (J. Org. Chem., 1995, 60, 7508), at least a stoichiometric amount of compound 9A and bis(pinacolato)diboron are combined, in an inert diluent such as DMSO, 1,4-dioxane, and the like, in the presence of a suitable catalyst such as PdCl2(dppf)2, PdCl2(PPh3)2, and the like. The reaction is typically maintained at from 80 °C to 110 °C until it is substantially complete. Conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 10A. [0115] In the next step, a conventional Suzuki coupling reaction wherein at least a stoichiometric equivalent of compound 10A, is combined with a suitable aryl halide, in an inert diluent such as tetrahydrofuran, dioxane, toluene, dimethoxyethane, and the like, typically in the presence of a palladium catalyst (e.g, palladium diacetate) and a suitable base such as diisopropylethylamine, triethylamine, pyridine, potassium carbonate, and the like. The reaction is typically maintained at from 10 °C to 65 °C until it is substantially complete. Conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 11A. [0116] In the final step, the t-butoxycarbonyl (BOC) protecting group is removed by conventional conditions. The BOC group is illustrative only and other conventional amino blocking groups such as benzyl, 9-fluorenylmethoxycarbonyl (Fmoc), benzyloxycarbonyl (Cbz), p-nitrobenzyloxycarbonyl and the like. Upon reaction completion, conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 12A. [0117] Compound 12A can be used in scheme 2A.
Figure imgf000055_0001
Scheme 2A [0118] As to the reaction in Scheme 2A, the first step is a conventional alkylation reaction wherein at least a stoichiometric equivalent of a suitable amine, compound 12A (R2 = H, R1 = CH3, X1 and X2 = CH2, CAS# 6639-92-5), is combined with a suitably functionalized benzylic halide, in an inert diluent such as tetrahydrofuran, dioxane, toluene, dimethoxyethane, and the like, typically in the presence of a suitable base such as diisopropylethylamine, triethylamine, pyridine, potassium carbonate, and the like. The reaction is typically maintained at from 80 °C to 120 °C until it is substantially complete. Conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 13A. [0119] In the final step, at least a stoichiometric amount of compound 13A is combined with a terminal alkyne under conventional Sonogashira coupling conditions, in an inert diluent such as tetrahydrofuran, dioxane, DMSO, DMF, and the like, typically in the presence of a palladium catalyst (e.g, Bis(triphenylphosphine)palladium(II) dichloride), a copper(I) co-catalyst (e.g. CuI), and a suitable base such as diisopropylethylamine, triethylamine, pyridine, potassium carbonate, and the like. The reaction is typically conducted at from about 30 ºC to about 120 ºC for a period of time sufficient for substantial completion of the reaction as evidenced by e.g., thin layer chromatography. Upon reaction completion, conventional workup of the reaction solution can be followed by isolation / purification processes such as crystallization, chromatography, high performance liquid chromatography (HPLC), and the like to provide for compound 14A. [0120] Other variants of compound 12A are commercially available or can be prepared by conventional reaction conditions well known in the art. [0121] Other starting materials used herein are either well known in the art, commercially available, or can be prepared by conventional synthetic methods. Methods [0122] In one embodiment, the compounds and compositions described herein are useful in methods for treating a BRD4 dependent disease or disorder or a disease or disorder that is mediated, at least in part by, BRD4. The methods comprise administering to a subject suffering from a BRD4 dependent disease or disorder an effective amount of a compound, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof or a pharmaceutical composition comprising said compound, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof as described herein. [0123] In one embodiment, there is provided a compound, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof or a pharmaceutical composition comprising said compound, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof as described herein for use in treating an BRD4 dependent disease or disorder. [0124] In one embodiment, the method relates a compound, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof or a pharmaceutical composition comprising said compound, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof as described herein for use in manufacture of a medicament for reducing BRD4 protein levels where reduction of such protein levels treats or ameliorates the diseases or disorder. [0125] In one embodiment, the method relates a compound, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof or a pharmaceutical composition comprising said compound, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof as described herein for use as described herein, wherein the BRD4 degradation at 1µM concentration of the compounds described herein is at least 50% or at least 70%. The BRD4 degradation is measured by the assay described in the biological example. [0126] The compounds and compositions described herein are useful in treating BRD4 dependent diseases or disorders such as liposarcoma, glioblastoma, bladder cancer, adrenocortical cancer, multiple myeloma, colorectal cancer, non-small cell lung cancer, Human Papilloma Virus-associated cervical, oropharyngeal, penis, anal, thyroid, or vaginal cancer or Epstein-Barr Virus-associated nasopharyngeal carcinoma, gastric cancer, rectal cancer, thyroid cancer, Hodgkin lymphoma or diffuse large B-cell lymphoma. The cancer may be selected from prostate cancer, breast carcinoma, lymphomas, leukemia, myeloma, bladder carcinoma, colon cancer, cutaneous melanoma, hepatocellular carcinoma, endometrial cancer, ovarian cancer, cervical cancer, lung cancer, renal cancer, glioblastoma multiform, glioma, thyroid cancer, parathyroid tumor, nasopharyngeal cancer, tongue cancer, pancreatic cancer, esophageal cancer, cholangiocarcinoma, gastric cancer, soft tissue sarcomas, rhabdomyosarcoma (RMS), synovial sarcoma, osteosarcoma, rhabdoid cancers, cancer for which the immune response is deficient, an immunogenic cancer, and Ewing’s sarcoma. In one embodiment, the BRD4-dependent disease or disorder is a disease or disorder is selected from non-small cell lung cancer (NSCLC), melanoma, triple-negative breast cancer (TNBC), nasopharyngeal cancer (NPC), microsatellite stable colorectal cancer (mssCRC), thymoma, carcinoid, and gastrointestinal stromal tumor (GIST). In another embodiment, the cancer is selected from non-small cell lung cancer (NSCLC), melanoma, triple-negative breast cancer (TNBC), nasopharyngeal cancer (NPC), microsatellite stable colorectal cancer (mssCRC), thymoma, carcinoid, acute myelogenous leukemia, and gastrointestinal stromal tumor (GIST). In another embodiment, the BRD4-dependent disease or disorder is a disease or disorder is selected from non-small cell lung cancer (NSCLC), melanoma, triple- negative breast cancer (TNBC), nasopharyngeal cancer (NPC), and microsatellite stable colorectal cancer (mssCRC). [0127] The compounds of the disclosure can be administered in effective amounts to treat or prevent a disorder and/or prevent the development thereof in subjects. [0128] In general, methods of using the compounds of the present application comprise administering to a subject in need thereof an effective amount of a compound as described herein. [0129] In certain embodiments, compounds as described herein are useful in the treatment of proliferative disorders (e.g., cancer, benign neoplasms, inflammatory disease, and autoimmune diseases). In certain embodiments, according to the methods of treatment of the present application, levels of cell proteins of interest, e.g., pathogenic and oncogenic proteins are modulated, or their expression is inhibited or the proteins are modulated or degraded by contacting said cells with a compound or composition, as described herein. In other embodiments, the compounds are useful in treating cancer. [0130] Thus, in another aspect of the application, methods for the treatment of cancer are provided comprising administering an effective amount of compound or composition, as described herein, to a subject in need thereof. In certain embodiments, a method for the treatment of cancer is provided comprising administering an effective amount of a compound, or a pharmaceutical composition comprising a compound as described herein to a subject in need thereof, in such amounts and for such time as is necessary to achieve the desired result. In some embodiments, the compounds of present application are administered orally. The compounds and compositions, according to the method of the present application, are administered orally to a subject using any amount and any route of administration effective for killing or inhibiting the growth of tumor cells. Thus, the expression “amount effective to kill or inhibit the growth of tumor cells,” as used herein, refers to a sufficient amount of agent to kill or inhibit the growth of tumor cells. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the disease, and the like. [0131] In certain embodiments, the method involves the administration of a therapeutically effective amount of the compound or a pharmaceutically acceptable salt thereof to a subject (including, but not limited to a human or other mammal in need of it. In certain embodiments, the compounds or compositions described herein are useful for the treatment of cancer (including, but not limited to, glioblastoma, retinoblastoma, breast cancer, cervical cancer, colon and rectal cancer, leukemia, lymphoma, lung cancer (including, but not limited to small cell lung cancer), melanoma and/or skin cancer, multiple myeloma, non-Hodgkin's lymphoma, ovarian cancer, pancreatic cancer, prostate cancer and gastric cancer, bladder cancer, uterine cancer, kidney cancer, testicular cancer, stomach cancer, brain cancer, liver cancer, or esophageal cancer). [0132] In certain embodiments, the compounds or compositions described herein are useful in the treatment of cancers and other proliferative disorders, including, but not limited to breast cancer, cervical cancer, colon and rectal cancer, leukemia, lung cancer, melanoma, multiple myeloma, non-Hodgkin's lymphoma, ovarian cancer, pancreatic cancer, prostate cancer, and gastric cancer. In certain embodiments, compounds or compositions described herein are active against solid tumors. [0133] Additionally, the present application provides pharmaceutically acceptable derivatives of the compounds, and methods of treating a subject using these compounds, pharmaceutical compositions thereof, or either of these in combination with one or more additional therapeutic agents. [0134] Another aspect of the application relates to a method of treating or lessening the severity of a disease or condition associated with a proliferation disorder in a patient, said method comprising a step of administering to said patient, a compound of Formula I or Formula I-1, or a composition comprising said compound. [0135] It will be appreciated that the compounds and compositions, according to the method of the present application, may be administered using any amount and any route of administration effective for the treatment of cancer and/or disorders associated with cell hyperproliferation. [0136] The present application provides methods for the treatment of a proliferative disorder in a subject in need thereof by administering to a subject in need of such treatment, a therapeutically effective amount of a compound of the present application, or a pharmaceutically acceptable salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof. The proliferative disorder can be cancer or a precancerous condition. The present application further provides the use of a compound of the present application, or a pharmaceutically acceptable salt, salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof, for the preparation of a medicament useful for the treatment of a proliferative disorder. [0137] The present application also provides methods of protecting against a proliferative disorder in a subject in need thereof by administering a therapeutically effective amount of compound of the present application, or a pharmaceutically acceptable salt, salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof, to a subject in need of such treatment. The proliferative disorder can be cancer or a precancerous condition. The present application also provides the use of compound of the present application, or a pharmaceutically acceptable salt, salt, solvate, stereoisomer, mixture of stereoisomers, or tautomer of each thereof, for the preparation of a medicament useful for the prevention of a proliferative disorder. [0138] As used herein, the term “proliferative disorder” refers to conditions in which unregulated or abnormal growth, or both, of cells can lead to the development of an unwanted condition or disease, which may or may not be cancerous. Exemplary proliferative disorders of the application encompass a variety of conditions wherein cell division is deregulated. Exemplary proliferative disorders include, but are not limited to, neoplasms, benign tumors, malignant tumors, uterine fibroids, pre-cancerous conditions, in situ tumors, encapsulated tumors, metastatic tumors, liquid tumors, solid tumors, immunological tumors, hematological tumors, cancers, carcinomas, leukemias, lymphomas, sarcomas, and rapidly dividing cells. The term “rapidly dividing cell” as used herein is defined as any cell that divides at a rate that exceeds or is greater than what is expected or observed among neighboring or juxtaposed cells within the same tissue. A proliferative disorder includes a precancer or a precancerous condition. A proliferative disorder includes cancer. In some embodiments, the methods provided herein are used to treat or alleviate a symptom of cancer. The term “cancer” includes solid tumors, as well as, hematologic tumors and/or malignancies. A “precancer cell” or “precancerous cell” is a cell manifesting a proliferative disorder that is a precancer or a precancerous condition. A “cancer cell” or “cancerous cell” is a cell manifesting a proliferative disorder that is a cancer. Any reproducible means of measurement may be used to identify cancer cells or precancerous cells. Cancer cells or precancerous cells can be identified by histological typing or grading of a tissue sample (e.g., a biopsy sample). Cancer cells or precancerous cells can be identified through the use of appropriate molecular markers. [0139] Exemplary non-cancerous conditions or disorders include, but are not limited to, rheumatoid arthritis; inflammation; autoimmune disease; lymphoproliferative conditions; acromegaly; rheumatoid spondylitis; osteoarthritis; gout, other arthritic conditions; sepsis; septic shock; endotoxic shock; gram- negative sepsis; toxic shock syndrome; asthma; adult respiratory distress syndrome; chronic obstructive pulmonary disease; chronic pulmonary inflammation; inflammatory bowel disease; Crohn's disease; psoriasis; eczema; ulcerative colitis; pancreatic fibrosis; hepatic fibrosis; acute and chronic renal disease; irritable bowel syndrome; pyresis; restenosis; cerebral malaria; stroke and ischemic injury; neural trauma; Alzheimer's disease; Huntington's disease; Parkinson's disease; acute and chronic pain; allergic rhinitis; allergic conjunctivitis; chronic heart failure; acute coronary syndrome; cachexia; malaria; leprosy; leishmaniasis; Lyme disease; Reiter's syndrome; acute synovitis; muscle degeneration, bursitis; tendonitis; tenosynovitis; herniated, ruptures, or prolapsed intervertebral disk syndrome; osteopetrosis; thrombosis; restenosis; silicosis; pulmonary sarcoidosis; bone resorption diseases, such as osteoporosis; graft-versus-host reaction; Multiple Sclerosis; lupus; fibromyalgia; AIDS and other viral diseases such as Herpes Zoster, Herpes Simplex I or II, influenza virus and cytomegalovirus; and diabetes mellitus. [0140] Exemplary cancers include, but are not limited to, adrenocortical carcinoma, AIDS-related cancers, AIDS-related lymphoma, anal cancer, anorectal cancer, cancer of the anal canal, appendix cancer, childhood cerebellar astrocytoma, childhood cerebral astrocytoma, basal cell carcinoma, skin cancer (non-melanoma), biliary cancer, extrahepatic bile duct cancer, intrahepatic bile duct cancer, bladder cancer, urinary bladder cancer, bone and joint cancer, osteosarcoma and malignant fibrous histiocytoma, brain cancer, brain tumor, brain stem glioma, cerebellar astrocytoma, cerebral astrocytoma/malignant glioma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumors, visual pathway and hypothalamic glioma, breast cancer, bronchial adenomas/carcinoids, carcinoid tumor, gastrointestinal, nervous system cancer, nervous system lymphoma, central nervous system cancer, central nervous system lymphoma, cervical cancer, childhood cancers, chronic lymphocytic leukemia, chronic myelogenous leukemia, chronic myeloproliferative disorders, colon cancer, colorectal cancer, cutaneous T-cell lymphoma, lymphoid neoplasm, mycosis fungoides, Sezary Syndrome, endometrial cancer, esophageal cancer, extracranial germ cell tumor, extragonadal germ cell tumor, extrahepatic bile duct cancer, eye cancer, intraocular melanoma, retinoblastoma, gallbladder cancer, gastric (stomach) cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (GIST), germ cell tumor, ovarian germ cell tumor, gestational trophoblastic tumor glioma, head and neck cancer, hepatocellular (liver) cancer, Hodgkin lymphoma, hypopharyngeal cancer, intraocular melanoma, ocular cancer, islet cell tumors (endocrine pancreas), Kaposi Sarcoma, kidney cancer, renal cancer, kidney cancer, laryngeal cancer, acute lymphoblastic leukemia, acute myeloid leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, hairy cell leukemia, lip and oral cavity cancer, liver cancer, lung cancer, non-small cell lung cancer, small cell lung cancer, AIDS-related lymphoma, non-Hodgkin lymphoma, primary central nervous system lymphoma, Waldenstrom macroglobulinemia, medulloblastoma, melanoma, intraocular (eye) melanoma, merkel cell carcinoma, mesothelioma malignant, mesothelioma, metastatic squamous neck cancer, mouth cancer, cancer of the tongue, multiple endocrine neoplasia syndrome, mycosis fungoides, myelodysplastic syndromes, myelodysplastic/myeloproliferative diseases, chronic myelogenous leukemia, acute myeloid leukemia, multiple myeloma, chronic myeloproliferative disorders, nasopharyngeal cancer, neuroblastoma, oral cancer, oral cavity cancer, oropharyngeal cancer, ovarian cancer, ovarian epithelial cancer, ovarian low malignant potential tumor, pancreatic cancer, islet cell pancreatic cancer, paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pheochromocytoma, pineoblastoma and supratentorial primitive neuroectodermal tumors, pituitary tumor, plasma cell neoplasm/multiple myeloma, pleuropulmonary blastoma, prostate cancer, rectal cancer, renal pelvis and ureter, transitional cell cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, Ewing family of sarcoma tumors, Kaposi Sarcoma, soft tissue sarcoma, uterine cancer, uterine sarcoma, skin cancer (non-melanoma), skin cancer (melanoma), merkel cell skin carcinoma, small intestine cancer, soft tissue sarcoma, squamous cell carcinoma, stomach (gastric) cancer, supratentorial primitive neuroectodermal tumors, testicular cancer, throat cancer, thymoma, thymoma and thymic carcinoma, thyroid cancer, transitional cell cancer of the renal pelvis and ureter and other urinary organs, gestational trophoblastic tumor, urethral cancer, endometrial uterine cancer, uterine sarcoma, uterine corpus cancer, vaginal cancer, vulvar cancer, and Wilms’ Tumor. [0141] A “proliferative disorder of the hematologic system” is a proliferative disorder involving cells of the hematologic system. A proliferative disorder of the hematologic system can include lymphoma, leukemia, myeloid neoplasms, mast cell neoplasms, myelodysplasia, benign monoclonal gammopathy, lymphomatoid granulomatosis, lymphomatoid papulosis, polycythemia vera, chronic myelocytic leukemia, agnogenic myeloid metaplasia, and essential thrombocythemia. A proliferative disorder of the hematologic system can include hyperplasia, dysplasia, and metaplasia of cells of the hematologic system. In some embodiments, the compositions of the present application may be used to treat a cancer selected from the group consisting of a hematologic cancer of the present application or a hematologic proliferative disorder of the present application. A hematologic cancer of the present application can include multiple myeloma, lymphoma (including Hodgkin's lymphoma, non-Hodgkin's lymphoma, childhood lymphomas, and lymphomas of lymphocytic and cutaneous origin), leukemia (including childhood leukemia, hairy-cell leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, chronic myelogenous leukemia, and mast cell leukemia), myeloid neoplasms and mast cell neoplasms. [0142] A “proliferative disorder of the lung” is a proliferative disorder involving cells of the lung. Proliferative disorders of the lung can include all forms of proliferative disorders affecting lung cells. Proliferative disorders of the lung can include lung cancer, a precancer or precancerous condition of the lung, benign growths or lesions of the lung, and malignant growths or lesions of the lung, and metastatic lesions in tissue and organs in the body other than the lung. In some embodiments, the compositions of the present application may be used to treat lung cancer or proliferative disorders of the lung. Lung cancer can include all forms of cancer of the lung. Lung cancer can include malignant lung neoplasms, carcinoma in situ, typical carcinoid tumors, and atypical carcinoid tumors. Lung cancer can include small cell lung cancer (“SCLC”), non-small cell lung cancer (“NSCLC”), squamous cell carcinoma, adenocarcinoma, small cell carcinoma, large cell carcinoma, adenosquamous cell carcinoma, and mesothelioma. Lung cancer can include “scar carcinoma”, bronchioalveolar carcinoma, giant cell carcinoma, spindle cell carcinoma, and large cell neuroendocrine carcinoma. Lung cancer can include lung neoplasms having histologic and ultrastructual heterogeneity (e.g., mixed cell types). [0143] Proliferative disorders of the lung can include all forms of proliferative disorders affecting lung cells. Proliferative disorders of the lung can include lung cancer, precancerous conditions of the lung. Proliferative disorders of the lung can include hyperplasia, metaplasia, and dysplasia of the lung. Proliferative disorders of the lung can include asbestos-induced hyperplasia, squamous metaplasia, and benign reactive mesothelial metaplasia. Proliferative disorders of the lung can include replacement of columnar epithelium with stratified squamous epithelium, and mucosal dysplasia. Individuals exposed to inhaled injurious environmental agents such as cigarette smoke and asbestos may be at increased risk for developing proliferative disorders of the lung. Prior lung diseases that may predispose individuals to development of proliferative disorders of the lung can include chronic interstitial lung disease, necrotizing pulmonary disease, scleroderma, rheumatoid disease, sarcoidosis, interstitial pneumonitis, tuberculosis, repeated pneumonias, idiopathic pulmonary fibrosis, granulomata, asbestosis, fibrosing alveolitis, and Hodgkin's disease. [0144] A “proliferative disorder of the colon” is a proliferative disorder involving cells of the colon. In one embodiment, the proliferative disorder of the colon is colon cancer. In one embodiment, compositions of the present application may be used to treat colon cancer or proliferative disorders of the colon. Colon cancer can include all forms of cancer of the colon. Colon cancer can include sporadic and hereditary colon cancers. Colon cancer can include malignant colon neoplasms, carcinoma in situ, typical carcinoid tumors, and atypical carcinoid tumors. Colon cancer can include adenocarcinoma, squamous cell carcinoma, and adenosquamous cell carcinoma. Colon cancer can be associated with a hereditary syndrome selected from the group consisting of hereditary nonpolyposis colorectal cancer, familial adenomatous polyposis, Gardner's syndrome, Peutz-Jeghers syndrome, Turcot's syndrome and juvenile polyposis. Colon cancer can be caused by a hereditary syndrome selected from the group consisting of hereditary nonpolyposis colorectal cancer, familial adenomatous polyposis, Gardner's syndrome, Peutz- Jeghers syndrome, Turcot's syndrome and juvenile polyposis. [0145] Proliferative disorders of the colon can include all forms of proliferative disorders affecting colon cells. Proliferative disorders of the colon can include colon cancer, precancerous conditions of the colon, adenomatous polyps of the colon and metachronous lesions of the colon. A proliferative disorder of the colon can include adenoma. Proliferative disorders of the colon can be characterized by hyperplasia, metaplasia, and dysplasia of the colon. Prior colon diseases that may predispose individuals to development of proliferative disorders of the colon can include prior colon cancer. Current disease that may predispose individuals to development of proliferative disorders of the colon can include Crohn's disease and ulcerative colitis. A proliferative disorder of the colon can be associated with a mutation in a gene selected from the group consisting of p53, ras, FAP and DCC. An individual can have an elevated risk of developing a proliferative disorder of the colon due to the presence of a mutation in a gene selected from the group consisting of p53, ras, FAP and DCC. [0146] A “proliferative disorder of the pancreas” is a proliferative disorder involving cells of the pancreas. Proliferative disorders of the pancreas can include all forms of proliferative disorders affecting pancreatic cells. Proliferative disorders of the pancreas can include pancreas cancer, a precancer or precancerous condition of the pancreas, hyperplasia of the pancreas, and dysplasia of the pancreas, benign growths or lesions of the pancreas, and malignant growths or lesions of the pancreas, and metastatic lesions in tissue and organs in the body other than the pancreas. Pancreatic cancer includes all forms of cancer of the pancreas. Pancreatic cancer can include ductal adenocarcinoma, adenosquamous carcinoma, pleomorphic giant cell carcinoma, mucinous adenocarcinoma, osteoclast-like giant cell carcinoma, mucinous cystadenocarcinoma, acinar carcinoma, unclassified large cell carcinoma, small cell carcinoma, pancreatoblastoma, papillary neoplasm, mucinous cystadenoma, papillary cystic neoplasm, and serous cystadenoma. Pancreatic cancer can also include pancreatic neoplasms having histologic and ultrastructual heterogeneity (e.g., mixed cell types). [0147] A “proliferative disorder of the prostate” is a proliferative disorder involving cells of the prostate. Proliferative disorders of the prostate can include all forms of proliferative disorders affecting prostate cells. Proliferative disorders of the prostate can include prostate cancer, a precancer or precancerous condition of the prostate, benign growths or lesions of the prostate, and malignant growths or lesions of the prostate, and metastatic lesions in tissue and organs in the body other than the prostate. Proliferative disorders of the prostate can include hyperplasia, metaplasia, and dysplasia of the prostate. [0148] A “proliferative disorder of the skin” is a proliferative disorder involving cells of the skin. Proliferative disorders of the skin can include all forms of proliferative disorders affecting skin cells. Proliferative disorders of the skin can include a precancer or precancerous condition of the skin, benign growths or lesions of the skin, melanoma, malignant melanoma and other malignant growths or lesions of the skin, and metastatic lesions in tissue and organs in the body other than the skin. Proliferative disorders of the skin can include hyperplasia, metaplasia, and dysplasia of the skin. [0149] A “proliferative disorder of the ovary” is a proliferative disorder involving cells of the ovary. Proliferative disorders of the ovary can include all forms of proliferative disorders affecting cells of the ovary. Proliferative disorders of the ovary can include a precancer or precancerous condition of the ovary, benign growths or lesions of the ovary, ovarian cancer, malignant growths or lesions of the ovary, and metastatic lesions in tissue and organs in the body other than the ovary. Proliferative disorders of the skin can include hyperplasia, metaplasia, and dysplasia of cells of the ovary. [0150] A “proliferative disorder of the breast” is a proliferative disorder involving cells of the breast. Proliferative disorders of the breast can include all forms of proliferative disorders affecting breast cells. Proliferative disorders of the breast can include breast cancer, a precancer or precancerous condition of the breast, benign growths or lesions of the breast, and malignant growths or lesions of the breast, and metastatic lesions in tissue and organs in the body other than the breast. Proliferative disorders of the breast can include hyperplasia, metaplasia, and dysplasia of the breast. [0151] A cancer that is to be treated can be staged according to the American Joint Committee on Cancer (AJCC) TNM classification system, where the tumor (T) has been assigned a stage of TX, T1, T1mic, T1a, T1b, T1c, T2, T3, T4, T4a, T4b, T4c, or T4d; and where the regional lymph nodes (N) have been assigned a stage of NX, N0, N1, N2, N2a, N2b, N3, N3a, N3b, or N3c; and where distant metastasis (M) can be assigned a stage of MX, M0, or M1. A cancer that is to be treated can be staged according to an American Joint Committee on Cancer (AJCC) classification as Stage I, Stage IIA, Stage IIB, Stage IIIA, Stage IIIB, Stage IIIC, or Stage IV. A cancer that is to be treated can be assigned a grade according to an AJCC classification as Grade GX (e.g., grade cannot be assessed), Grade 1, Grade 2, Grade 3 or Grade 4. A cancer that is to be treated can be staged according to an AJCC pathologic classification (pN) of pNX, pN0, PN0 (I-), PN0 (I+), PN0 (mol-), PN0 (mol+), PN1, PN1(mi), PN1a, PN1b, PN1c, pN2, pN2a, pN2b, pN3, pN3a, pN3b, or pN3c. [0152] A cancer that is to be treated can include a tumor that has been determined to be less than or equal to about 2 centimeters in diameter. A cancer that is to be treated can include a tumor that has been determined to be from about 2 to about 5 centimeters in diameter. A cancer that is to be treated can include a tumor that has been determined to be greater than or equal to about 3 centimeters in diameter. A cancer that is to be treated can include a tumor that has been determined to be greater than 5 centimeters in diameter. A cancer that is to be treated can be classified by microscopic appearance as well differentiated, moderately differentiated, poorly differentiated, or undifferentiated. A cancer that is to be treated can be classified by microscopic appearance with respect to mitosis count (e.g., amount of cell division) or nuclear pleiomorphism (e.g., change in cells). A cancer that is to be treated can be classified by microscopic appearance as being associated with areas of necrosis (e.g., areas of dying or degenerating cells). A cancer that is to be treated can be classified as having an abnormal karyotype, having an abnormal number of chromosomes, or having one or more chromosomes that are abnormal in appearance. A cancer that is to be treated can be classified as being aneuploid, triploid, tetraploid, or as having an altered ploidy. A cancer that is to be treated can be classified as having a chromosomal translocation, or a deletion or duplication of an entire chromosome, or a region of deletion, duplication or amplification of a portion of a chromosome. [0153] A cancer that is to be treated can be evaluated by DNA cytometry, flow cytometry, or image cytometry. A cancer that is to be treated can be typed as having 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of cells in the synthesis stage of cell division (e.g., in S phase of cell division). A cancer that is to be treated can be typed as having a low S-phase fraction or a high S-phase fraction. [0154] As used herein, a “normal cell” is a cell that cannot be classified as part of a “proliferative disorder”. A normal cell lacks unregulated or abnormal growth, or both, that can lead to the development of an unwanted condition or disease. In one embodiment, a normal cell possesses normally functioning cell cycle checkpoint control mechanisms. [0155] One skilled in the art may refer to general reference texts for detailed descriptions of known techniques discussed herein or equivalent techniques. These texts include Ausubel et al., Current Protocols in Molecular Biology, John Wiley and Sons, Inc. (2005); Sambrook et al., Molecular Cloning, A Laboratory Manual (3rd edition), Cold Spring Harbor Press, Cold Spring Harbor, N.Y. (2000); Coligan et al., Current Protocols in Immunology, John Wiley & Sons, N.Y.; Erma et al., Current Protocols in Pharmacology, John Wiley & Sons, N.Y.; Fingl et al., The Pharmacological Basis of Therapeutics (1975), Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 18th edition (1990). These texts can, of course, also be referred to in making or using an aspect of the application. [0156] In certain embodiments, compounds of the application are useful in the treatment of proliferative disorders (e.g., cancer, benign neoplasms, inflammatory disease, and autoimmune diseases). In certain embodiments, according to the methods of treatment of the present application, levels of cell proteins of interest, e.g., pathogenic and oncogenic proteins are modulated, or their growth is inhibited by contacting said cells with a compound or composition, as described herein. In other embodiments, the compounds are useful in treating cancer. [0157] In certain embodiments, the method involves the administration of a therapeutically effective amount of the compound or a pharmaceutically acceptable derivative thereof to a subject (including, but not limited to a human or animal) in need of it. In certain embodiments, the compounds are useful for the treatment of cancer (including, but not limited to, glioblastoma, retinoblastoma, breast cancer, cervical cancer, colon and rectal cancer, leukemia, lymphoma, lung cancer (including, but not limited to small cell lung cancer), melanoma and/or skin cancer, multiple myeloma, non-Hodgkin's lymphoma, ovarian cancer, pancreatic cancer, prostate cancer and gastric cancer, bladder cancer, uterine cancer, kidney cancer, testicular cancer, stomach cancer, brain cancer, liver cancer, or esophageal cancer). [0158] In certain embodiments, the anticancer agents are useful in the treatment of cancers and other proliferative disorders, including, but not limited to breast cancer, cervical cancer, colon and rectal cancer, leukemia, lung cancer, melanoma, multiple myeloma, non-Hodgkin's lymphoma, ovarian cancer, pancreatic cancer, prostate cancer, and gastric cancer. In certain embodiments, the anticancer agents are active against solid tumors. [0159] Additionally, the present application provides pharmaceutically acceptable derivatives of the compounds, and methods of treating a subject using these compounds, pharmaceutical compositions thereof, or either of these in combination with one or more additional therapeutic agents. [0160] For example, other therapies or anticancer agents that may be used in combination with the compounds disclosed herein including surgery, radiotherapy, endocrine therapy, biologic response modifiers (interferons, interleukins, and tumor necrosis factor (TNF), to name a few), hyperthermia and cryotherapy, agents to attenuate any adverse effects (e.g., antiemetics), and other approved chemotherapeutic drugs, including, but not limited to, alkylating drugs (mechlorethamine, chlorambucil, Cyclophosphamide, Melphalan, Ifosfamide), antimetabolites (Methotrexate), purine antagonists and pyrimidine antagonists (6-Mercaptopurine, 5-Fluorouracil, Cytarabile, Gemcitabine), spindle poisons (Vinblastine, Vincristine, Vinorelbine, Paclitaxel), podophyllotoxins (Etoposide, Irinotecan, Topotecan), antibiotics (Doxorubicin, Bleomycin, Mitomycin), nitrosoureas (Carmustine, Lomustine), inorganic ions (Cisplatin, Carboplatin), enzymes (Asparaginase), and hormones (Tamoxifen, Leuprolide, Flutamide, and Megestrol), to name a few. For a more comprehensive discussion of overview of cancer therapy see The Merck Manual, Twentieth Ed.2020, the entire contents of which are hereby incorporated by reference. See also the National Cancer Institute (NCI) website (www.nci.nih.gov) and the Food and Drug Administration (FDA) website for a list of the FDA approved oncology drugs (www.fda.gov/cder/cancer/druglistframe). [0161] In certain embodiments, the pharmaceutical compositions comprising the compounds disclosed herein further comprise one or more additional therapeutically active ingredients (e.g., chemotherapeutic and/or palliative). For purposes of the application, the term “palliative” refers to treatment that is focused on the relief of symptoms of a disease and/or side effects of a therapeutic regimen, but is not curative. For example, palliative treatment encompasses painkillers, antinausea medications and anti-sickness drugs. In addition, chemotherapy, radiotherapy and surgery can all be used palliatively (that is, to reduce symptoms without going for cure; e.g., for shrinking tumors and reducing pressure, bleeding, pain and other symptoms of cancer). Administration, Pharmaceutical Compositions [0162] Administration of the disclosed compounds and pharmaceutical compositions can be accomplished via any mode of administration for therapeutic agents. These modes include systemic or local administration such as oral, nasal, parenteral, transdermal, subcutaneous, vaginal, buccal, rectal or topical administration modes. [0163] Depending on the intended mode of administration, the disclosed compositions can be in solid, semi-solid or liquid dosage form, such as, for example, injectables, tablets, suppositories, pills, time- release capsules, elixirs, tinctures, emulsions, syrups, powders, liquids, suspensions, or the like, sometimes in unit dosages and consistent with conventional pharmaceutical practices. Likewise, they can also be administered in intravenous (both bolus and infusion), intraperitoneal, subcutaneous or intramuscular form, and all using forms well known to those skilled in the pharmaceutical arts. [0164] Illustrative pharmaceutical compositions are tablets and gelatin capsules comprising a compound of the disclosure and a pharmaceutically acceptable carrier, such as a) a diluent, e.g., purified water, triglyceride oils, such as hydrogenated or partially hydrogenated vegetable oil, or mixtures thereof, com oil, olive oil, sunflower oil, safflower oil, fish oils, such as EPA or DHA, or their esters or triglycerides or mixtures thereof, omega-3 fatty acids or derivatives thereof, lactose, dextrose, sucrose, mannitol, sorbitol, cellulose, sodium, saccharin, glucose and/or glycine; b) a lubricant, e.g., silica, talcum, stearic acid, its magnesium or calcium salt, sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and/or polyethylene glycol; for tablets also; c) a binder, e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, magnesium carbonate, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, waxes, and/or polyvinylpyrrolidone, if desired; d) a disintegrant, e.g., starches, agar, methyl cellulose, bentonite, xanthan gum, algic acid or its sodium salt, or effervescent mixtures; e) absorbent, colorant, flavorant and sweetener; f) an emulsifier or dispersing agent, such as Tween 80, Labrasol, HPMC, DOSS, caproyl 909, labrafac, labrafil, peceol, transcutol, capmul MCM, capmul PG-12, captex 355, gelucire, vitamin E TGPS or other acceptable emulsifier; and/or g) an agent that enhances absorption of the compound such as cyclodextrin, hydroxypropyl-cyclodextrin, PEG400, PEG200. [0165] Liquid, particularly injectable, compositions can, for example, be prepared by dissolution, dispersion, etc. For example, the disclosed compound is dissolved in or mixed with a pharmaceutically acceptable solvent such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form an injectable isotonic solution or suspension. Proteins such as albumin, chylomicron particles, or serum proteins can be used to solubilize the disclosed compounds. [0166] The disclosed compounds can be also formulated as a suppository that can be prepared from fatty emulsions or suspensions; using polyalkylene glycols such as propylene glycol, as the carrier. [0167] The disclosed compounds can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, containing cholesterol, stearylamine or phosphatidylcholines. [0168] In some embodiments, a film of lipid components is hydrated with an aqueous solution of drug to a form lipid layer encapsulating the drug, as described in U.S. Pat. No.5,262,564, which is hereby incorporated by reference in its entirety. [0169] Disclosed compounds can also be delivered by the use of monoclonal antibodies as individual carriers to which the disclosed compounds are coupled. The disclosed compounds can also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspanamidephenol, or polyethyleneoxidepolylysine substituted with palmitoyl residues. Furthermore, the disclosed compounds can be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and cross-linked or amphipathic block copolymers of hydrogels. In one embodiment, disclosed compounds are not covalently bound to a polymer, e.g., a polycarboxylic acid polymer, or a polyacrylate. [0170] Parental injectable administration is generally used for subcutaneous, intramuscular or intravenous injections and infusions. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions or solid forms suitable for dissolving in liquid prior to injection. [0171] Another aspect of the disclosure is directed to pharmaceutical compositions comprising a compound of Formula I, I-1, IA, or IA-1 and a pharmaceutically acceptable carrier. Another aspect of the disclosure is directed to pharmaceutical compositions comprising a compound of Formula I, I-1, IA, or IA-1, or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof and a pharmaceutically acceptable carrier. The pharmaceutically acceptable carrier may further include an excipient, diluent, or surfactant. [0172] Compositions can be prepared according to conventional mixing, granulating or coating methods, respectively, and the present pharmaceutical compositions can contain from about 0.1% to about 99%, from about 5% to about 90%, or from about 1% to about 20% of the disclosed compound by weight or volume. [0173] In one embodiment, the disclosure provides a kit comprising two or more separate pharmaceutical compositions, at least one of which contains a compound of the present disclosure. In one embodiment, the kit comprises means for separately retaining said compositions, such as a container, divided bottle, or divided foil packet. An example of such a kit is a blister pack, as typically used for the packaging of tablets, capsules and the like. [0174] The kit of the disclosure may be used for administering different dosage forms, for example, oral and parenteral, for administering the separate compositions at different dosage intervals, or for titrating the separate compositions against one another. To assist compliance, the kit of the disclosure typically comprises directions for administration. [0175] Pharmaceutical dosage forms of a compound of this disclosure may be manufactured by any of the methods well-known in the art, such as, for example, by conventional mixing, sieving, dissolving, melting, granulating, dragee-making, tableting, suspending, extruding, spray-drying, levigating, emulsifying, (nano-/micro-) encapsulating, entrapping, or lyophilization processes. As noted above, the compositions of this disclosure can include one or more physiologically acceptable inactive ingredients that facilitate processing of active molecules into preparations for pharmaceutical use. [0176] As noted above, the compositions are comprised of, in general, a compound of this disclosure in combination with at least one pharmaceutically acceptable excipient. Acceptable excipients are non- toxic, aid administration, and do not adversely affect the therapeutic benefit of the claimed compounds. Such excipient may be any solid, liquid, semi-solid or, in the case of an aerosol composition, gaseous excipient that is generally available to one of skill in the art. [0177] Solid pharmaceutical excipients include starch, cellulose, talc, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, magnesium stearate, sodium stearate, glycerol monostearate, sodium chloride, dried skim milk and the like. Liquid and semi-solid excipients may be selected from glycerol, propylene glycol, water, ethanol and various oils, including those of petroleum, animal, vegetable or synthetic origin, e.g., peanut oil, soybean oil, mineral oil, sesame oil, etc. In some embodiments, liquid carriers, particularly for injectable solutions, include water, saline, aqueous dextrose, and glycols. [0178] Compressed gases may be used to disperse a compound of this disclosure in an aerosol form. Inert gases suitable for this purpose are nitrogen, carbon dioxide, etc. Other suitable pharmaceutical excipients and their formulations are described in Remington’s Pharmaceutical Sciences, edited by E. W. Martin (Mack Publishing Company, 18th ed., 1990). [0179] The compositions of this disclosure may, if desired, be presented in a pack or dispenser device containing one or more unit dosage forms containing the active ingredient. Such a pack or device may, for example, comprise metal or plastic foil, such as a blister pack, or glass, and rubber stoppers such as in vials. The pack or dispenser device may be accompanied by instructions for administration. Compositions comprising a compound of this disclosure that can be formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition. [0180] The amount of the compound in a formulation can vary within the full range employed by those skilled in the art. Typically, the formulation will contain, on a weight percent (wt %) basis, from about 0.01-99.99 wt % of a compound of this disclosure based on the total formulation, with the balance being one or more suitable pharmaceutical excipients. In one embodiment, the compound is present at a level of about 1-80 wt %. Representative pharmaceutical formulations are described below. Formulation Examples [0181] The following are representative pharmaceutical formulations containing a compound of this disclosure. Formulation Example 1 -- Tablet formulation [0182] The following ingredients are mixed intimately and pressed into single scored tablets. Quantity per
Figure imgf000070_0001
Formulation Example 2 -- Capsule formulation [0183] The following ingredients are mixed intimately and loaded into a hard-shell gelatin capsule Quantity per
Figure imgf000070_0002
Formulation Example 3 -- Suspension formulation [0184] The following ingredients are mixed to form a suspension for oral administration.
Figure imgf000071_0001
Formulation Example 4 -- Injectable formulation [0185] The following ingredients are mixed to form an injectable formulation. di
Figure imgf000071_0002
Formulation Example 5 -- Suppository Formulation [0186] A suppository of total weight 2.5 g is prepared by mixing the compound of this disclosure with Witepsol® H-15 (triglycerides of saturated vegetable fatty acid; Riches-Nelson, Inc., New York), and has the following composition: Ingredient Amount
Figure imgf000071_0003
Dosing [0187] The dosage regimen utilizing the disclosed compound is selected in accordance with a variety of factors including type, species, age, weight, sex, and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal or hepatic function of the patient; and the particular disclosed compound employed. A physician or veterinarian of ordinary skill in the art can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition. [0188] Effective dosage amounts of the disclosed compounds, when used for the indicated effects, range from about 0.5 mg to about 5000 mg of the disclosed compound as needed to treat the condition. Compositions for in vivo or in vitro use can contain about 0.5, 5, 20, 50, 75, 100, 150, 250, 500, 750, 1000, 1250, 2500, 3500, or 5000 mg of the disclosed compound, or, in a range of from one amount to another amount in the list of doses. In one embodiment, the compositions are in the form of a tablet that can be scored. EXAMPLES [0189] The following synthetic and biological examples are offered to illustrate this disclosure and are not to be construed in any way as limiting the scope of this disclosure. Unless otherwise stated, all temperatures are in degrees Celsius. [0190] This disclosure is further understood by reference to the following examples, which are intended to be purely exemplary of this disclosure. This disclosure is not limited in scope by the exemplified embodiments, which are intended as illustrations of single aspects of this disclosure only. Any methods that are functionally equivalent are within the scope of this disclosure. Various modifications of this disclosure in addition to those described herein will become apparent to those skilled in the art from the foregoing description and accompanying figures. Such modifications fall within the scope of the appended claims. [0191] In the specification and in the examples below, all temperatures are in degrees Celsius. In addition, the following abbreviations have the following meanings. If not defined, these abbreviations have their art recognized meaning. Abbreviation Meaning δ chemical shift (ppm) ACN or MeCN acetonitrile Boc tert -butoxycarbonyl BPD bis(pinacolato)diboron BRET bioluminescence resonance energy transfer Cbz benzyloxycarbonyl DAPI 4’,6-diamidino-2-phenylindole DC50 concentration that resulted in a 50% targeted protein degradation DCC N,N-dicyclohexylcarbodiimide DCM dichloromethane DMA dimethylacetamide DMF N,N-dimethylformamide DMP 2,6-dimethylpyridine DMSO dimethylsulfoxide CDCl3 deuterated chloroform d6-DMSO deuterated dimethylsulfoxide d4-MeOD deuterated methanol eq. equivalent(s) ESI electrospray ionization EtOAc ethyl acetate EtOH ethanol g grams 1H NMR proton nuclear magnetic resonance spectroscopy h hour(s) HPLC high performance liquid chromatography L liter LC liquid chromatography LC-MS liquid chromatography – mass spectrometry M molar MeOH methanol mg milligram mL milliliter mmol millimole μL microliter umol or μmol micromole μM micromolar m/z mass-to-charge ratio min minute(s) N normal nm nanometer PBS phosphate-buffered saline Pd(Cy*Phine)2Cl2 bis[dicyclohexyl(2;,4;,6;-triisopropyl [1,1':3',1"-terphenyl]-2- yl)phosphane]palladium(II) dichloride Pd(dppf)Cl2 [1,1’-bis(diphenylphosphino)ferrocene]dichloropalladium(II) pM picomolar Pyr pyridine q.s. amount which is sufficient rt room temperature Select-Fluor 1-chloromethyl-4-fluoro-1,4-diazoniabicyclo[2.2.2]octane bis(tetrafluoroborate) t-Bu tert-butyl THF tetrahydrofuran TMSOTf trimethylsilyl trifluoromethanesulfonate UV ultraviolet v/v volume/volume ratio XPhos-Pd-G2 chloro(2-dicyclohexylphosphino-2’,4’,6’-triisopropyl-1,1’- biphenyl)[2-(2’-amino-1,1’-biphenyl)]palladium(II) NMR abbreviations br = broad d = doublet dd = doublet of doublets ddt = doublet of doublet of triplets dtd = doublet of triplet of doublets m = multiplet qd = quartet of doublets quin = quintet s = singlet t = triplet LC-MS Methods (General Method) [0192] Experiments were performed using a Phenomenex Gemini-NX C1875×30mm×3µm LC Column, at a flow rate of 25 mL/min, and a mass spectrometer using ESI as ionization source. The solvent A was 0.075% v/v NH4HCO3 in water, and solvent B was acetonitrile. The gradient consisted of 10-100% solvent B over 20 minutes, LC column temperature was 40 °C. UV absorbance was collected at 220 nm and 254 nm. Starting Materials Compound A: 1-[(2S)-2-methyl-1,2,3,4-tetrahydroquinoxalin-1-yl]ethan-1-one [0193] Compound A, CAS # 1563614-79-
Figure imgf000075_0001
8 s co e ca y available from ASW MedChem, New Brunswick, New Jersey, USA. Example 1 Preparation of (R)-1-(4-((4-(3-aminoprop-1-yn-1-yl)phenyl)amino)-3,4-dihydroquinolin-1(2H)- yl)ethan-1-one (Compound P-2): S
Figure imgf000076_0001
[0194] To a solution of 2,3-dihydro-1H-quinolin-4-one (2.0 g, 13.6 mmol, 1.0 eq) in DCM (20 mL) was added pyridine (2.1 g, 27.2 mmol, 2.2 mL, 2.0 eq) at 25 °C. Acetyl chloride (1.2 g, 14.9 mmol, 1.1 mL, 1.1 eq) was added dropwise at 0 °C. The resulting mixture was stirred at 25 °C for 20 hr. The solvents were evaporated in vacuo, and the residue diluted with water (20 mL), then extracted with ethyl acetate (40 mL). The organic phase was separated, washed with brine (20 mL), dried over Na2SO4, filtered and the solvents were evaporated in vacuo to give 1-acetyl-2,3-dihydroquinolin-4(1H)-one, which was used in the next step without further purification. m/z (ESI+) 231.1 (M+42)+. 1H NMR (400 MHz, d4-DMSO) δ 7.86 (dd, J = 1.4, 8.0 Hz, 1 H), 7.70 (d, J = 8.0 Hz, 1H), 7.60 (dt, J = 1.6, 7.2 Hz, 1H), 7.29 (t, J = 8.0 Hz, 1H), 4.12 (t, J = 6.0 Hz, 2H), 2.78 (t, J = 6.4 Hz, 2H), 2.29 (s, 3H). Step 2: [0195] To a solution
Figure imgf000076_0002
o -acety - , - y roqu no n- -one . g, . mmo , .0 eq) and (R)-2- methylpropane-2-sulfinamide (2.2 g, 18.3 mmol, 3.0 eq) in THF (15 mL), was added titanium(IV)tetraisopropoxide (10.4 g, 36.7 mmol, 10.8 mL, 6.0 eq) dropwise at 0 °C. The reaction was degassed, purged with N2, and stirred at 80 °C for 20 hrs. The mixture was cooled to -78 °C and NaBH4 (1.4 g, 36.7 mmol, 6.0 eq) was added and stirred at -78 °C for 2 hrs. The reaction mixture was quenched by slow addition 0.5 N hydrochloric acid (20 mL) and diluted with H2O (50 mL), then extracted with ethyl acetate (50 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and the solvents were evaporated in vacuo. The residue was purified by column chromatography (SiO2, 0 to 50% ethyl acetate in petroleum ether) to give (R)-N-((R)-1-acetyl-1,2,3,4-tetrahydroquinolin- 4-yl)-2-methylpropane-2-sulfinamide. m/z (ESI+) 295.1 (M+42)+. 1H NMR (400 MHz, d6-DMSO) δ 7.58 (d, J = 7.2 Hz, 1H), 7.54 - 7.34 (m, 1H), 7.24 (t, J = 7.2 Hz, 1H), 7.19 - 7.11 (m, 1H), 5.78 (d, J = 7.2 Hz, 1H), 4.41 - 4.32 (m, 1H), 3.85 - 3.63 (m, 2H), 2.20 - 2.13 (m, 3H), 2.01 - 1.83 (m, 2H), 1.19 - 1.11 (m, 9H). Step 3:
Figure imgf000077_0001
[0196] To a solution of (R)-N-((R)-1-acetyl-1,2,3,4-tetrahydroquinolin-4-yl)-2-methylpropane-2- sulfinamide (1.2 g, 4.3 mmol, 1.0 eq) in dioxane (13 mL) was added HCl/dioxane (6.4 mL) dropwise at 0 °C. The mixture was stirred at 25 °C for 3 hr. The reaction mixture was quenched by addition of triethylamine (20 mL) in portions, and then diluted with H2O (50 mL) and extracted with ethyl acetate (50 mL). The combined organic layers were washed with brine (10 mL), dried over by Na2SO4, filtered, and the solvents were evaporated in vacuo. The residue was purified by prep-HPLC to give (R)-1-(4-amino- 3,4-dihydroquinolin-1(2H)-yl)ethan-1-one. m/z (ESI+) 174.1 (M-16)+. Step 4:
Figure imgf000077_0002
[0197] To a solution of (R)-1-(4-amino-3,4-dihydroquinolin-1(2H)-yl)ethan-1-one (50.0 mg, 262.8 µmol, 1.0 eq) and [4-[3-(1,3-dioxoisoindolin-2-yl)prop-1-ynyl]phenyl]boronic acid (96.2 mg, 315.3 µmol, 1.2 eq) in MeCN (4 mL), was added pyridine (62.3 mg, 788.4 µmol, 63.6 µL, 3.0 eq) and Cu(OAc)2 (95.4 mg, 525.6 µmol, 2.0 eq). The mixture was stirred at 25 °C for 2 hr. Water (10 mL) was added at 0 °C, and then diluted with DCM (10 mL) and extracted with DCM (10 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered, and the solvents were evaporated in vacuo. The residue was purified by column chromatography (SiO2, 0 to 50% ethyl acetate in petroleum ether) to give (R)-2-(3-(4-((1-acetyl-1,2,3,4-tetrahydroquinolin-4-yl)amino)phenyl)prop-2-yn-1-yl)isoindoline-1,3- dione. m/z (ESI+) 450.2 (M+1)+. Step 5:
Figure imgf000078_0001
[0198] To a solution of (R)-2-(3-(4-((1-acetyl-1,2,3,4-tetrahydroquinolin-4-yl)amino)phenyl)prop-2-yn- 1-yl)isoindoline-1,3-dione (50.0 mg, 111.2 µmol, 1.0 eq) in EtOH (0.3 mL) was added hydrazine hydrate (139.2 mg, 2.7 mmol, 135.1 µL, 25.0 eq). The mixture was stirred at 25 °C for 2 hr. The reaction mixture was partitioned between H2O (3 mL) and DCM (3 mL). The organic phase was separated, washed with brine (3 mL), dried over Na2SO4, filtered, and the solvents were evaporated in vacuo. The crude product was purified by prep-HPLC to give (R)-1-(4-((4-(3-aminoprop-1-yn-1-yl)phenyl)amino)- 3,4-dihydroquinolin-1(2H)-yl)ethan-1-one. 1H NMR (400 MHz, d4-MeOD) δ 7.49 - 7.24 (m, 3H), 7.18 (d, J = 8.6 Hz, 3H), 6.62 (d, J = 8.6 Hz, 2H), 4.19 - 4.10 (m, 1H), 3.66 - 3.61 (m, 2H), 2.36 - 2.24 (m, 4H), 1.87 (d, J = 9.0 Hz, 1H), 1.30 (br s, 2H).
Example 2 Preparation of 1-((2S,4R)-4-((4-(3-aminoprop-1-yn-1-yl)phenyl)amino)-6-bromo-2-methyl-3,4- dihydroquinolin-1(2H)-yl)ethan-1-one (Compound P-3):
Figure imgf000079_0001
Step 1:
Figure imgf000079_0002
[0199] To a mixture of (S)-3-aminobutanoic acid (20 g, 193.95 mmol) in ethanol (100 mL) was added thionyl chloride (39.23 g, 329.71 mmol) dropwise at 0 ℃. The mixture was stirred at 25 °C for 12 hours. The mixture was concentrated under pressure to give ethyl (S)-3-aminobutanoate, which was used directly without purification. m/z (ESI+) 132.2 (M+H)+. Step 2: [0200]
Figure imgf000079_0003
To a mixture of ethyl (S)-3-aminobutanoate (32 g, 243.95 mmol) and 1-bromo-4-iodobenzene (57.51 g, 203.29 mmol) in DMF (500 mL), was added CuI (3.87 g, 20.33 mmol), 2-(2- methylpropanoyl)cyclohexanone (6.84 g, 40.66 mmol), and Cs2CO3 (132.47 g, 406.59 mmol) at 20 ℃. The mixture was stirred at 100 °C for 12 h under N2. The mixture was poured into water (2 L) and extracted with ethyl acetate (3x500 mL). The combined organic phases were washed with brine (200 mL), dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography (SiO2, 20 to 50% ethyl acetate in petroleum ether) to give ethyl (S)-3-((4-bromophenyl)amino)butanoate. 1H NMR (400 MHz, CDCl3) δ 7.26 (d, J = 9.2 Hz, 2H), 6.49 (d, J = 7.2 Hz, 2H), 4.19 - 4.09 (m, 3H), 2.65 - 2.53 (m, 1H), 2.43 (dd, J = 6.4, 15.2 Hz, 1H), 1.29 - 1.25 (m, 6H). Step 3:
Figure imgf000080_0001
[0201] To (S)-3-((4-bromophenyl)amino)butanoate (10 g, 34.94 mmol) was added 7 N NH3 in MeOH (17.14 g, 349.45 mmol, 60 mL) at 20 °C. One additional vial was set up as above describe, and the two reactions were combined for work up. The mixture was concentrated in vacuo. The residue was purified by column chromatography (SiO2, 25 to 100% ethyl acetate in petroleum ether) to give (S)-3-((4- bromophenyl)amino)butanamide. 1H NMR (400 MHz, d6-DMSO) δ 7.23 - 7.15 (m, 2H), 6.55 - 6.48 (m, 2H), 5.67 (d, J = 8.8 Hz, 1H), 3.78 - 3.63 (m, 1H), 2.39 - 2.29 (m, 1H), 2.08 - 1.98 (m, 1H), 1.11 (d, J = 6.4 Hz, 3H). Step 4:
Figure imgf000080_0002
[0202] To a mixture of (S)-3-((4-bromophenyl)amino)butanamide (12 g, 46.67 mmol) in ethyl acetate (100 mL) was added isopropyl carbonochloridate (7.44 g, 60.67 mmol, 8.42 mL) at -10 °C. Lithium tert- butoxide (8.97 g, 112.01 mmol, 10.10 mL) in tetrahydrofuran (150 mL) was added dropwise at -10 ℃ to the mixture. The reaction was stirred 0 ℃ for 0.5 h. The mixture was partitioned between ethyl acetate (200 mL) and 2N HCl (300 mL). The organic phase was washed with brine (200 mL), dried over Na2SO4, filtered, and the solvents evaporated in vacuo. The residue was purified by column chromatography (SiO2, 20 to 50% ethyl acetate in petroleum ether) to give isopropyl (S)-(3-((4- bromophenyl)amino)butanoyl)carbamate. 1H NMR (400 MHz, CDCl3) δ 7.24 - 7.18 (m, 2H), 6.52 - 6.44 (m, 2H), 4.95 (q, J = 6.4 Hz, 1H), 4.02 - 3.92 (m, 1H), 3.07 (dd, J = 5.6, 16.0 Hz, 1H), 2.88 (dd, J = 6.0, 16.0 Hz, 1H), 1.29 - 1.26 (m, 9H). Step 5:
Figure imgf000080_0003
[0203] To a mixture of isopropyl (S)-(3-((4-bromophenyl)amino)butanoyl)carbamate (12 g, 34.96 mmol) in ethanol (120 mL) was added NaBH4 (992.06 mg, 26.22 mmol) at -20 °C. MgCl2 (3.66 g, 38.46 mmol) in water (12 mL) was added dropwise at -20 ℃. The mixture was stirred at 0 ℃ for 1 h. The reaction was poured into citric acid (40.30 g, 209.78 mmol) in 1N HCl (150 mL) and extracted with dichloromethane (3x100 mL). The combined organic phases were washed with brine (200 mL), dried over Na2SO4, filtered, and the solvents evaporated in vacuo. The residue was purified by column chromatography (SiO2, 20 to 50% ethyl acetate in petroleum ether) to give isopropyl ((2S,4R)-6-bromo-2- methyl-1,2,3,4-tetrahydroquinolin-4-yl)carbamate. 1H NMR (400 MHz, CDCl3) δ 7.47 (s, 1H), 7.31 - 7.27 (m, 1H), 6.95 (d, J = 8.0 Hz, 1H), 5.03 - 4.96 (m, 2H), 3.68 - 3.56 (m, 1H), 2.46 - 2.33 (m, 1H), 1.91 - 1.71 (m, 1H), 1.31 (dd, J = 6.0, 15.2 Hz, 6H), 1.27 - 1.23 (m, 3H). Step 6:
Figure imgf000081_0001
[0204] To a solution of isopropyl ((2S,4R)-6-bromo-2-methyl-1,2,3,4-tetrahydroquinolin-4-yl)carbamate (8.6 g, 26.28 mmol) and pyridine (8.32 g, 105.13 mmol, 8.49 mL) in dichloromethane (90 mL), was added acetyl chloride (6.19 g, 78.85 mmol, 5.63 mL) dropwise under nitrogen at 20 °C. The mixture was stirred for 1 h. The mixture was poured into saturated NaHCO3 solution (200 mL) and extracted with dichloromethane (3x100 mL). The organic phase was washed with brine (200 mL), dried over MgSO4, filtered, and the solvents evaporated in vacuo. The residue was purified by column chromatography (SiO2, 25 to 50% ethyl acetate in petroleum ether) to give isopropyl ((2S,4R)-1-acetyl-6-bromo-2-methyl- 1,2,3,4-tetrahydroquinolin-4-yl)carbamate. 1H NMR (400 MHz, d4-methanol) δ 7.48 (dd, J = 2.0, 8.4 Hz, 1H), 7.35 (s, 1H), 7.22 (d, J = 8.8 Hz, 1H), 4.93 (td, J = 6.4, 12.8 Hz, 1H), 4.85 - 4.72 (m, 1H), 4.47 (dd, J = 4.0, 12.4 Hz, 1H), 2.53 (ddd, J = 4.4, 8.8, 12.8 Hz, 1H), 2.13 (s, 3H), 1.36 - 1.21 (m, 7H), 1.12 (d, J = 6.4 Hz, 3H). Step 7:
Figure imgf000081_0002
[0205] A solution of aluminum trichloride (10.11 g, 75.83 mmol) in dichloromethane (60 mL) was added dropwise to isopropyl ((2S,4R)-1-acetyl-6-bromo-2-methyl-1,2,3,4-tetrahydroquinolin-4-yl)carbamate (7 g, 18.96 mmol) in dichloromethane (20 mL) at 0 °C under nitrogen. The mixture was stirred at 0 °C for 0.5 h. Triethylamine (23.02 g, 227.49 mmol, 31.66 mL) in methanol (8 mL) was added at 0 ℃. Ethyl acetate (50 mL) was added and the mixture was stirred at 20 ℃ for 30 minutes, and then filtered. The filter cake was dissolved in dichloromethane (200 mL) and NaHCO3 (300 mL). Sodium potassium tartrate (85 g) was added and the resulting mixture was stirred at 20 ℃ for 2 h. The organic phase was washed with brine (300 mL), dried over Na2SO4, filtered, and the solvents evaporated in vacuo. The residue was purified by prep-HPLC to give 1-((2S,4R)-4-amino-6-bromo-2-methyl-3,4-dihydroquinolin- 1(2H)-yl)ethan-1-one. 1H NMR (400 MHz, CDCl3) δ 7.66 (s, 1H), 7.41 (dd, J = 2.0, 8.4 Hz, 1H), 7.04 - 6.93 (m, 1H), 4.90 - 4.73 (m, 1H), 3.73 (dd, J = 4.4, 12.4 Hz, 1H), 2.54 (ddd, J = 4.4, 8.4, 12.8 Hz, 1H), 2.10 (s, 3H), 1.12 (d, J = 6.4 Hz, 3H). Step 8:
Figure imgf000082_0001
[0206] To a solution of 1-((2S,4R)-4-amino-6-bromo-2-methyl-3,4-dihydroquinolin-1(2H)-yl)ethan-1- one (1 g, 3.5 mmol, 1.0 eq) in ACN (10 mL), was added (4-(3-((tert-butoxycarbonyl)amino)prop-1-yn-1- yl)phenyl)boronic acid (1.9 g, 7.1 mmol, 2.0 eq), Cu(OAc)2 (641.4 mg, 3.5 mmol, 1.0 eq), and pyridine (558.7 mg, 7.1 mmol, 570.1 µL, 2.0 eq). The mixture was stirred at 25 °C for 10 hrs. The residue was diluted with H2O (20 mL) and extracted with ethyl acetate (3x15 mL). The combined organic layers were washed with brine, dried over Na2SO4, filtered, and the solvents evaporated in vacuo. The residue was purified by column chromatography (SiO2, 20 to 100% ethyl acetate in petroleum ether) to give tert-butyl (3-(4-(((2S,4R)-1-acetyl-6-bromo-2-methyl-1,2,3,4-tetrahydroquinolin-4-yl)amino)phenyl)prop-2-yn-1- yl)carbamate. 1H NMR (400 MHz, d6-DMSO) δ 7.46 (dd, J = 2.4, 8.4 Hz, 1H), 7.34 - 7.25 (m, 2H), 7.19 - 7.13 (m, 3H), 6.65 (d, J = 8.6 Hz, 2H), 6.48 (d, J = 7.8 Hz, 1H), 4.74 - 4.61 (m, 1H), 4.28 -4.26 (m, 1H), 3.93 (d, J = 5.4 Hz, 2H), 2.62 - 2.60 (m, 1H), 2.10 (s, 3H), 1.40 (s, 9H), 1.28 - 1.20 (m, 1H), 1.06 (d, J = 6.4 Hz, 3H). Step 9:
Figure imgf000083_0001
[0207] To a solution of tert-butyl (3-(4-(((2S,4R)-1-acetyl-6-bromo-2-methyl-1,2,3,4-tetrahydroquinolin- 4-yl)amino)phenyl)prop-2-yn-1-yl)carbamate (200 mg, 390.3 µmol, 1.0 eq) in DCM (3 mL), was added 2,6-dimethylpyridine (250.9 mg, 2.3 mmol, 272.7 µL, 6.0 eq) at 25 °C. The reaction was cooled to 0 °C, and trimethylsilyl trifluoromethanesulfonate (347.0 mg, 1.6 mmol, 282.10 µL, 4.0 eq) was added. The reaction was allowed to warm to room temperature, and then stirred for 2 hrs. The reaction was diluted with H2O (9 mL) and extracted into ethyl acetate (3x5 mL). The combined organic layers were washed with brine, dried over Na2SO4, filtered, and the solvents evaporated in vacuo. The residue was purified by prep-HPLC to give 1-((2S,4R)-4-((4-(3-aminoprop-1-yn-1-yl)phenyl)amino)-6-bromo-2-methyl-3,4- dihydroquinolin-1(2H)-yl)ethan-1-one. 1H NMR (400 MHz, DMSO-d6) δ 7.46 (dd, J = 2.0, 8.4 Hz, 1H), 7.32 (d, J = 8.0 Hz, 1H), 7.19 - 7.12 (m, 3H), 6.64 (d, J = 8.8 Hz, 2H), 6.44 (d, J = 7.6 Hz, 1H), 4.67 – 4.65 (m, 1H), 4.31 - 4.23 (m, 1H), 3.45 (s, 2H), 2.62 - 2.56 (m, 1H), 2.10 (s, 3H), 1.97 - 1.68 (m, 2H), 1.29 - 1.12 (m, 1H), 1.10 - 1.01 (m, 3H). Example 3 Preparation of 1-((2S,4R)-4-((4-(3-aminoprop-1-yn-1-yl)phenyl)amino)-2,6-dimethyl-3,4- dihydroquinolin-1(2H)-yl)ethan-1-one (Compound P-1):
Figure imgf000083_0002
Step 1:
Figure imgf000084_0001
[0208] A mixture tert-butyl (3-(4-(((2S,4R)-1-acetyl-6-bromo-2-methyl-1,2,3,4-tetrahydroquinolin-4- yl)amino)phenyl)prop-2-yn-1-yl)carbamate (180 mg, 351.26 µmol, 1 eq), 2,4,6-trimethyl-1,3,5,2,4,6- trioxatriborinane (431.90 mg, 1.72 mmol, 480.96 µL, 50% purity, 4.90 eq), Pd(dppf)Cl2 (25.70 mg, 35.13 µmol, 0.1 eq), and Cs2CO3 (228.90 mg, 702.52 µmol, 2 eq) in H2O (0.5 mL) and THF (2 mL) was degassed and purged with N2. The mixture was stirred at 80 °C for 2 hours. The reaction was filtered and the solvents evaporated in vacuo. The residue was purified by column chromatography (silica, 50% ethyl acetate in petroleum ether) to give tert-butyl (3-(4-(((2R,4S)-1-acetyl-2,6-dimethyl-1,2,3,4- tetrahydroquinolin-4-yl)amino)phenyl)prop-2-yn-1-yl)carbamate. m/z (ESI+): 447.25 (M+42)+. Step 2: [0209]
Figure imgf000084_0002
To a solution of tert-butyl (3-(4-(((2R,4S)-1-acetyl-2,6-dimethyl-1,2,3,4-tetrahydroquinolin-4- yl)amino)phenyl)prop-2-yn-1-yl)carbamate (20 mg, 44.69 µmol, 1 eq) in CH2Cl2 (0.5 mL), was added TMSOTf (59.59 mg, 268.12 µmol, 48.45 µL, 6 eq) and 2,6-dimethylpyridine (19.15 mg, 178.74 µmol, 20.82 µL, 4 eq). The mixture was stirred at 25 °C for 2 hr. The reaction mixture was partitioned between H2O (20 mL) and ethyl acetate (20 mL). The organic phase was separated, washed with ethyl acetate (3x10 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by prep-HPLC to give 1-((2S,4R)-4-((4-(3-aminoprop-1-yn-1-yl)phenyl)amino)-2,6-dimethyl- 3,4-dihydroquinolin-1(2H)-yl)ethan-1-one.1H NMR (400 MHz, d4-methanol) δ 7.20 - 7.10 (m, 4H), 7.05 (s, 1H), 6.59 (d, J = 8.6 Hz, 2H), 4.66 - 4.52 (m, 1H), 4.18 (br dd, J = 4.0, 12.2 Hz, 1H), 3.59 (br s, 2H), 2.65 – 2.58 (m, 1H), 2.28 (s, 3H), 2.15 (s, 3H), 1.15 - 1.09 (m, 4H). Example 4 Preparation of 1-((2S,4R)-4-((4-(3-aminoprop-1-yn-1-yl)phenyl)amino)-6-fluoro-2-methyl-3,4- dihydroquinolin-1(2H)-yl)ethan-1-one (Compound P-4):
Figure imgf000085_0001
Step 1:
Figure imgf000085_0002
[0210] A mixture of isopropyl ((2S,4R)-1-acetyl-6-bromo-2-methyl-1,2,3,4-tetrahydroquinolin-4- yl)carbamate (0.5 g, 1.35 mmol, 1 eq), bis(pinacolato)diboron (0.5 g, 2.03 mmol, 1.5 eq), KOAc (266 mg, 2.71 mmol, 2 eq), and Pd(dppf)Cl2 (100 mg, 135 µmol, 0.1 eq) in dioxane (5 mL) was de-gassed and then heated to 80 °C for 1 h under N2. The reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (2x10 mL). The combined organic layers were washed with brine (3x10 mL), dried over Na2SO4, filtered, and the solvents evaporated in vacuo. The residue was purified by column chromatography (SiO2, 30 to 50% ethyl acetate in petroleum ether) to give the crude product, isopropyl ((2S,4R)-1-acetyl-2-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,2,3,4-tetrahydroquinolin-4- yl)carbamate, which was used into the next step without further purification. m/z (ESI+) 417.2 (M+H)+. Step 2: [0211] To a solu
Figure imgf000085_0003
ton o a . mg, . mmo , . eq n e m at , was added isopropyl ((2S,4R)-1-acetyl-2-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,2,3,4- tetrahydroquinolin-4-yl)carbamate (400 mg, 960.81 µmol, 1 eq). After stirring for 15 min at 20 °C, the reaction mixture was cooled to 0 °C and silver trifluoromethanesulfonate (740.60 mg, 2.88 mmol, 3 eq) was added. The reaction was stirred at 0 ºC for 30 min. The solvents were evaporated in vacuo at 0 ºC, and the residual MeOH was removed by co-evaporation with acetone (2x2 mL). The residue was diluted with acetone (8 mL), and Select-Fluor™ (357.39 mg, 1.01 mmol, 1.05 eq) was added. The mixture was stirred for 12 h at 20 °C. The mixture was filtered and the solvents were evaporated in vacuo. The residue was purified by prep-HPLC to give the product, isopropyl ((2S,4R)-1-acetyl-6-fluoro-2-methyl- 1,2,3,4-tetrahydroquinolin-4-yl)carbamate. 1H NMR (400 MHz, CDCl3) δ 7.14 - 7.05 (m, 1H), 7.03 - 6.95 (m, 2H), 4.99 (td, J = 5.6, 12.4 Hz, 1H), 4.93 - 4.82 (m, 1H), 4.68 - 4.56 (m, 1H), 2.60 (ddd, J = 4.4, 8.4, 12.4 Hz, 1H), 2.11 (s, 3H), 1.30 (br dd, J = 6.0, 12.0 Hz, 6H), 1.21 (br dd, J = 8.8, 12.4 Hz, 1H), 1.13 (d, J = 6.4 Hz, 3H). Step 3:
Figure imgf000086_0001
[0212] To a mixture of AlCl3 (173 mg, 1.30 mmol, 63.80 µL, 4 eq) in DCM (1 mL) at 0 °C was added a solution of isopropyl ((2S,4R)-1-acetyl-6-fluoro-2-methyl-1,2,3,4-tetrahydroquinolin-4-yl)carbamate (100 mg, 324.31 µmol, 1 eq) in DCM (1 mL) at 0 °C. The mixture was stirred at 0 °C for 30 minutes. Triethylamine (2 mL) was slowly added to the reaction at 0 °C. The reaction mixture was diluted with water (5 mL) and extracted with DCM (2x10 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give the crude product, 1-((2S,4R)-4-amino-6-fluoro-2-methyl-3,4-dihydroquinolin-1(2H)-yl)ethan-1-one, which was used in the next step without further purification. m/z (ESI+): 206.0 (benzylic cation)+. Step 4:
Figure imgf000086_0002
[0213] A solution of 1-((2S,4R)-4-amino-6-fluoro-2-methyl-3,4-dihydroquinolin-1(2H)-yl)ethan-1-one (70 mg, 314.95 µmol, 1 eq), [4-[3-(tert-butoxycarbonylamino)prop-1-ynyl]phenyl]boronic acid (173.29 mg, 629.90 µmol, 2 eq), Cu(OAc)2 (57.20 mg, 314.95 µmol, 1 eq), and pyridine (49.82 mg, 629.90 µmol, 50.84 µL, 2 eq) in ACN (1 mL) was stirred at 20 °C for 12 h. The reaction mixture was filtered and the solvents were evaporated in vacuo. The residue was purified by prep-HPLC to give the product, tert- butyl (3-(4-(((2S,4R)-1-acetyl-6-fluoro-2-methyl-1,2,3,4-tetrahydroquinolin-4-yl)amino)phenyl)prop-2- yn-1-yl)carbamate. 1H NMR (400 MHz, CDCl3) δ 7.23 - 7.07 (m, 2H), 7.06 - 6.94 (m, 3H), 6.54 (d, J = 8.4 Hz, 2H), 4.99 - 4.86 (m, 1H), 4.83 - 4.70 (m, 1H), 4.24 - 4.07 (m, 4H), 2.67 (dt, J = 4.4, 8.4 Hz, 1H), 2.18 (s, 3H), 1.47 (s, 9H), 1.32 - 1.29 (m, 1H), 1.15 (br d, J = 6.4 Hz, 3H). Step 5:
Figure imgf000087_0001
[0214] To a solution of tert-butyl (3-(4-(((2S,4R)-1-acetyl-6-fluoro-2-methyl-1,2,3,4-tetrahydroquinolin- 4-yl)amino)phenyl)prop-2-yn-1-yl)carbamate (30 mg, 66.44 µmol, 1 eq) and lutidine (28.48 mg, 265.76 µmol, 30.95 µL, 4 eq) in DCM (1 mL), was added trimethylsilyl trifluoromethanesulfonate (44.30 mg, 199.32 µmol, 36.02 µL, 3 eq) dropwise at 20 °C, and the mixture was stirred for 2 h. The solvents were evaporated in vacuo. The residue was purified by prep-HPLC to give 1-((2S,4R)-4-((4-(3-aminoprop-1- yn-1-yl)phenyl)amino)-6-fluoro-2-methyl-3,4-dihydroquinolin-1(2H)-yl)ethan-1-one. 1H NMR (400 MHz, CDCl3) δ 7.26 - 7.06 (m, 3H), 7.04 - 6.93 (m, 2H), 6.54 (d, J = 8.4 Hz, 2H), 5.02 - 4.84 (m, 1H), 4.23 - 4.13 (m, 1H), 3.92 (br d, J = 7.6 Hz, 1H), 3.64 (s, 2H), 2.66 (ddd, J = 4.0, 8.4, 12.4 Hz, 1H), 2.18 (s, 3H), 1.33 - 1.27 (m, 1H), 1.15 (d, J = 6.4 Hz, 3H).
Example 5 Preparation of 1-((2S,4R)-4-((4-(3-aminoprop-1-yn-1-yl)phenyl)amino)-2-methyl-6-(1-methyl-1H- pyrazol-5-yl)-3,4-dihydroquinolin-1(2H)-yl)ethan-1-one (Compound P-7): S
Figure imgf000088_0001
[0215] To a solution of 1-((2S,4R)-4-amino-6-bromo-2-methyl-3,4-dihydroquinolin-1(2H)-yl)ethan-1- one (300.0 mg, 1.1 mmol, 1.0 eq) in DCM (5 mL), was added (4-chlorophenyl)boronic acid (248.5 mg, 1.6 mmol, 1.5 eq), Cu(OAc)2 (192.4 mg, 1.1 mmol, 1.0 eq), and Et3N (321.6 mg, 3.2 mmol, 442.4 µL, 3.0 eq). The reaction was stirred at 25 °C for 10 hrs. Water (15 mL) was added and the product extracted into ethyl acetate (3x5 mL). The combined organic layers were washed with brine, dried over Na2SO4, filtered, and the solvents were evaporated in vacuo. The residue was purified by column chromatography (SiO2, 20 to 50% ethyl acetate in petroleum ether) to give 1-((2S,4R)-6-bromo-4-((4- chlorophenyl)amino)-2-methyl-3,4-dihydroquinolin-1(2H)-yl)ethan-1-one. 1H NMR (400 MHz, d6- DMSO) δ ppm 7.50 - 7.39 (m, 2H), 7.29 (d, J = 8.2 Hz, 1H), 7.22 - 7.14 (m, 2H), 7.10 (d, J = 8.6 Hz, 2H), 6.66 (d, J = 8.6 Hz, 2H), 6.36 - 6.22 (m, 1H), 4.78 - 4.43 (m, 3H), 4.27 - 4.15 (m, 2H), 2.07 (br s, 3H), 2.03 - 1.91 (m, 1H), 1.25 - 1.10 (m, 3H), 1.03 (d, J = 6.0 Hz, 3H), 0.93 (t, J = 6.8 Hz, 7H). Step 2:
Figure imgf000089_0001
[0216] To a solution of 1-((2S,4R)-6-bromo-4-((4-chlorophenyl)amino)-2-methyl-3,4-dihydroquinolin- 1(2H)-yl)ethan-1-one (50.0 mg, 127.0 µmol, 1.0 eq) in THF (0.8 mL) and H2O (0.2 mL), was added 1- methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (39.6 mg, 190.5 µmol, 1.5 eq), K3PO4 (80.8 mg, 381.0 µmol, 3.0 eq), and XPhos-Pd-G2 (10.0 mg, 12.7 µmol, 0.1 eq). The mixture was stirred at 90 °C for 12 hrs. Water (3 mL) was added to the reaction mixture, and the product was extracted into ethyl acetate (3x1 mL). The combined organic layers were washed with brine, dried over Na2SO4, filtered, and the solvents were evaporated in vacuo. The residue was purified by prep-HPLC to give 1-((2S,4R)-4-((4-chlorophenyl)amino)-2-methyl-6-(1-methyl-1H-pyrazol-5-yl)-3,4-dihydroquinolin- 1(2H)-yl)ethan-1-one.1H NMR (400 MHz, d6-DMSO) δ 7.49 - 7.40 (m, 3H), 7.18 (s, 1H), 7.16 - 7.07 (m, 2H), 6.72 (d, J = 8.8 Hz, 2H), 6.35 - 6.27 (m, 2H), 4.71 (d, J = 6.4 Hz, 1H), 4.31 (m, J = 4.0, 8.0, 11.7 Hz, 1H), 3.69 (s, 3H), 2.62 (m, J = 4.0, 8.4, 12.4 Hz, 1H), 2.15 (s, 3H), 1.31 - 1.19 (m, 1H), 1.11 (d, J = 6.4 Hz, 3H). Step 3: [02
Figure imgf000089_0002
7] .5 m otage m crowave reactor was c arge wt -[( S, )- -[( -c orop eny)am no]- - methyl-6-(1-methyl-1H-pyrazol-5-yl)-1,2,3,4-tetrahydroquinolin-1-yl]ethan-1-one (52.0 mg, 132 µmol), prop-2-yn-1-amine (25.3 µL, 3 eq., 395 µmol), cesium carbonate (257 mg, 6 eq., 790 µmol), degassed ACN (3 mL), and Pd(Cy*Phine)2Cl2 (14.0 mg, 132 µmol). The microwave tube was then sealed under an argon atmosphere and heated at 90 °C under microwave irradiation for 2 hours. The solvents were evaporated in vacuo and the resulting residue was dissolved in a mixture of DCM and DMA (2 mL). The insoluble solid was filtered and the solvents were evaporated in vacuo. The resulting residue was purified by prep-HPLC to give 1-((2S,4R)-4-((4-(3-aminoprop-1-yn-1-yl)phenyl)amino)-2-methyl-6-(1-methyl- 1H-pyrazol-5-yl)-3,4-dihydroquinolin-1(2H)-yl)ethan-1-one. 1H NMR (499 MHz, d6-DMSO) δ 8.30 (s, 1H), 7.48 – 7.40 (m, 3H), 7.17 – 7.12 (m, 3H), 6.70 – 6.64 (m, 2H), 6.46 (d, J = 8.0 Hz, 1H), 6.28 (d, J = 1.9 Hz, 1H), 4.73 – 4.68 (m, 1H), 4.34 (ddd, J = 12.1, 8.0, 4.2 Hz, 1H), 3.66 (s, 3H), 3.53 (s, 2H), 2.62 (ddd, J = 12.5, 8.5, 4.1 Hz, 1H), 2.15 (s, 3H), 1.27 (td, J = 12.3, 9.3 Hz, 1H), 1.11 (d, J = 6.3 Hz, 3H). [0218] Each of the compounds set forth in Table 3 was prepared following one of the procedures set forth above. Table 3
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0003
Example 6 Preparation of 1-((2S)-4-(1-(4-(3-aminoprop-1-yn-1-yl)phenyl)ethyl)-2-methyl-3,4- dihydroquinoxalin-1(2H)-yl)ethan-1-one (Compound P-10A & P-3A):
Figure imgf000098_0001
Step 1: [0219] To a soluti
Figure imgf000098_0002
on of 1-[(2S)-2-methyl-3,4-dihydro-2H-quinoxalin-1-yl]ethanone (1 g, 5.26 mmol, 1 eq) and 1-bromo-4-(1-bromoethyl)benzene (1.65 g, 6.31 mmol, 1.2 eq) in DMF (15 mL) was added K2CO3 (1.45 g, 10.51 mmol, 2 eq). The mixture was stirred at 100 °C for 2 hr. The reaction mixture was partitioned between H2O (20 mL) and ethyl acetate (20 mL). The organic phase was separated, washed with ethyl acetate (3x10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC to give 1-((2S)-4-(1-(4-bromophenyl)ethyl)-2-methyl-3,4- dihydroquinoxalin-1(2H)-yl)ethan-1-one. Step 2:
Figure imgf000099_0001
[0220] To a solution of 1-((2S)-4-(1-(4-bromophenyl)ethyl)-2-methyl-3,4-dihydroquinoxalin-1(2H)- yl)ethan-1-one (50 mg, 139 µmol, 1 eq) and prop-2-yn-1-amine (15.33 mg, 278 µmol, 17.83 µL, 2 eq) in DMSO (2 mL) was added CuI (2.65 mg, 13.92 µmol, 0.1 eq), Pd(PPh3)2Cl2 (9.77 mg, 13.92 µmol, 0.1 eq) and Cs2CO3 (136.04 mg, 417.53 µmol, 3 eq). The mixture was stirred at 80 °C for 8 hr. The reaction mixture was partitioned between ethyl acetate (3 mL) and H2O (3 mL). The organic phase was separated, washed with ethyl acetate (3x3 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC to give 1-((2S)-4-(1-(4-(3-aminoprop-1-yn-1-yl)phenyl)ethyl)-2- methyl-3,4-dihydroquinoxalin-1(2H)-yl)ethan-1-one as two separable isomers. [0221] Isomer 1: MS (ESI+): m/z 348.1 (M+H)+. 1H NMR (400 MHz, d6-DMSO) δ 7.67 - 7.54 (m, 1H), 7.41 - 7.35 (m, 2H), 7.33 - 7.27 (m, 2H), 6.98 (d, J = 6.4 Hz, 1H), 6.93 - 6.86 (m, 1H), 6.59 (t, J = 7.6 Hz, 1H), 5.31 - 5.21 (m, 1H), 3.50 (s, 2H), 3.20 - 3.10 (m, 1H), 3.02 - 2.89 (m, 1H), 2.12 (s, 3H), 1.49 (d, J = 7.2 Hz, 3H), 0.95 (d, J = 3.2 Hz, 3H). [0222] Isomer 2: MS (ESI+): m/z 348.1 (M+H)+. 1H NMR (400 MHz, d6-DMSO) δ 7.66 - 7.52 (m, 1H), 7.40 - 7.35 (m, 2H), 7.34 - 7.29 (m, 2H), 7.00 - 6.92 (m, 1H), 6.87 (d, J = 8.4 Hz, 1H), 6.57 (t, J = 7.6 Hz, 1H), 5.21 (q, J = 6.4 Hz, 1H), 3.52 (s, 2H), 3.26 (m, 1H), 2.98 (d, J = 11.6 Hz, 1H), 2.12 (s, 3H), 1.51 (d, J = 7.2 Hz, 3H), 0.85 (s, 3H). Example 7 (S)-1-(4-(4-(3-aminoprop-1-yn-1-yl)benzyl)-6-fluoro-2-methyl-3,4-dihydroquinoxalin-1(2H)- yl)ethan-1-one (Compound P-2A):
Figure imgf000100_0001
Step 1:
Figure imgf000100_0002
[0223] To a mixture of 4-bromo-1-fluoro-2-nitrobenzene (100 g, 454.55 mmol, 55.87 mL) and (2S)-2- aminopropan-1-ol (37.56 g, 500.01 mmol, 38.92 mL) in DMF (500 mL) was added K2CO3 (69.10 g, 500.01 mmol). The mixture was stirred at 60°C for 12 h. Water (2 L) was added dropwise to the reaction mixture to give a precipitate. The precipitated solid was filtered. The solid was washed with water (1 L) and dried to give (S)-2-((4-bromo-2-nitrophenyl)amino)propan-1-ol. m/z (ESI+): 275.0 (M+H)+. Step 2: [0224] A mixture o
Figure imgf000100_0003
Fe (233.45 g, 4.18 mo ) an NH4C (2.24 g, 41.80 mmo) n EtOH (600 mL) and H2O (360 mL) was heated to 80 °C. (S)-2-((4-bromo-2-nitrophenyl)amino)propan-1-ol (115 g, 418.03 mmol) in EtOH (600 mL) was added dropwise. The mixture was stirred at 80°C for 4 h. The mixture was cooled to 20°C, filtered and concentrated to give (S)-2-((2-amino-4-bromophenyl)amino)propan-1-ol. 1H NMR (400 MHz, d6-DMSO) δ = 6.67 (d, J = 2.4 Hz, 1H), 6.57 (dd, J = 2.4, 8.4 Hz, 1H), 6.37 (d, J = 8.8 Hz, 1H), 4.81 (s, 2H), 4.68 (t, J = 5.6 Hz, 1H), 4.20 (d, J = 6.4 Hz, 1H), 3.49 - 3.42 (m, 1H), 3.30 - 3.23 (m, 1H), 1.11 (d, J = 6.4 Hz, 3H). Step 3:
Figure imgf000101_0001
[0225] A mixture of (S)-2-((2-amino-4-bromophenyl)amino)propan-1-ol (93 g, 379.41 mmol) in pyridine (1 L) was added 4-methylbenzene-1-sulfonyl chloride (72.33 g, 379.41 mmol) dropwise at 0 °C. The mixture was stirred at 0°C for 2 h. The mixture was poured into water (2 L) and extracted with ethyl acetate (3 × 1 L). The organic phase was washed with brine (1.5 L), dried over Na2SO4 and concentrated to give a residue. The residue was purified by column chromatography (SiO2, 25 to 50% ethyl acetate in petroleum ether) to give (S)-N-(5-bromo-2-((1-hydroxypropan-2-yl)amino)phenyl)-4- methylbenzenesulfonamide. 1H NMR (400 MHz, d6-DMSO) δ 7.55 (d, J = 8.0 Hz, 2H), 7.35 (d, J = 8.0 Hz, 2H), 7.13 (dd, J = 2.4, 8.8 Hz, 1H), 6.81 (d, J = 2.4 Hz, 1H), 6.52 (d, J = 9.2 Hz, 1H), 4.69 (t, J = 5.2 Hz, 1H), 3.29 (dd, J = 4.0, 7.2 Hz, 2H), 3.16 - 3.04 (m, 1H), 2.36 (s, 3H), 0.94 (d, J = 6.0 Hz, 3H). Step 4:
Figure imgf000101_0002
[0226] To a mixture of (S)-N-(5-bromo-2-((1-hydroxypropan-2-yl)amino)phenyl)-4- methylbenzenesulfonamide (140 g, 350.61 mmol) and PPh3 (110.35 g, 420.73 mmol) in THF (2.5 L) was added DIAD (77.99 g, 385.67 mmol, 74.99 mL) dropwise at 0°C. The mixture was stirred at 0°C for 1.5 h. The mixture was concentrated under pressure to give a residue. The residue was purified by column chromatography (SiO2, 25 to 50% ethyl acetate in petroleum ether) to give (S)-7-bromo-3-methyl-1-tosyl- 1,2,3,4-tetrahydroquinoxaline.1H NMR (400 MHz, CDCl3) δ 7.81 (d, J = 2.4 Hz, 1H), 7.48 (d, J = 8.4 Hz, 2H), 7.21 (d, J = 8.4 Hz, 2H), 7.03 (dd, J = 2.4, 8.8 Hz, 1H), 6.34 (d, J = 8.8 Hz, 1H), 4.24 - 4.13 (m, 1H), 3.77 (s, 1H), 2.84 - 2.74 (m, 2H), 2.38 (s, 3H), 1.03 (d, J = 5.6 Hz, 3H). Step 5:
Figure imgf000102_0001
[0227] To a mixture of (S)-7-bromo-3-methyl-1-tosyl-1,2,3,4-tetrahydroquinoxaline (133 g, 348.82 mmol) and pyridine (41.39 g, 523.23 mmol, 42.23 mL) in DCM (3 L) at 0°C was added acetyl chloride (32.86 g, 418.58 mmol, 29.87 mL) dropwise. The mixture was stirred at 0°C for 1.5 h. The mixture was concentrated under pressure to give (S)-1-(6-bromo-2-methyl-4-tosyl-3,4-dihydroquinoxalin-1(2H)- yl)ethan-1-one. m/z (ESI+): 423.0 (M+H)+. Step 6:
Figure imgf000102_0002
[0228] To a mixture of (S)-1-(6-bromo-2-methyl-4-tosyl-3,4-dihydroquinoxalin-1(2H)-yl)ethan-1-one (130 g, 307.09 mmol) in DCM (650 mL) at 0°C was added H2SO4 (180.72 g, 1.84 mol, 98.22 mL) dropwise. The mixture was stirred at 0°C for 1.5 h. The reaction mixture was added to ice water (5 L) slowly to maintain the solution temperature below 10°C. The mixture was extracted with dichloromethane (3 × 1 L). The combined organic phases were washed with brine (1 L), dried over Na2SO4 and concentrated to give a residue. The residue was purified by column chromatography (SiO2, 50 to 70% ethyl acetate in petroleum ether) to give (S)-1-(6-bromo-2-methyl-3,4-dihydroquinoxalin- 1(2H)-yl)ethan-1-one. 1H NMR (400 MHz, CDCl3) δ 6.83 (br s, 1H), 6.78 - 6.65 (m, 2H), 5.20 (br s, 1H), 4.18 (br s, 1H), 3.42 (dd, J = 4.0, 11.6 Hz, 1H), 3.22 (d, J = 11.6 Hz, 1H), 2.23 (s, 3H), 1.06 (s, 3H). Step 7: [0229] To a solution
Figure imgf000102_0003
o (S)- -(6- romo- -met y-3, - y roqu noxa n- ( )-y )ethan-1-one (2 g, 7.43 mmol, 1 eq) and DMAP (90.79 mg, 743.12 µmol, 0.1 eq) in DCE (30 mL) was added Boc2O (2.43 g, 11.15 mmol, 2.56 mL, 1.5 eq) dropwise. The mixture was stirred at 50°C for 1 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, 10 to 25% ethyl acetate in petroleum ether) to give tert-butyl (S)-4-acetyl-7- bromo-3-methyl-3,4-dihydroquinoxaline-1(2H)-carboxylate. 1H NMR (400 MHz, CDCl3) δ 8.12 (s, 1H), 7.19 (dd, J = 2.4, 8.4 Hz, 1H), 7.04 (s, 1H), 5.13 (s, 1H), 3.93 (dd, J = 6.4, 12.4 Hz, 1H), 3.56 - 3.42 (m, 1H), 2.19 (s, 3H), 1.54 (s, 9H), 1.04 (d, J = 6.8 Hz, 3H). Step 8:
Figure imgf000103_0001
[0230] A mixture of tert-butyl (S)-4-acetyl-7-bromo-3-methyl-3,4-dihydroquinoxaline-1(2H)- carboxylate (1.5 g, 4.06 mmol, 1 eq), BPD (3.09 g, 12.19 mmol, 3 eq), Pd(dppf)Cl2 (297.24 mg, 406.23 µmol, 0.1 eq) and KOAc (1.20 g, 12.19 mmol, 3 eq) in DMSO (15 mL) was heated at 80°C for 12 h under a N2 atmosphere. The reaction mixture was diluted with water (100 mL) and extracted with ethyl acetate (2 × 50 mL). The combined organic layers were washed with brine (4 × 100 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, 20 to 35% ethyl acetate in petroleum ether) to give tert-butyl (S)-4-acetyl-3- methyl-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,4-dihydroquinoxaline-1(2H)-carboxylate. 1H NMR (400 MHz, CDCl3) δ 8.20 (s, 1H), 7.51 (dd, J = 1.2, 8.0 Hz, 1H), 7.14 (d, J = 7.6 Hz, 1H), 5.21 - 5.06 (m, 1H), 4.08 (dd, J = 7.2, 12.8 Hz, 1H), 3.34 (dd, J = 4.4, 12.8 Hz, 1H), 2.21 (s, 3H), 1.55 (s, 9H), 1.35 (s, 12H), 1.05 (d, J = 6.8 Hz, 3H). Step 9: [0231] To a
Figure imgf000103_0002
mixture of KOtBu (424.51 mg, 3.78 mmol, 1.05 eq) in MeOH (15 mL) was added tert-butyl (S)-4-acetyl-3-methyl-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,4-dihydroquinoxaline-1(2H)- carboxylate (1.5 g, 3.60 mmol, 1 eq) at 20°C. After stirring for 15 min, the reaction mixture was cooled to 0°C and AgOTf (2.78 g, 10.81 mmol, 3 eq) was added. After stirring for 30 min at 0 °C, the solvent was removed under reduced pressure at 0°C and the residual MeOH was removed by co-evaporation with acetone (20 mL × 3) to give a residue. The residue was dissolved in acetone (20 mL) and 1- (chloromethyl)-4-fluoro-1,4-diazoniabicyclo[2.2.2]octane;ditetrafluoroborate (1.34 g, 3.78 mmol, 1.05 eq) was added. The mixture was stirred at 20°C for 0.5 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give tert-butyl (S)-4-acetyl-7-fluoro-3-methyl-3,4-dihydroquinoxaline-1(2H)-carboxylate. 1H NMR (400 MHz, CDCl3) δ 7.76 (d, J = 9.6 Hz, 1H), 7.09 (br s, 1H), 6.86 - 6.72 (m, 1H), 5.19 (br s, 1H), 3.94 (dd, J = 6.4, 12.4 Hz, 1H), 3.48 (d, J = 12.4 Hz, 1H), 2.18 (s, 3H), 1.55 (s, 9H), 1.03 (d, J = 6.8 Hz, 3H). Step 10:
Figure imgf000104_0001
[0232] To a solution of tert-butyl (S)-4-acetyl-7-fluoro-3-methyl-3,4-dihydroquinoxaline-1(2H)- carboxylate (0.46 g, 1.49 mmol, 1 eq) in DCM (10 mL) was added TFA (1.70 g, 14.92 mmol, 1.10 mL, 10 eq) dropwise. The mixture was stirred for 12 h. The reaction mixture was concentrated under reduced pressure to give (S)-1-(6-fluoro-2-methyl-3,4-dihydroquinoxalin-1(2H)-yl)ethan-1-one as a TFA salt, which was used into the next step without purification. 1H NMR (400 MHz, CDCl3) δ 6.93 (br s, 1H), 6.42 (dt, J = 2.4, 8.4 Hz, 1H), 6.36 (dd, J = 2.4, 9.6 Hz, 1H), 5.24 (br s, 1H), 3.49 (d, J = 8.8 Hz, 1H), 3.26 (d, J = 11.6 Hz, 1H), 2.34 (s, 3H), 1.08 (br s, 3H). Step 11: I [0233] A so
Figure imgf000104_0002
lutio of (S) 1 (6 fluo o 2 ethyl 3,4 dihyd oqui o ali 1(2H) yl)etha 1 o e (400 mg, 1.24 mmol, 1 eq, TFA salt), KI (20.61 mg, 124.13 µmol, 0.1 eq), DIEA (802.12 mg, 6.21 mmol, 1.08 mL, 5 eq) and 1-(bromomethyl)-4-iodo-benzene (1.11 g, 3.72 mmol, 3 eq) in DMF (4 mL) was stirred at 50°C for 1 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, 10 to 25% ethyl acetate in petroleum ether to give (S)-1-(6-fluoro-4-(4-iodobenzyl)-2-methyl-3,4-dihydroquinoxalin-1(2H)-yl)ethan-1-one. 1H NMR (400 MHz, CDCl3) δ 7.67 (d, J = 8.4 Hz, 2H), 6.97 (d, J = 8.4 Hz, 2H), 6.94 (d, J = 2.0 Hz, 1H), 6.38 (dt, J = 2.4, 8.4 Hz, 1H), 6.32 (dd, J = 2.4, 11.2 Hz, 1H), 5.36 - 5.17 (m, 1H), 4.43 (s, 2H), 3.55 (dd, J = 4.4, 11.2 Hz, 1H), 3.12 (d, J = 11.6 Hz, 1H), 2.22 (s, 3H), 1.08 (d, J = 4.4 Hz, 3H). Step 12:
Figure imgf000105_0001
[0234] To a solution of (S)-1-(6-fluoro-4-(4-iodobenzyl)-2-methyl-3,4-dihydroquinoxalin-1(2H)- yl)ethan-1-one (20 mg, 47.14 µmol, 1 eq) and prop-2-yn-1-amine (7.79 mg, 141.43 µmol, 9.06 µL, 3 eq) in ACN (1 mL) was added Pd(PPh3)2Cl2 (3.31 mg, 4.71 µmol, 0.1 eq), Cs2CO3 (46.08 mg, 141.43 µmol, 3 eq) and CuI (897.82 µg, 4.71 µmol, 0.1 eq) under a N2 atmosphere. The mixture was stirred at 30°C for 12 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (Prep-HPLC method: column: Phenomenex Luna C18 75×30mm×3µm;mobile phase: [water(FA)-ACN]; B%: 1%-40%, 8min) to give (S)-1-(4-(4-(3- aminoprop-1-yn-1-yl)benzyl)-6-fluoro-2-methyl-3,4-dihydroquinoxalin-1(2H)-yl)ethan-1-one. 1H NMR (400 MHz, d4-MeOD) δ 8.62 - 8.43 (m, 1H), 7.44 (d, J = 8.0 Hz, 2H), 7.26 (d, J = 8.0 Hz, 2H), 7.15 - 7.03 (m, 1H), 6.46 - 6.31 (m, 2H), 5.26 - 5.08 (m, 1H), 4.67 - 4.46 (m, 2H), 3.86 (s, 2H), 3.60 - 3.50 (m, 1H), 3.26 - 3.19 (m, 1H), 2.20 (s, 3H), 1.06 (dd, J = 1.6, 3.6 Hz, 3H). [0235] Each of the compounds set forth in Table 3A was prepared following one of the procedures set forth above. Table 3A Compound
Figure imgf000105_0002
Figure imgf000106_0001
Figure imgf000107_0001
Biological Examples Target engagement assay [0236] The ability of a compound to bind to BRD4 is assessed using HEK-293T cells and the NanoBRET Target Engagement Intracellular BET BRD Assay from Promega (Cat. No. N2131). Assays were run according to manufacturer’s specifications and luminescent readout was recorded using a CLARIOstar Plus instrument (BMG Labtech). Resulting BRET ratios were plotted and IC50s determined by non-linear regression analyses (GraphPad Prism). BRD4 degradation assay [0237] BRD4 degradation was monitored by immunofluorescence in HEK-293T cells. In brief, 96-well plates (black, clear-bottom) were seeded with HEK-293T cells at 10,000 cells/well and incubated overnight at 37 °C to allow cell attachment. After overnight incubation, test compounds were added in a 10-point dilution series (typically 30 µM to 100 pM) using a TECAN D300e Digital Dispenser, and plates were subsequently incubated for 24 hours at 37 °C. Media was carefully removed, and cells were fixed in PBS + 2.5% formalin (50 µL) for 20 minutes at 37 °C. Following formalin fixation, cells were washed once with PBS and methanol was added (30 µL). Plates were wrapped in parafilm and incubated at -20 °C for 1 hour to overnight. For immunostaining, plates were removed from -20 °C and cells were washed with PBS (3x).50 µL of blocking solution (PBS + 1X fish gelatin, 0.3% Triton X-100) were added to each well and plates were incubated at room temperature for 30 minutes to 1 hour. Blocking buffer was removed, and 40 µL primary BRD4 antibody (Sigma-Aldrich HPA061646; diluted 1:1000 in PBS + 1X fish gelatin, 0.1% Triton X-100) was added. Plates were incubated at 4 °C for 4 hours to overnight, after which cells were washed (3x) with PBS + 0.1% tween-20 at room temperature. DAPI (1 µM final) plus secondary antibody (Southern Biotech 4030-30 anti-Rabbit IgG Alexa Fluor 488; diluted 1:2000 in PBS + 1X fish gelatin, 0.1% Triton X-100) were added at 40 µL/well, and plates were incubated at room temperature for 2 hours, covered with foil. Cells were washed (3x) with PBS + 0.1% tween-20, followed by one wash with PBS, and the addition of 100 µL of PBS for imaging. Images were acquired using the ImageXpress Pico system (Molecular Devices). Cell Reporter Xpress software was utilized to segment cells and determine fluorescence intensities, which were used to construct dose- response curves and calculation of degradation DC50s (GraphPad Prism). Results for certain compounds are reported in Table 4 and 4A below. Table 4
Figure imgf000108_0001
Figure imgf000109_0001
Table 4A
Figure imgf000110_0001

Claims

WHAT IS CLAIMED IS: 1. A compound of formula I:
Figure imgf000111_0001
or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein: m is an integer from 0 to 4; n is an integer from 0 to 4; Y is CH(R4), NR5, or O; each of X1 and X2 is independently CH(R4) or NR5; Q is H, -CH3, -CH2OH, -CH(CH3)N(R13)2, -CH2N(R13)2, -CH2N(R13)C(O)R1, or -CH2CH2N(R13)2; R1 is C1-C6 alkyl, C3-C10 cycloalkyl, aryl, heteroaryl, or heterocyclyl, wherein each C1-C6 alkyl, C3-C10 cycloalkyl, aryl, heteroaryl, or heterocyclyl is independently unsubstituted or substituted with one to five Z1a; each of R2 and R3 is independently selected from halo, -N(R13)2, -OR14, C1-C6 alkyl, aryl, cyano, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, guanidino, and nitro, wherein each C1-C6 alkyl, aryl, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, and guanidino is independently unsubstituted or substituted with one to five Z1a; each R4 is independently H, halo, cyano, hydroxy, -SH, -N(R13)2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl, wherein each of C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each R5 is independently H, -C(O)R13, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl, wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each Z1a is independently halo, cyano, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -N(R13)2, -OR13, -C(O)R13, -C(O)OR13, -S(O)0-2R13, -NR13S(O)1-2-R13, -S(O)1-2N(R13)2, -NR13S(O)1-2N(R13)2, -NR13C(O)N(R13)2, -C(O)N(R13)2, -NR13C(O)R13, -OC(O)N(R13)2, or -NR13C(O)OR13; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl is independently unsubstituted or substituted with one to five Z1b; each R13 is independently H, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R13 is independently unsubstituted or substituted with one to five Z1b; each R14 is independently H, C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R14 is independently unsubstituted or substituted with one to five Z1b; each Z1b is independently halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -L-C1-6 alkyl, -L-C2-6 alkenyl, -L-C2-6 alkynyl, -L-C1-6 haloalkyl, -L-C3-10 cycloalkyl, -L-heterocyclyl, -L-aryl, or -L-heteroaryl; each L is independently -O-, -NH-, -S-, -S(O)-, -S(O)2-, -N(C1-C6 alkyl)-, -N(C2-C6 alkenyl), -N(C2-C6 alkynyl)-, -N(C1-C6 haloalkyl)-, -N(C3-10 cycloalkyl)-, -N(heterocyclyl)-, -N(aryl)-, -N(heteroaryl)-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)N(C1-C6 alkyl)-, -C(O)N(C2-C6 alkenyl)-, -C(O)N(C2-C6 alkynyl)-, -C(O)N(C1-6 haloalkyl)-, -C(O)N(C3-10 cycloalkyl)-, -C(O)N(heterocyclyl)-, -C(O)N(aryl)-, -C(O)N(heteroaryl)-, -NHC(O)-, -NHC(O)O-, -NHC(O)NH-, -NHS(O)-, or -S(O)2NH-; and wherein each C1-C6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, and heteroaryl of Z1b and L is further independently unsubstituted or substituted with one to five halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl.
2. A compound of formula I:
Figure imgf000113_0001
or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein: m is an integer from 0 to 4; n is an integer from 0 to 4; Y is CH(R4), NR5, or O; each of X1 and X2 is independently CH(R4) or NR5; Q is H, -CH3, -CH2OH, -CH(CH3)N(R13)2, -CH2N(R13)2, -CH2N(R13)C(O)R1, or -CH2CH2N(R13)2; R1 is C1-C6 alkyl, C3-C10 cycloalkyl, aryl, heteroaryl, or heterocyclyl, wherein each C1-C6 alkyl, C3-C10 cycloalkyl, aryl, heteroaryl, or heterocyclyl is independently unsubstituted or substituted with one to five Z1a; each of R2 and R3 is independently selected from halo, -N(R13)2, -OR14, C1-C6 alkyl, aryl, cyano, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, guanidino, and nitro, wherein each C1-C6 alkyl, aryl, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, and guanidino is independently unsubstituted or substituted with one to five Z1a; each R4 is independently H, halo, cyano, hydroxy, -SH, -N(R13)2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl, wherein each of C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each R5 is independently H, -C(O)R13, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl, wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each Z1a is independently halo, cyano, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -N(R13)2, -OR13, -C(O)R13, -C(O)OR13, -S(O)0-2R13, -NR13S(O)1-2-R13, -S(O)1-2N(R13)2, -NR13S(O)1-2N(R13)2, -NR13C(O)N(R13)2, -C(O)N(R13)2, -NR13C(O)R13, -OC(O)N(R13)2, or -NR13C(O)OR13; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl is independently unsubstituted or substituted with one to five Z1b; each R13 is independently H, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R13 is independently unsubstituted or substituted with one to five Z1b; each R14 is independently H, C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R14 is independently unsubstituted or substituted with one to five Z1b; each Z1b is independently halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -L-C1-6 alkyl, -L-C2-6 alkenyl, -L-C2-6 alkynyl, -L-C1-6 haloalkyl, -L-C3-10 cycloalkyl, -L-heterocyclyl, -L-aryl, or -L-heteroaryl; each L is independently -O-, -NH-, -S-, -S(O)-, -S(O)2-, -N(C1-C6 alkyl)-, -N(C2-C6 alkenyl), -N(C2-C6 alkynyl)-, -N(C1-C6 haloalkyl)-, -N(C3-10 cycloalkyl)-, -N(heterocyclyl)-, -N(aryl)-, -N(heteroaryl)-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)N(C1-C6 alkyl)-, -C(O)N(C2-C6 alkenyl)-, -C(O)N(C2-C6 alkynyl)-, -C(O)N(C1-6 haloalkyl)-, -C(O)N(C3-10 cycloalkyl)-, -C(O)N(heterocyclyl)-, -C(O)N(aryl)-, -C(O)N(heteroaryl)-, -NHC(O)-, -NHC(O)O-, -NHC(O)NH-, -NHS(O)-, or -S(O)2NH-; and wherein each C1-C6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, and heteroaryl of Z1b and L is further independently unsubstituted or substituted with one to five halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl, provided that the compound at 1 µM concentration degrades BRD4 by 30% or more.
3. A compound of formula IA:
Figure imgf000115_0001
or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein: m is an integer from 0 to 4; n is an integer from 0 to 4; Y is CH(R4), NR5, or O; each of X1 and X2 is independently CH(R4) or NR5; each R2 is independently selected from halo, -N(R13)2, -OR14, C1-C6 alkyl, aryl, cyano, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, guanidino, and nitro, wherein each C1-C6 alkyl, aryl, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, and guanidino is independently unsubstituted or substituted with one to five Z1a, provided that R2 is not imidazolyl, isooxazolyl, pyrazol-4-yl, or pyrimidin-5-yl; each R3 is independently selected from halo, -N(R13)2, -OR14, C1-C6 alkyl, aryl, cyano, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, guanidino, and nitro, wherein each C1-C6 alkyl, aryl, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, and guanidino is independently unsubstituted or substituted with one to five Z1a; each R4 is independently H, halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl, wherein each of C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each R5 is independently H, -C(O)R13, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl, wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each Z1a is independently halo, cyano, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -N(R13)2, -OR13, -C(O)R13, -C(O)OR13, -S(O)0-2R13, -NR13S(O)1-2-R13, -S(O)1-2N(R13)2, -NR13S(O)1-2N(R13)2, -NR13C(O)N(R13)2, -C(O)N(R13)2, -NR13C(O)R13, -OC(O)N(R13)2, or -NR13C(O)OR13; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl is independently unsubstituted or substituted with one to five Z1b; each R13 is independently H, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R13 is independently unsubstituted or substituted with one to five Z1b; each R14 is independently H, C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R14 is independently unsubstituted or substituted with one to five Z1b; each Z1b is independently halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -L-C1-6 alkyl, -L-C2-6 alkenyl, -L-C2-6 alkynyl, -L-C1-6 haloalkyl, -L-C3-10 cycloalkyl, -L-heterocyclyl, -L-aryl, or -L-heteroaryl; each L is independently -O-, -NH-, -S-, -S(O)-, -S(O)2-, -N(C1-C6 alkyl)-, -N(C2-C6 alkenyl), -N(C2-C6 alkynyl)-, -N(C1-C6 haloalkyl)-, -N(C3-10 cycloalkyl)-, -N(heterocyclyl)-, -N(aryl)-, -N(heteroaryl)-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)N(C1-C6 alkyl)-, -C(O)N(C2-C6 alkenyl)-, -C(O)N(C2-C6 alkynyl)-, -C(O)N(C1-6 haloalkyl)-, -C(O)N(C3-10 cycloalkyl)-, -C(O)N(heterocyclyl)-, -C(O)N(aryl)-, -C(O)N(heteroaryl)-, -NHC(O)-, -NHC(O)O-, -NHC(O)NH-, -NHS(O)-, or -S(O)2NH-; and wherein each C1-C6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, and heteroaryl of Z1b and L is further independently unsubstituted or substituted with one to five halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl, provided that the following compounds are excluded:
, ,
Figure imgf000117_0001
, , ,
Figure imgf000118_0001
Figure imgf000119_0001
.
4. The compound of claim 3, wherein the compound is represented by formula IB: or a pharmaceutically accepta
Figure imgf000119_0002
ble salt, solvate, stereoisomer, or tautomer thereof.
5. The compound of claim 4, wherein the compound is represented by formula IC: or a pharmaceutically accepta
Figure imgf000119_0003
, , , eof.
6. The compound of claim 5, wherein the compound is represented by formula ID:
Figure imgf000120_0001
or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof.
7. The compound of claim 6, wherein R4 is C1-C6 alkyl and n is 0.
8. The compound of claim 6, wherein the compound is represented by formula 1E: NH or a pharmaceutically acceptable s
Figure imgf000120_0002
alt, solvate, stereoisomer, or tautomer thereof.
9. The compound of claim 8, wherein R2 is H, halo, C1-C6 alkyl, phenyl substituted with C1-C6 alkoxy, pyridyl, or 1-methylpyrazol-5-yl.
10. A compound of formula I-1:
Figure imgf000121_0001
or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein: m is an integer from 0 to 4; n is an integer from 0 to 4; each of X1 and X2 is independently CH(R4) or NR5; Q is H, -CH2OH, -CH2N(R13)2, -CH2CH2N(R13)2, -CH2N(R13)C(O)R1, or heterocyclyl; R1 is C1-C6 alkyl, C3-C10 cycloalkyl, aryl, heteroaryl, or heterocyclyl, wherein each C1-C6 alkyl, C3-C10 cycloalkyl, aryl, heteroaryl, or heterocyclyl is independently unsubstituted or substituted with one to five Z1a; each of R2 and R3 is independently selected from halo, -N(R13)2, -OR14, C1-C6 alkyl, aryl, cyano, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, guanidino, and nitro, wherein each C1-C6 alkyl, aryl, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, and guanidino is independently unsubstituted or substituted with one to five Z1a; each R4 is independently H, halo, cyano, hydroxy, -SH, -N(R13)2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl, wherein each of C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each R5 is independently H, -C(O)R13, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl, wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each Z1a is independently halo, cyano, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -N(R13)2, -OR13, -C(O)R13, -C(O)OR13, -S(O)0-2R13, -NR13S(O)1-2-R13, -S(O)1-2N(R13)2, -NR13S(O)1-2N(R13)2, -NR13C(O)N(R13)2, -C(O)N(R13)2, -NR13C(O)R13, -OC(O)N(R13)2, or -NR13C(O)OR13; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl is independently unsubstituted or substituted with one to five Z1b; each R13 is independently H, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R13 is independently unsubstituted or substituted with one to five Z1b; each R14 is independently H, C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R14 is independently unsubstituted or substituted with one to five Z1b; each Z1b is independently halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -L-C1-6 alkyl, -L-C2-6 alkenyl, -L-C2-6 alkynyl, -L-C1-6 haloalkyl, -L-C3-10 cycloalkyl, -L-heterocyclyl, -L-aryl, or -L-heteroaryl; each L is independently -O-, -NH-, -S-, -S(O)-, -S(O)2-, -N(C1-C6 alkyl)-, -N(C2-C6 alkenyl), -N(C2-C6 alkynyl)-, -N(C1-C6 haloalkyl)-, -N(C3-10 cycloalkyl)-, -N(heterocyclyl)-, -N(aryl)-, -N(heteroaryl)-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)N(C1-C6 alkyl)-, -C(O)N(C2-C6 alkenyl)-, -C(O)N(C2-C6 alkynyl)-, -C(O)N(C1-6 haloalkyl)-, -C(O)N(C3-10 cycloalkyl)-, -C(O)N(heterocyclyl)-, -C(O)N(aryl)-, -C(O)N(heteroaryl)-, -NHC(O)-, -NHC(O)O-, -NHC(O)NH-, -NHS(O)-, or -S(O)2NH-; and wherein each C1-C6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, and heteroaryl of Z1b and L is further independently unsubstituted or substituted with one to five halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl.
11. A compound of formula I-1:
Figure imgf000122_0001
I-1 or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, wherein: m is an integer from 0 to 4; n is an integer from 0 to 4; each of X1 and X2 is independently CH(R4) or NR5; Q is H, -CH2OH, -CH2N(R13)2, -CH2CH2N(R13)2, -CH2N(R13)C(O)R1, or heterocyclyl; R1 is C1-C6 alkyl, C3-C10 cycloalkyl, aryl, heteroaryl, or heterocyclyl, wherein each C1-C6 alkyl, C3-C10 cycloalkyl, aryl, heteroaryl, or heterocyclyl is independently unsubstituted or substituted with one to five Z1a; each of R2 and R3 is independently selected from halo, -N(R13)2, -OR14, C1-C6 alkyl, aryl, cyano, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, guanidino, and nitro, wherein each C1-C6 alkyl, aryl, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, and guanidino is independently unsubstituted or substituted with one to five Z1a; each R4 is independently H, halo, cyano, hydroxy, -SH, -N(R13)2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl, wherein each of C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each R5 is independently H, -C(O)R13, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl, wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each Z1a is independently halo, cyano, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -N(R13)2, -OR13, -C(O)R13, -C(O)OR13, -S(O)0-2R13, -NR13S(O)1-2-R13, -S(O)1-2N(R13)2, -NR13S(O)1-2N(R13)2, -NR13C(O)N(R13)2, -C(O)N(R13)2, -NR13C(O)R13, -OC(O)N(R13)2, or -NR13C(O)OR13; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl is independently unsubstituted or substituted with one to five Z1b; each R13 is independently H, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R13 is independently unsubstituted or substituted with one to five Z1b; each R14 is independently H, C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R14 is independently unsubstituted or substituted with one to five Z1b; each Z1b is independently halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -L-C1-6 alkyl, -L-C2-6 alkenyl, -L-C2-6 alkynyl, -L-C1-6 haloalkyl, -L-C3-10 cycloalkyl, -L-heterocyclyl, -L-aryl, or -L-heteroaryl; each L is independently -O-, -NH-, -S-, -S(O)-, -S(O)2-, -N(C1-C6 alkyl)-, -N(C2-C6 alkenyl), -N(C2-C6 alkynyl)-, -N(C1-C6 haloalkyl)-, -N(C3-10 cycloalkyl)-, -N(heterocyclyl)-, -N(aryl)-, -N(heteroaryl)-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)N(C1-C6 alkyl)-, -C(O)N(C2-C6 alkenyl)-, -C(O)N(C2-C6 alkynyl)-, -C(O)N(C1-6 haloalkyl)-, -C(O)N(C3-10 cycloalkyl)-, -C(O)N(heterocyclyl)-, -C(O)N(aryl)-, -C(O)N(heteroaryl)-, -NHC(O)-, -NHC(O)O-, -NHC(O)NH-, -NHS(O)-, or -S(O)2NH-; and wherein each C1-C6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, and heteroaryl of Z1b and L is further independently unsubstituted or substituted with one to five halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl, provided that the compound at 1 µM concentration degrades BRD4 by 30% or more.
12. A compound of formula IA-1: or a pharmaceutically a
Figure imgf000124_0001
cceptable salt, solvate, stereoisomer, or tautomer thereof, wherein: m is an integer from 0 to 4; n is an integer from 0 to 4; each of X1 and X2 is independently CH(R4) or NR5; each of R2 and R3 is independently selected from halo, -N(R13)2, -OR14, C1-C6 alkyl, aryl, cyano, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, guanidino, and nitro, wherein each C1-C6 alkyl, aryl, C3-C10 cycloalkyl, heteroaryl, heterocyclyl, and guanidino is independently unsubstituted or substituted with one to five Z1a; each R4 is independently H, halo, cyano, hydroxy, -SH, -N(R13)2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl, wherein each of C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each R5 is independently H, -C(O)R13, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl, wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -C(O)OC1-C6 alkyl, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to five Z1a; each Z1a is independently halo, cyano, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -N(R13)2, -OR13, -C(O)R13, -C(O)OR13, -S(O)0-2R13, -NR13S(O)1-2-R13, -S(O)1-2N(R13)2, -NR13S(O)1-2N(R13)2, -NR13C(O)N(R13)2, -C(O)N(R13)2, -NR13C(O)R13, -OC(O)N(R13)2, or -NR13C(O)OR13; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl is independently unsubstituted or substituted with one to five Z1b; each R13 is independently H, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R13 is independently unsubstituted or substituted with one to five Z1b; each R14 is independently H, C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl; wherein each C1-C6 alkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl of R14 is independently unsubstituted or substituted with one to five Z1b; each Z1b is independently halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C3-10 cycloalkyl, heterocyclyl, aryl, heteroaryl, -L-C1-6 alkyl, -L-C2-6 alkenyl, -L-C2-6 alkynyl, -L-C1-6 haloalkyl, -L-C3-10 cycloalkyl, -L-heterocyclyl, -L-aryl, or -L-heteroaryl; each L is independently -O-, -NH-, -S-, -S(O)-, -S(O)2-, -N(C1-C6 alkyl)-, -N(C2-C6 alkenyl), -N(C2-C6 alkynyl)-, -N(C1-C6 haloalkyl)-, -N(C3-10 cycloalkyl)-, -N(heterocyclyl)-, -N(aryl)-, -N(heteroaryl)-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)N(C1-C6 alkyl)-, -C(O)N(C2-C6 alkenyl)-, -C(O)N(C2-C6 alkynyl)-, -C(O)N(C1-6 haloalkyl)-, -C(O)N(C3-10 cycloalkyl)-, -C(O)N(heterocyclyl)-, -C(O)N(aryl)-, -C(O)N(heteroaryl)-, -NHC(O)-, -NHC(O)O-, -NHC(O)NH-, -NHS(O)-, or -S(O)2NH-; and wherein each C1-C6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-C6 haloalkyl, C3-C10 cycloalkyl, heterocyclyl, aryl, and heteroaryl of Z1b and L is further independently unsubstituted or substituted with one to five halo, cyano, hydroxy, -SH, -NH2, -NO2, -SF5, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C3-C10 cycloalkyl, heterocyclyl, aryl, or heteroaryl, provided that the following compounds are excluded:
Figure imgf000126_0001
, .
Figure imgf000127_0001
13. The compound of claim 11 or claim 12, wherein the compound is represented by formula IB-1: or a pharmaceutically acceptable
Figure imgf000127_0002
salt, solvate, stereoisomer, or tautomer thereof.
14. The compound of claim 13, wherein each R4 independently is H or C1-C6 alkyl.
15. The compound of claim 14, wherein the compound is represented by formula IC-1:
Figure imgf000128_0001
or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof.
16. The compound of claim 15, wherein each R4 independently is H or methyl.
17. A pharmaceutical composition comprising a pharmaceutically acceptable excipient and an effective amount of a compound of claim 1.
18. A pharmaceutical composition comprising a pharmaceutically acceptable excipient and an effective amount of a compound of claim 10.
19 A pharmaceutical composition comprising a pharmaceutically acceptable excipient and an effective amount of a compound of any one of claims 2-9.
20 A pharmaceutical composition comprising a pharmaceutically acceptable excipient and an effective amount of a compound of any one of claims 11-16.
21. A compound or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, selected from Table 1.
22. A compound or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, selected from Table 1A.
23. A compound or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, selected from Table 2.
24. A compound or a pharmaceutically acceptable salt, solvate, stereoisomer, or tautomer thereof, selected from Table 2A.
25. A method for modulating or degrading BRD4 which method comprises contacting BRD4 with an effective amount of a compound according to claim 1.
26. A method for modulating or degrading BRD4 which method comprises contacting BRD4 with an effective amount of a compound according to claim 2 or claim 3 under conditions wherein BRD4 is modulated or degraded.
27. A method for modulating or degrading BRD4 in a subject, which method comprises administering to said subject an effective amount of a compound according to claim 2 or claim 3.
28. A method for modulating or degrading BRD4 which method comprises contacting BRD4 with an effective amount of a compound according to claim 10.
29. A method for modulating or degrading BRD4 which method comprises contacting BRD4 with an effective amount of a compound according to claim 11 or claim 12 under conditions wherein BRD4 is modulated or degraded.
30. A method for modulating or degrading BRD4 in a subject, which method comprises administering to said subject an effective amount of a compound according to claim 11 or claim 12.
31. A method for modulating or degrading BRD4 in a subject, which method comprises administering to said subject an effective amount of a pharmaceutical composition according to claim 19.
32. A method for modulating or degrading BRD4 in a subject, which method comprises administering to said subject an effective amount of a pharmaceutical composition according to claim 20.
33. A method for treating cancer in a subject in need thereof which method comprises administering to said subject an effective amount of a compound according to claim 2 or claim 3.
34. A method for treating cancer in a subject in need thereof which method comprises administering to said subject an effective amount of a pharmaceutical composition comprising a pharmaceutically acceptable excipient and an effective amount of a compound according to claim 2 or claim 3.
35. A method for treating cancer in a subject in need thereof which method comprises administering to said subject an effective amount of a compound according to claim 11 or claim 12.
36. A method for treating cancer in a subject in need thereof which method comprises administering to said subject an effective amount of a pharmaceutical composition comprising a pharmaceutically acceptable excipient and an effective amount of a compound according to claim 11 or claim 12.
PCT/US2023/017751 2022-04-08 2023-04-06 Propargyl compounds and pharmaceutical compositions that modulate brd4 WO2023196512A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202263329310P 2022-04-08 2022-04-08
US202263329308P 2022-04-08 2022-04-08
US63/329,310 2022-04-08
US63/329,308 2022-04-08

Publications (1)

Publication Number Publication Date
WO2023196512A1 true WO2023196512A1 (en) 2023-10-12

Family

ID=86387245

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/017751 WO2023196512A1 (en) 2022-04-08 2023-04-06 Propargyl compounds and pharmaceutical compositions that modulate brd4

Country Status (1)

Country Link
WO (1) WO2023196512A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5262564A (en) 1992-10-30 1993-11-16 Octamer, Inc. Sulfinic acid adducts of organo nitroso compounds useful as retroviral inactivating agents anti-retroviral agents and anti-tumor agents
WO2014140076A1 (en) * 2013-03-14 2014-09-18 Glaxosmithkline Intellectual Property (No.2) Limited 2,3-disubstituted 1 -acyl-4-amino-1,2,3,4-tetrahydroquinoline derivatives and their use as bromodomain inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5262564A (en) 1992-10-30 1993-11-16 Octamer, Inc. Sulfinic acid adducts of organo nitroso compounds useful as retroviral inactivating agents anti-retroviral agents and anti-tumor agents
WO2014140076A1 (en) * 2013-03-14 2014-09-18 Glaxosmithkline Intellectual Property (No.2) Limited 2,3-disubstituted 1 -acyl-4-amino-1,2,3,4-tetrahydroquinoline derivatives and their use as bromodomain inhibitors

Non-Patent Citations (25)

* Cited by examiner, † Cited by third party
Title
"Bioreversible Carriers in Drug Design", 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS
"Design of Prodrugs", 1985, ELSEVIER
"Fieser and Fieser's Reagents for Organic Synthesis", vol. 1, 2016, JOHN WILEY, AND SONS
"Larock's Comprehensive Organic Transformations", 1989, VCH PUBLISHERS INC.
"March's Advanced Organic Chemistry", vol. 1, 2019, JOHN WILEY, AND SONS
"Rodd's Chemistry of Carbon Compounds", vol. 1, 2001, ELSEVIER SCIENCE PUBLISHERS
ANGEWANDTE CHEMIE-INTERNATIONAL EDITION, vol. 59, 2020, pages 3028 - 3032
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 2005, JOHN WILEY AND SONS, INC.
COLIGAN ET AL.: "Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING COMPANY, article "The Pharmacological Basis of Therapeutics"
DUAN YINGCHAO ET AL: "Targeting Brd4 for cancer therapy: inhibitors and degraders", MEDCHEMCOMM, vol. 9, no. 11, 1 January 2018 (2018-01-01), United Kingdom, pages 1779 - 1802, XP055842447, ISSN: 2040-2503, DOI: 10.1039/C8MD00198G *
FOSTER: "Deuterium Isotope Effects in Studies of Drug Metabolism", TRENDS PHARMACOL. SCI., vol. 5, no. 12, 1984, pages 524 - 527, XP025943358, DOI: 10.1016/0165-6147(84)90534-0
J. ORG. CHEM., vol. 60, 1995, pages 7508
JEANMOUGIN F. ET AL., TRENDS BIOCHEM. SCI., vol. 22, no. 5, 1997, pages 151 - 153
JOURNAL OF GENERAL CHEMISTRY OF THE USSR, vol. 36, no. 8, 1966, pages 1448 - 1450
JOURNAL OF MEDICINAL CHEMISTRY, vol. 48, no. 25, 2005, pages 8045 - 8054
JOURNAL OF MEDICINAL CHEMISTRY, vol. 57, no. 19, 2014, pages 8111 - 8131
MULLER ET AL., EXPERT REV., vol. 13, no. 29, 2011, pages 1 - 20
PRINJHA ET AL., TRENDS PHARM. SCI., vol. 33, no. 3, 2012, pages 146 - 153
ROMAIN GOSMINI ET AL: "The Discovery of I-BET726 (GSK1324726A), a Potent Tetrahydroquinoline ApoA1 Up-Regulator and Selective BET Bromodomain Inhibitor", JOURNAL OF MEDICINAL CHEMISTRY, vol. 57, no. 19, 9 October 2014 (2014-10-09), US, pages 8111 - 8131, XP055221328, ISSN: 0022-2623, DOI: 10.1021/jm5010539 *
SAMBROOK ET AL.: "Molecular Cloning, A Laboratory Manual", 2000, COLD SPRING HARBOR PRESS
STRUHL K., GENES DEV., vol. 12, no. 5, 1989, pages 599 - 606
T. HIGUCHIV. STELLA: "A.C.S. Symposium Series", vol. 14, article "Pro-drugs as Novel Delivery Systems"
T. W. GREENEP. G. M. WUTS: "Protecting Groups in Organic Synthesis", 1999, WILEY
TAMKUN J. W. ET AL., CELL, vol. 7, no. 3, 1992, pages 561 - 572
TETRAHEDRON LETT., vol. 39, no. 19, pages 2933 - 293

Similar Documents

Publication Publication Date Title
AU2019200723B2 (en) 2-amino-6-fluoro-N-[5-fluoro-pyridin-3-yl]pyrazolo[1,5-a]pyrimidin-3-carboxamide compound useful as ATR kinase inhibitor, its preparation, different solid forms and radiolabelled derivatives thereof
EP2970323B1 (en) Furopyridines as bromodomain inhibitors.
ES2751741T3 (en) Procedure for making compounds useful as ATR kinase inhibitors
CN115141215B (en) KRAS G12D protein inhibitors and uses thereof
WO2020167994A1 (en) Compounds, compositions and methods
WO2023283428A1 (en) Heterocycloalkyl and heteroaryl compounds and pharmaceutical compositions that modulate ikzf2
JP6933384B2 (en) Heterocyclic compounds for the treatment of diseases
NZ713645A (en) Use of high acyl gellan in whipping cream
AU2022306297A1 (en) Aryl compounds and pharmaceutical compositions that modulate ikzf2
WO2023196512A1 (en) Propargyl compounds and pharmaceutical compositions that modulate brd4
AU2022202524B2 (en) Compounds, compositions and methods
WO2023283430A1 (en) Cycloalkyl compounds and pharmaceutical compositions that modulate ikzf2
CN117222654A (en) KRAS G12C inhibitors and uses thereof
AU2019424628B2 (en) 1,2,3,4-tetrahydroquinoxaline derivative, preparation method therefor and application thereof
RU2672725C2 (en) Method of preparing compound
WO2022221786A2 (en) Compounds and pharmaceutical compositions that modulate brd4
WO2023244806A1 (en) Compounds and pharmaceutical compositions that degrade swi/snf-related matrix-associated actin-dependent regulator of chromatin subfamily a
BR112019026096A2 (en) compound, medication, methods for positive allosteric modulation of the ch1 cholinergic muscarinic receptor and for the prophylaxis or treatment of constipation, and, use of the compound
EP4072670B1 (en) Quinazoline derivatives as lpa receptor 2 inhibitors
BR112019017931A2 (en) compound, pharmaceutical composition, combination, use of a compound, and method of treating a disease or condition.
CN114907387B (en) Pyrimido pyrrole KRAS inhibitor and preparation method and application thereof
CN117836288A (en) Aryl compounds and pharmaceutical compositions for modulating IKZF2
CN107406452B (en) Pyrazolo [3,4-d] pyrimidine derivatives and its purposes for treating leishmaniasis
WO2023208005A1 (en) Cyclic compounds, preparation methods and medicinal uses thereof
TW202320767A (en) Heteroaryl derivative PARP inhibitor and use thereof

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: 23724459

Country of ref document: EP

Kind code of ref document: A1