WO2023173057A1 - Mek inhibitors and uses thereof - Google Patents

Mek inhibitors and uses thereof Download PDF

Info

Publication number
WO2023173057A1
WO2023173057A1 PCT/US2023/064091 US2023064091W WO2023173057A1 WO 2023173057 A1 WO2023173057 A1 WO 2023173057A1 US 2023064091 W US2023064091 W US 2023064091W WO 2023173057 A1 WO2023173057 A1 WO 2023173057A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
cancer
compound
ring
membered
Prior art date
Application number
PCT/US2023/064091
Other languages
French (fr)
Inventor
Michael J. Burke
Sabine K. RUPPEL
Alfredo C. Castro
Sergio L. SANTILLANA SOTO
Eric HAINES
Lan Xu
Oksana ZAVIDIJ
Original Assignee
Ikena Oncology, 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 Ikena Oncology, Inc. filed Critical Ikena Oncology, Inc.
Publication of WO2023173057A1 publication Critical patent/WO2023173057A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems

Definitions

  • MEK INHIBITORS AND USES THEREOF CROSS REFERENCED TO RELATED APPLICATIONS This application claims the priority of US provisional application 63/269,140 filed March 10, 2022, US provisional application number 63/375,864 filed September 16, 2022, and US provisional application 63/479,118 filed January 9, 2023, each of which is hereby incorporated by reference in its entirety.
  • TECHNICAL FIELD [0002] The present disclosure relates to compounds and methods useful for inhibiting mitogen- activated protein kinase/extracellular signal-regulated kinase (MEK).
  • the disclosure also provides pharmaceutically acceptable compositions comprising compounds of the present disclosure and methods of using said compositions in the treatment of proliferative disorders, such as cancers.
  • MEK mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase (MEK)-ERK
  • ERK mitogen-activated protein kinase/extracellular signal-regulated kinase
  • MEK-ERK pathway is often activated by mutation of upstream factors, BRAF or Ras, or by the signals of constitutively activated cell-surface receptors.
  • Inhibition of MEK can be a promising strategy for controlling the growth of tumors, for example, the tumors associated with MEK pathway signaling.
  • SUMMARY [0004] It has now been found that compounds of the present disclosure, and pharmaceutically acceptable compositions thereof, are effective as MEK inhibitors.
  • the instant disclosure provides a compound of Formula (I): (I), or a pharmaceutically acceptable salt thereof, wherein each variable is as defined and described herein.
  • Another aspect of the disclosure provides a method of treating a disorder mediated by MEK in a subject. The method comprises administering a therapeutically effective amount of a compound described herein to a subject in need thereof to treat the disorder mediated by MEK, as further described in the detailed description.
  • Another aspect of the disclosure provides a method of inhibiting MEK activity. The method comprises contacting MEK or a KSR-MEK complex or a RAF-MEK complex with an effective amount of a compound described herein, as further described in the detailed description.
  • compounds of the present disclosure, and pharmaceutically acceptable compositions thereof are useful for treating proliferative disorders, such as the cancers as described herein.
  • DETAILED DESCRIPTION Compounds of the present disclosure, and pharmaceutical compositions thereof, are useful as MEK inhibitors. Without wishing to be bound by any particular theory, it is believed that the compounds of the present disclosure may directly engage kinase suppressor of Ras (KSR) at the MEK interface, and that KSR may remodel the prototypical allosteric pocket of the MEK inhibitor in the KSR-MEK complex. Without wishing to be bound by any particular theory, it is believed that the compounds of the present disclosure may bind the KSR-MEK complex and may disrupt the related RAF–MEK complex. 1.
  • KSR kinase suppressor of Ras
  • aliphatic or “aliphatic group”, as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as "carbocycle,” “cycloaliphatic” or “cycloalkyl”), that has a single point of attachment to the rest of the molecule.
  • aliphatic groups contain 1-6 aliphatic carbon atoms.
  • aliphatic groups contain 1-5 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms.
  • “cycloaliphatic” (or “carbocycle” or “cycloalkyl”) refers to a monocyclic C3-C6 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule.
  • Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl.
  • bicyclic ring or “bicyclic ring system” refers to any bicyclic ring system, i.e. , carbocyclic or heterocyclic, saturated or having one or more units of unsaturation, having one or more atoms in common between the two rings of the ring system.
  • the term includes any permissible ring fusion, such as ortho-fused or spirocyclic.
  • heterocyclic is a subset of “bicyclic” that requires that one or more heteroatoms are present in one or both rings of the bicycle.
  • Such heteroatoms may be present at ring junctions and are optionally substituted and may be selected from nitrogen (including N oxides) oxygen sulfur (including oxidized forms such as sulfones and sulfonates), phosphorus (including oxidized forms such as phosphates), boron, etc.
  • a bicyclic group has 7-12 ring members and 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • bridged bicyclic refers to any bicyclic ring system, i.e., carbocyclic or heterocyclic, saturated or partially unsaturated, having at least one bridge.
  • a “bridge” is an unbranched chain of atoms or an atom or a valence bond connecting two bridgeheads, where a “bridgehead” is any skeletal atom of the ring system which is bonded to three or more skeletal atoms (excluding hydrogen).
  • a bridged bicyclic group has 7-12 ring members and 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Such bridged bicyclic groups are well known in the art and include those groups set forth below where each group is attached to the rest of the molecule at any substitutable carbon or nitrogen atom.
  • a bridged bicyclic group is optionally substituted with one or more substituents as set forth for aliphatic groups. Additionally or alternatively, any substitutable nitrogen of a bridged bicyclic group is optionally substituted.
  • Exemplary bicyclic rings include: Exemplary bridged bicyclics include:
  • lower alkyl refers to a C 1-4 straight or branched alkyl group.
  • exemplary lower alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and tert-butyl.
  • lower haloalkyl refers to a C1-4 straight or branched alkyl group that is substituted with one or more halogen atoms.
  • heteroatom means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR + (as in N-substituted pyrrolidinyl)).
  • unsaturated as used herein, means that a moiety has one or more units of unsaturation.
  • bivalent C 1-8 (or C 1-6 ) saturated or unsaturated, straight or branched, hydrocarbon chain refers to bivalent alkylene, alkenylene, and alkynylene chains that are straight or branched as defined herein.
  • alkylene refers to a bivalent alkyl group.
  • An “alkylene chain” is a polymethylene group, i.e., –(CH 2 ) n –, wherein n is a positive integer, preferably from 1 to 6, from 1 to 4, from 1 to 3, from 1 to 2, or from 2 to 3.
  • a substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
  • alkenylene refers to a bivalent alkenyl group.
  • a substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
  • cyclopropylenyl refers to a bivalent cyclopropyl group of the following structure: .
  • halogen means F, Cl, Br, or I.
  • aryl used alone or as part of a larger moiety as in “aralkyl,” “aralkoxy,” or “aryloxyalkyl,” refers to monocyclic or bicyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members.
  • aryl may be used interchangeably with the term “aryl ring.”
  • aryl refers to an aromatic ring system which includes, but not limited to, phenyl, biphenyl, naphthyl, anthracyl and the like, which may bear one or more substituents.
  • aryl is a group in which an aromatic ring is fused to one or more non–aromatic rings, such as indanyl, phthalimidyl, naphthimidyl, phenanthridinyl, or tetrahydronaphthyl, and the like.
  • heteroaryl and “heteroar—,” used alone or as part of a larger moiety, e.g., “heteroaralkyl,” or “heteroaralkoxy,” refer to groups having 5 to 10 ring atoms, preferably 5, 6, or 9 ring atoms; having 6, 10, or 14 ⁇ electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms.
  • heteroatom refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen.
  • Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl.
  • heteroaryl and “heteroar—”, as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring.
  • Nonlimiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H–quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[2,3–b]–1,4–oxazin–3(4H)–one.
  • heteroaryl group may be mono– or bicyclic.
  • heteroaryl may be used interchangeably with the terms “heteroaryl ring,” “heteroaryl group,” or “heteroaromatic,” any of which terms include rings that are optionally substituted.
  • heteroarylkyl refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted.
  • heterocycle As used herein, the terms “heterocycle,” “heterocyclyl,” “heterocyclic radical,” and “heterocyclic ring” are used interchangeably and refer to a stable 5– to 7–membered monocyclic or 7–10–membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above.
  • nitrogen includes a substituted nitrogen.
  • the nitrogen may be N (as in 3,4–dihydro– 2H–pyrrolyl), NH (as in pyrrolidinyl), or + NR (as in N–substituted pyrrolidinyl).
  • a heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted.
  • saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenyl, pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and quinuclidinyl.
  • heterocycle used interchangeably herein, and also include groups in which a heterocyclyl ring is fused to one or more aryl, heteroaryl, or cycloaliphatic rings, such as indolinyl, 3H–indolyl, chromanyl, phenanthridinyl, or tetrahydroquinolinyl.
  • a heterocyclyl group may be mono– or bicyclic.
  • heterocyclylalkyl refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted.
  • partially unsaturated refers to a ring moiety that includes at least one double or triple bond.
  • partially unsaturated is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined.
  • compounds of the invention may contain “optionally substituted” moieties.
  • substituted means that one or more hydrogens of the designated moiety are replaced with a suitable substituent.
  • an “optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position.
  • Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds.
  • R * is C 1–6 aliphatic
  • R * is optionally substituted with halogen, – R ⁇ , -(haloR ⁇ ), -OH, –OR ⁇ , –O(haloR ⁇ ), –CN, –C(O)OH, –C(O)OR ⁇ , –NH 2 , –NHR ⁇ , –NR ⁇ 2, or – NO 2 , wherein each R ⁇ is independently selected from C 1–4 aliphatic, –CH 2 Ph, –O(CH 2 ) 0 –1Ph, or a 5–6–membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein each R ⁇ is unsubstituted or where preceded by halo is substituted only with one or more halogens.
  • An optional substituent on a substitutable nitrogen is independently –R ⁇ , –NR ⁇ 2 , – C(O)R ⁇ , –C(O)OR ⁇ , –C(O)C(O)R ⁇ , –C(O)CH 2 C(O)R ⁇ , -S(O) 2 R ⁇ , -S(O) 2 NR ⁇ 2, –C(S)NR ⁇ 2, – C(NH)NR ⁇ 2, or –N(R ⁇ )S(O) 2 R ⁇ ; wherein each R ⁇ is independently hydrogen, C 1–6 aliphatic, unsubstituted –OPh, or an unsubstituted 5–6–membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, two independent occurrences of R ⁇ , taken together with their intervening atom(s) form an unsubstit
  • alkyl refers to saturated monovalent hydrocarbon radicals having straight or branched hydrocarbon chains or, in the event that at least 3 carbon atoms are present, cyclic hydrocarbons or combinations thereof and contains 1 to 20 carbon atoms (C 1 -C 20 alkyl), 1 to 10 carbon atoms (C 1 -C 10 alkyl), 1 to 8 carbon atoms (C 1 - C 8 alkyl), 1 to 6 carbon atoms (C 1 -C 4 alkyl), or 1 to 4 carbon atoms (C 1 -C 4 alkyl).
  • alkyl radicals include methyl, ethyl, propyl, isopropyl, n- butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isoamyl, hexyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like.
  • alkoxy refers to —O-(alkyl), wherein “alkyl” is as defined above.
  • cycloalkyl as used alone or in combination, means a saturated or partially saturated monocyclic, bicyclic or tricyclic alkyl radical wherein each cyclic moiety contains from 3 to 8 carbon atoms, or from 3 to 6 carbon atoms.
  • cycloalkyl radicals include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like.
  • halogen or halo is F, Cl, Br, or I.
  • the term “pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1–19, incorporated herein by reference.
  • Pharmaceutically acceptable salts of the compounds of this disclosure include those derived from suitable inorganic and organic acids and bases.
  • Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid
  • organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2– hydroxy–ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2–naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pec
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (C 1 -C4alkyl)4 salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
  • structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the disclosure. Unless otherwise stated, all tautomeric forms of the compounds of the disclosure are within the scope of the disclosure.
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13 C- or 14 C-enriched carbon are within the scope of this disclosure.
  • Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present disclosure.
  • the term “provided compound” refers to any MEK inhibitor genus, subgenus, and/or species set forth herein.
  • the terms “inhibitor” or “MEK inhibitor” or “MEK antagonist” are defined as a compound that binds to and/or inhibits MEK with measurable affinity. In some embodiments, inhibition in the presence of a MEK inhibitor or a MEK antagonist is observed in a dose-dependent manner.
  • the measured signal (e.g., signaling activity or biological activity) is at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, or at least about 100% lower than the signal measured with a negative control under comparable conditions.
  • an inhibitor has an IC50 and/or binding constant of less than about 100 ⁇ M, less than about 50 ⁇ M, less than about 1 ⁇ M, less than about 500 nM, less than about 100 nM, less than about 10 nM, or less than about 1 nM.
  • measurable affinity and “measurably inhibit,” as used herein, means a measurable change or inhibition in MEK activity between a sample comprising a compound of the present disclosure, or composition thereof, and MEK, and an equivalent sample comprising MEK, in the absence of said compound, or composition thereof.
  • effective amount refers to the amount of a compound sufficient to effect beneficial or desired results (e.g., a therapeutic, ameliorative, inhibitory, or preventative result). An effective amount can be administered in one or more administrations, applications, or dosages and is not intended to be limited to a particular formulation or administration route.
  • treating includes any effect, e.g., lessening, reducing, modulating, ameliorating or eliminating, that results in the improvement of the condition, disease, disorder, and the like, or ameliorating a symptom thereof.
  • treatment can be administered after one or more symptoms have developed.
  • treatment can be administered in the absence of symptoms.
  • treatment can be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment can also be continued after symptoms have resolved, for example, to prevent or delay their recurrence.
  • a non-limiting example of a MEK-associated disease or disorder is a MEK-associated cancer.
  • the term “pharmaceutical composition” refers to the combination of an active agent with a carrier, inert or active, making the composition especially suitable for diagnostic or therapeutic use in vivo or ex vivo.
  • the term “pharmaceutically acceptable carrier” refers to any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, emulsions (e.g., such as an oil/water or water/oil emulsions), and various types of wetting agents.
  • the compositions also can include stabilizers and preservatives.
  • stabilizers and adjuvants see e.g., Martin, Remington’s Pharmaceutical Sciences, 15th Ed., Mack Publ. Co., Easton, PA [1975].
  • a “non-ATP competitive” or “ATP non-competitive” MEK inhibitor refers to an inhibitor of MEK that does not bind in the ATP pocket of MEK, or does not displace ATP from the MEK active site, and can form direct contacts when co-bound to the MEK-ATP complex.
  • Non-ATP competitive inhibition by a compound of the invention can be confirmed by art-recognized methods such as enzymology studies, competition assays, biophysical methods, including X-ray co-crystallography.
  • An exemplary non-ATP competitive inhibitor of the invention inhibits recombinant MEK1 or MEK2 with an IC50 of from about 1 nM to about 50 ⁇ M.
  • an exemplary non-ATP competitive inhibitor of the invention inhibits recombinant MEK1 or MEK2 with an IC 50 of about 1 nM to about 1000 nM, about 1 ⁇ M to about 50 ⁇ M, about 1 ⁇ M to about 25 ⁇ M, about 1 ⁇ M to about 10 ⁇ M, about 500 nM to about 5 ⁇ M, or about 10 nM to about 500 nM.
  • binding refers to when an inhibitor of the present disclosure binds to and stabilizes the CRAF-MEK complex in an inactive conformation.
  • a compound of the invention “allosterically binds an inhibitor pocket” when a compound binds outside the active site, including, for example, outside or adjacent to the ATP-binding site of a kinase.
  • an “inhibitor-inhibitor pocket complex” describes a species in which an inhibitor of the invention allosterically binds an inhibitor pocket formed at an interaction interface between human MEK (MEK1 or MEK2) and human Kinase Suppressor of Ras (KSR1, KSR2, or the KSR homolog BRAF or CRAF) adjacent to ATP in a physiological complex between MEK and KSR.
  • a 4 Angstrom contact is reasonable based on the nature of the trametinib-KSR interaction and precedence of known drug-receptor complexes. This contact is within the range of known contacts as defined by several independent groups (PMID 29308120, 26517868, 19221587). 2.
  • Ring A is an optionally substituted ring selected from a phenyl ring, a 5-6 membered heteroaromatic ring having 1-4 heteroatoms independently selected from N, O, or S, or a 8-11 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from N, O, or S.
  • Ring A is an optionally substituted ring selected from a phenyl ring, a 5 or 6 membered heteroaromatic ring having 1, 2, 3, or 4 heteroatoms independently selected from N, O, or S, or a 8, 9, 10, or 11 membered bicyclic heteroaromatic ring having 1, 2, 3, 4, or 5 heteroatoms independently selected from N, O, or S.
  • Ring A is an optionally substituted ring selected from a phenyl ring or a 6 membered heteroaromatic ring having 1-4 heteroatoms independently selected from N. In some embodiments, Ring A is an optionally substituted ring selected from a phenyl ring or a 6 membered heteroaromatic ring having 1, 2, 3, or 4 heteroatoms independently selected from N. In some embodiments, Ring A is an optionally substituted ring selected form a phenyl ring or a 5 or 6 membered heteroaromatic having 1, 2, 3, or 4 heteroatoms independent selected from N. In some embodiments, Ring A is pyridinyl, pyrimidinyl, pyrazinyl, or pyrazolyl.
  • Ring is independently halogen, R, -OR, or -N(R) 2 ; and R 8 is halogen, R, -OR, -N(R) 2 , each of R 10 and R 11 is independently H, optionally substituted C1-6 aliphatic, optionally substituted 3-8 membered carbocyclyl, optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S, optionally substituted phenyl, or optionally substituted 5-6 membered heteroaryl having 1-4 heteroatoms independently selected from N, O, or S, or R 10 and R 11 taken together with the N atom to which they attach, form optionally substituted 3-8 membered heterocyclyl having 0, 1, or 2 heteroatoms independently selected from N, O, or S in addition to the N atom to which R 10 and R 11 attach; and R 12 is H, optionally substituted C 1 -C 6 aliphatic, optionally substituted 3-8 membere
  • each R 7 is independently halogen, R, or -OR. In some embodiments, R 7 is independently H, or optionally substituted C 1 -C 6 aliphatic. In some embodiments, R 7 is independently H, halogen, or C 1 -C 6 aliphatic. In some embodiments, R 7 is unsubstituted C 1 -C 6 alkoxy, or optionally substituted C 1 -C 6 alkoxy. In some embodiments, each R 7 is independently halogen. In some embodiments, R 7 is independently H or fluoro. In some embodiments, R 7 is fluoro. In some embodiments, R 7 is H. [0058] In some embodiments, R 8 is , , or .
  • R 8 is , , or ; each of R 10 and R 11 is independently H, C 1 -C 6 aliphatic, or 3-8 membered heterocyclyl having 1, 2, or 3 N atoms; and R 12 is H, optionally substituted C 1 -C 6 aliphatic, or optionally substituted 3-8 membered carbocyclyl.
  • R 8 is unsubstituted C 1 -C 6 alkoxy, or optionally substituted C 1 -C 6 alkoxy.
  • each of R 10 and R 11 is independently hydrogen or C 1 -C 6 alkyl.
  • R 12 is hydrogen or C 1 -C 6 alkyl.
  • R 12 is C 1 -C 6 alkyl. In some embodiments, R 12 is hydrogen, methyl, ethyl, propyl, butyl, or hexyl.
  • Ring B is an optionally substituted ring selected from a phenyl ring, a 5-6 membered heteroaromatic ring having 1-4 heteroatoms independently selected from N, O, or S, or a 8-11 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from N, O, or S.
  • Ring B is an optionally substituted ring selected from a phenyl ring or a 6 membered heteroaromatic ring having 1-4 N atoms. In some embodiments, Ring B is optionally substituted pyridinyl or optionally substituted pyrimidinyl. In some embodiments, Ring B is , , , or , and R 1 is halogen, R, -OR, or -N(R) 2 . In some embodiments, R 1 is hydrogen or halogen. In some embodiments, R 1 is fluoro. In some embodiments, R 1 is R. In some embodiments, R 1 is unsubstituted, or optionally substituted C 1 -C 6 alkyl.
  • R 1 is unsubstituted C 1 -C 6 alkoxy, or optionally substituted C 1 -C 6 alkoxy.
  • X 1 is C(R 21 ) or N. In some embodiments, X 1 is C(R 21 ). In some embodiments, X 1 is N. In some embodiments, X 1 is C(H).
  • X 2 is C(R 22 ) or N. In some embodiments, X 2 is C(R 22 ). In some embodiments, X 2 is N. In some embodiments, X 2 is C(H). In some embodiments, X 2 is C(R 22 ), wherein R 22 is halogen.
  • X 3 is C(R 23 ) or N. In some embodiments, X 3 is C(R 23 ). In some embodiments, X 3 is N. In some embodiments, X 3 is C(H). In some embodiments, X 3 is C(R 23 ), wherein R 23 is halogen. [0067] In some embodiments, R 21 is halogen. In some embodiments, R 21 is R. In some embodiments, R 21 is H. In some embodiments, R 21 is optionally substituted C 1 -C 6 aliphatic. In some embodiments, R 21 is optionally substituted C 1 -C 6 alkyl. In some embodiments, R 21 is -OR.
  • R 21 is optionally substituted C 1 -C 6 alkoxy. In some embodiments, R 21 is - N(R) 2 . In some embodiments, R 21 is -N(R)-C(O)-R. In some embodiments, R 21 is -C(O)-N(R) 2 . [0068] In some embodiments, R 22 is halogen. In some embodiments, R 22 is R. In some embodiments, R 22 is H. In some embodiments, R 22 is optionally substituted C 1 -C 6 aliphatic. In some embodiments, R 22 is optionally substituted C 1 -C 6 alkyl. In some embodiments, R 22 is -OR.
  • R 22 is optionally substituted C 1 -C 6 alkoxy. In some embodiments, R 22 is - N(R) 2 . In some embodiments, R 22 is -N(R)-C(O)-R. In some embodiments, R 22 is -C(O)-N(R) 2 . [0069] In some embodiments, R 23 is halogen. In some embodiments, R 23 is R. In some embodiments, R 23 is H. In some embodiments, R 23 is optionally substituted C 1 -C 6 aliphatic. In some embodiments, R 23 is optionally substituted C 1 -C 6 alkyl. In some embodiments, R 23 is -OR.
  • R 23 is optionally substituted C 1 -C 6 alkoxy. In some embodiments, R 23 is - N(R) 2 . In some embodiments, R 23 is -N(R)-C(O)-R. In some embodiments, R 23 is -C(O)-N(R) 2 .
  • R 4 is unsubstituted C 1 -C 6 alkoxy, or optionally substituted C 1 -C 6 alkoxy.
  • R 3 and R 4 are independently hydrogen, -CH 3 , -CH 2 N(CH 3 ) 2 , or –(C 3 H 5 ).
  • R 3 and R 4 are independently R; and each R is independently H, optionally substituted C 1-6 aliphatic, or optionally substituted 3-8 membered carbocyclyl. In some embodiments, R 3 and R 4 are independently R; and each R is independently H, methyl, or cyclopropyl. In some embodiments, R 3 and R 4 are independently hydrogen, -CH 3 , or -CH 2 N(CH 3 ) 2 . In some embodiments, R 3 and R 4 are independently R; and each R is independently C 1 -C 6 aliphatic. In some embodiments, R 3 and R 4 are independently hydrogen or CH3.
  • R 3 is -T-N(R) 2 , wherein T is a bivalent C 1 -8 saturated or unsaturated, straight or branched, hydrocarbon chain wherein one, two or three methylene units of the chain are optionally and independently replaced by -Cy-, –C(R) 2 –, –S–, –N(R)–, –O–, –C(O)– , –OC(O)–, –C(O)O–, –C(O)N(R)–, –N(R)C(O)–, –S(O)–, –S(O) 2 – or –N(R)CH2C(O)–; and -Cy- is an optionally substituted 3-8 membered bivalent carbocyclic ring, or an optionally substituted 3-8 membered bivalent heterocyclic ring having 1, 2, or 3 heteroatoms independently selected from N, O, or S.
  • T is a bivalent C 1 -8 saturated or
  • R 4 is -T-N(R) 2 , wherein T is a bivalent C 1-8 saturated or unsaturated, straight or branched, hydrocarbon chain wherein one, two or three methylene units of the chain are optionally and independently replaced by -Cy-, –C(R) 2 –, –S–, –N(R)–, –O–, –C(O)– , –OC(O)–, –C(O)O–, –C(O)N(R)–, –N(R)C(O)–, –S(O)–, –S(O) 2 – or –N(R)CH2C(O)–; and -Cy- is an optionally substituted 3-8 membered bivalent carbocyclic ring, or an optionally substituted 3-8 membered bivalent heterocyclic ring having 1, 2, or 3 heteroatoms independently selected from N, O, or S.
  • T is a bivalent C 1-8 saturated or unsatur
  • L 1 is -C(R 5 )(R 6 )-, wherein each of R 5 and R 6 is independently halogen, R, -OR, or -N(R) 2 ; or R 5 and R 6 , together with the atom to which they attach, form an optionally substituted 3-8 membered carbocyclic ring or an optionally substituted 3-8 membered heterocyclic ring having 1-3 heteroatoms independently selected from N, O, or S.
  • R 5 is hydrogen
  • R 6 is R.
  • R 6 is hydrogen.
  • R 5 and R 6 are independently optionally substituted C 1 -C 6 aliphatic.
  • R 5 and R 6 are independently methyl, ethyl, propyl, butyl, or hexyl. In some embodiments, R 5 is hydrogen, and R 6 is methyl. In some embodiments, R 5 and R 6 are independently halogen or R. In some embodiments, R 5 and R 6 are independently hydrogen, halogen, or C 1 -C 6 aliphatic. [0075] In some embodiments, L 1 is -CH 2 -. In some embodiments, L 1 is -C(H)(CH 3 )-. [0076] In some embodiments, L 1 is -NR-, wherein R is as defined and described in embodiments herein. In some embodiments, L 1 is -NH-.
  • L 1 is -N(CH3)-.
  • each R is independently H, optionally substituted C 1 -C 6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S.
  • R is H.
  • R is unsubstituted, or optionally substituted C 1 -C 6 aliphatic.
  • R is unsubstituted, or optionally substituted C 1 -C 6 alkyl.
  • R is unsubstituted, or optionally substituted 3-8 membered carbocyclyl.
  • R is unsubstituted, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S.
  • the present disclosure provides a compound of Formula (IA): or a pharmaceutically acceptable salt thereof, wherein each variable is as defined above and described in embodiments herein, both singly and in combination.
  • the present disclosure provides a compound of Formula (IB): (IB) or a pharmaceutically acceptable salt thereof, wherein each variable is as defined above and described in embodiments herein, both singly and in combination.
  • the present disclosure provides a compound of Formula (IC): or a pharmaceutically acceptable salt thereof, wherein each variable is as defined above and described in embodiments herein, both singly and in combination.
  • the present disclosure provides a compound of Formula (ID): or a pharmaceutically acceptable salt thereof, wherein each variable is as defined above and described in embodiments herein, both singly and in combination.
  • the present disclosure provides a compound of Formula (IC-1), or a pharmaceutically acceptable salt thereof, wherein: R 1 is H or halogen; each of R 3 and R 4 is independently H or optionally substituted C 1 -C 6 aliphatic; R 22 is H or halogen; R 7 is H or halogen; each of R 10 and R 11 is independently H or optionally substituted C 1 -C 6 aliphatic; and R 12 is H or optionally substituted C 1 -C 6 aliphatic.
  • R 1 is H or halogen
  • each of R 3 and R 4 is independently H or optionally substituted C 1 -C 6 aliphatic
  • R 22 is H or halogen
  • R 7 is H or halogen
  • each of R 10 and R 11 is independently H or optionally substituted C 1 -C 6 aliphatic
  • R 12 is H or optionally substituted C 1 -C 6 aliphatic.
  • the present disclosure provides a compound of Formula (IC-2), or a pharmaceutically acceptable salt thereof, wherein: R 1 is H or halogen; R 3 is H or optionally substituted C 1 -C 6 alkyl; R 22 is H or halogen; R 7 is halogen; and each of R 10 and R 11 is independently H or optionally substituted C 1 -C 6 alkyl.
  • the present disclosure provides a compound of Formula (IC-3), or a pharmaceutically acceptable salt thereof, wherein: R 3 is H or optionally substituted C 1 -C 6 alkyl; R 22 is H or halogen; and each of R 10 and R 11 is independently H or optionally substituted C 1 -C 6 alkyl.
  • the present disclosure provides a compound of Formula (IC-4), or a pharmaceutically acceptable salt thereof, wherein: each of R 3 and R 4 is independently H or optionally substituted C 1 -C 6 aliphatic; R 22 is H or halogen; and each of R 10 and R 11 is independently H or optionally substituted C 1 -C 6 aliphatic; and R 12 is H or optionally substituted C 1 -C 6 aliphatic.
  • the present disclosure provides a compound of Formula (IC-5), or a pharmaceutically acceptable salt thereof, wherein: R 1 is H or halogen; each of R 3 and R 4 is independently H or optionally substituted C 1 -C 6 aliphatic; R 22 is H or halogen; and R 7 is H or halogen.
  • IE compound of Formula
  • the present disclosure provides a compound of Formula (IF): or a pharmaceutically acceptable salt thereof, wherein each variable is as defined above and described in embodiments herein, both singly and in combination.
  • the present disclosure provides a compound of Formula (II): or a pharmaceutically acceptable salt thereof, wherein W is S or S(O) 2 , and each of X 1 , X 2 , X 3 , R 3 , R 4 , L 1 , Ring A and Ring B is as defined above and described in embodiments herein, both singly and in combination.
  • W is S or S(O) 2
  • each of X 1 , X 2 , X 3 , R 3 , R 4 , L 1 , Ring A and Ring B is as defined above and described in embodiments herein, both singly and in combination.
  • the present disclosure provides a compound set forth in Table 1 above, or a pharmaceutically acceptable salt thereof. 3. Uses, Formulation, and Administration: Pharmaceutically acceptable compositions [0098] According to another embodiment, the disclosure provides a pharmaceutical composition comprising a compound of this disclosure or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle. The amount of compound in compositions of this disclosure is such that is effective to measurably inhibit MEK, or a variant or mutant thereof, in a biological sample or in a patient. In certain embodiments, the amount of compound in compositions of this disclosure is such that is effective to measurably inhibit MEK, or a variant or mutant thereof, in a biological sample or in a patient.
  • a composition of this disclosure is formulated for administration to a patient in need of such composition. In some embodiments, a composition of this disclosure is formulated for oral administration to a patient.
  • the terms “subject” and “patient” are used interchangeably and refer to organisms to be treated by the methods of the present disclosure. Such organisms preferably include, but are not limited to, mammals (e.g., murines, simians, equines, bovines, porcines, canines, felines, and the like), and, most preferably, includes humans.
  • compositions of this disclosure refers to a non- toxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated.
  • Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of this disclosure include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropy
  • a “pharmaceutically acceptable derivative” means any non-toxic salt, ester, salt of an ester or other derivative of a compound of this disclosure that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this disclosure or an active metabolite or residue thereof.
  • active metabolite or residue thereof means that a metabolite or residue thereof also inhibits MEK, or a variant or mutant thereof.
  • Compositions of the present disclosure can be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • the compositions are administered orally, intraperitoneally or intravenously.
  • Sterile injectable forms of the compositions of this disclosure may be aqueous or oleaginous suspension. These suspensions can be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • a non-toxic parenterally acceptable diluent or solvent for example as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that can be employed are water, Ringer’s solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or di- glycerides.
  • Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions can also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • a long-chain alcohol diluent or dispersant such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • Other commonly used surfactants such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
  • compositions of this disclosure can be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions.
  • carriers commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents can also be added.
  • pharmaceutically acceptable compositions of this disclosure can be administered in the form of suppositories for rectal administration.
  • compositions of this disclosure can also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
  • Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches can also be used.
  • compositions can be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers.
  • Carriers for topical administration of compounds of this disclosure include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • provided pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • provided pharmaceutically acceptable compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride.
  • the pharmaceutically acceptable compositions can be formulated in an ointment such as petrolatum.
  • compositions of this disclosure can also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well- known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents. [00112] Most preferably, pharmaceutically acceptable compositions of this disclosure are formulated for oral administration. Such formulations may be administered with or without food. In some embodiments, pharmaceutically acceptable compositions of this disclosure are administered without food. In other embodiments, pharmaceutically acceptable compositions of this disclosure are administered with food.
  • the pharmaceutical compositions of this disclosure are brain- penetrant or CNS-penetrant or provide brain exposure.
  • the terms “brain-penetrant, “CNS-penetrant, “ or “brain exposure” refers that the compounds and pharmaceutical compositions of this disclosure are capable of crossing the blood brain barrier (BBB), and thus are useful for treating a brain or CNS disease, condition, injury or disorder.
  • a brain or CNS disease, condition, injury or disorder is a neurodegenerative diseases, neuronal injury, stroke, genetic disorders, psychiatric disorders, developmental disorders, inflammation, infection or damage, and brain cancers, spinal cord injury (SCI) and traumatic brain injury (TBI).
  • a brain disorder is selected from epilepsy, meningitis, encephalitis including HIV Encephalitis, progressive multifocal leukoencephalopathy, neuromyelitis optica, multiple sclerosis, late-stage neurological trypanosomiasis, amyotrophic lateral sclerosis (ALS), progressive bulbar palsy (PBP), primary lateral sclerosis (PLS), progressive muscular atrophy (PMA), Alzheimer's disease, Parkinson's disease, Huntington's disease, De Vivo disease, and any type of tumor, cancer or hyperproliferative disease in the brain or CNS.
  • epilepsy meningitis
  • encephalitis including HIV Encephalitis
  • progressive multifocal leukoencephalopathy progressive multifocal leukoencephalopathy
  • neuromyelitis optica multiple sclerosis
  • late-stage neurological trypanosomiasis amyotrophic lateral sclerosis (ALS), progressive bulbar palsy (PBP), primary lateral sclerosis (PLS), progressive muscular atrophy (
  • a brain or CNS disease, condition, injury or disorder is a neurological disorder which affects the CNS and/or which has an etiology in the CNS, which includes, but is not limited to, neuropathy, amyloidosis, cancer, an ocular disease or disorder, viral or microbial infection, inflammation, ischemia, neurodegenerative disease, seizure, behavioral disorders, and a lysosomal storage disease.
  • the CNS will be understood to include the eye, which is normally sequestered from the rest of the body by the blood-retina barrier.
  • neurological disorders include, but are not limited to, neurodegenerative diseases (including, but not limited to, Lewy body disease, postpoliomyelitis syndrome, Shy-Draeger syndrome, olivopontocerebellar atrophy, Parkinson's disease, multiple system atrophy, striatonigral degeneration, tauopathies (including, but not limited to, Alzheimer disease and supranuclear palsy), prion diseases (including, but not limited to, bovine spongiform encephalopathy, scrapie, Creutzfeldt- Jakob syndrome, kuru, Gerstmann-Straussler-Scheinker disease, chronic wasting disease, and fatal familial insomnia), bulbar palsy, motor neuron disease, and nervous system hetero degenerative disorders (including, but not limited to, Canavan disease, Huntington's disease, neuronal ceroid- lipofuscinosis, Alexander's disease, Tourette's syndrome, Menkes kinky hair syndrome, Cockayne syndrome, Halervorden-Spatz syndrome, la
  • the “central nervous system” or “CNS” refers to the complex of nerve tissues that control bodily function, and includes the brain and spinal cord.
  • the amount of compounds of the present disclosure that can be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration. Preferably, provided compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions.
  • a specific dosage and treatment regimen for any particular patient depends upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated.
  • the amount of a compound of the present disclosure in the composition also depends upon the particular compound in the composition.
  • Uses of Compounds and Pharmaceutically Acceptable Compositions [00118]
  • the present disclosure provides a method of using a compound as described herein for treating a disease or disorder associated with MEK.
  • a disease or disorder associated with MEK is a proliferative disorder.
  • a disease or disorder associated with MEK is a cancer. In some embodiments, a disease or disorder associated with MEK is a cancer as described herein. [00119] In some aspects and embodiments, provided herein are methods of treating, reducing the severity of, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more symptoms thereof, of a disease or disorder characterized by or associated with increased MEK expression and/or increased MEK activity, comprising the step of administering to a patient in need thereof a therapeutically effective amount of a compound of the present disclosure, or pharmaceutically acceptable composition thereof.
  • provided herein are methods of treating, reducing the severity of, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more symptoms thereof of a disease or disorder in which inhibition or antagonizing of MEK activity is beneficial, comprising the step of administering to a patient in need thereof a therapeutically effective amount of a compound of the present disclosure, or pharmaceutically acceptable composition thereof.
  • the present disclosure provides a method for treating one or more disorders, diseases, and/or conditions wherein the disorder, disease, or condition includes, but is not limited to, a cellular proliferative disorder, comprising administering to a patient in need thereof, a MEK inhibitor compound as described herein, or a pharmaceutical salt or composition thereof.
  • the cellular proliferative disorder is cancer.
  • the cancer is characterized by increased MEK expression and/or increased MEK activity, i.e., “increased activated MEK.”
  • increased MEK expression and/or increased MEK activity i.e., “increased activated MEK.”
  • an increase can be by at least about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, about 100%, about 2-fold, about 3-fold, about 4- fold, about 5-fold, about 6-fold, about 7-fold, about 8-fold, about 9-fold, about 10-fold, about 20- fold, about 25-fold, about 50-fold, about 100-fold, or higher, relative to a control or baseline amount of a function, or activity, or concentration.
  • the terms “increased expression” and/or “increased activity” of a substance, such as MEK, in a sample or cancer or patient refers to an increase in the amount of the substance, such as MEK, of about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, about 100%, about 2-fold, about 3-fold, about 4-fold, about 5-fold, about 6-fold, about 7-fold, about 8-fold, about 9-fold, about 10-fold, about 20-fold, about 25-fold, about 50- fold, about 100-fold, or higher, relative to the amount of the substance, such as MEK, in a control sample or control samples, such as an individual or group of individuals who are not suffering from the disease or disorder (e.g.,
  • a subject can also be determined to have an "increased expression” or “increased activity” of MEK if the expression and/or activity of MEK is increased by one standard deviation, two standard deviations, three standard deviations, four standard deviations, five standard deviations, or more, relative to the mean (average) or median amount of MEK in a control group of samples or a baseline group of samples or a retrospective analysis of patient samples.
  • control or baseline expression levels can be previously determined, or measured prior to the measurement in the sample or cancer or subject, or can be obtained from a database of such control samples.
  • the present disclosure provides a method for treating or preventing or reducing the risk of a cancer in patient comprising administering to the patient a compound of the disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
  • a "cancer,” as used herein, refers a broad group of various diseases characterized by the uncontrolled growth of abnormal cells in the body. Unregulated cell division and growth divide and grow results in the formation of malignant tumors that invade neighboring tissues and can also metastasize to distant parts of the body through the lymphatic system or bloodstream.
  • the cancer or proliferative disorder or tumor to be treated using the compounds and methods and uses described herein include, but are not limited to, a hematological cancer, a lymphoma, a myeloma, a leukemia, a neurological cancer, skin cancer, breast cancer, a prostate cancer, a colorectal cancer, lung cancer, head and neck cancer, a gastrointestinal cancer, a liver cancer, a pancreatic cancer, a genitourinary cancer, a bone cancer, renal cancer, and a vascular cancer.
  • the cancer is a K-Ras mutant cancer.
  • the K-Ras mutant cancer is an activated mutant K-Ras cancer.
  • the K-Ras mutant cancer is a cancer having a mutant or variant K-Ras G12.
  • the mutant or variant K-Ras G12 is K-Ras G12D, K-Ras G12V, K-Ras G12C, K-Ras G12R, K-Ras G12A, or any combination thereof.
  • the mutant or variant K-Ras G12 is K-Ras G12D.
  • the mutant or variant K-Ras G12 is K-Ras G12V.
  • the mutant or variant K-Ras G12 is K-Ras G12C.
  • the mutant or variant K- Ras G12 is K-Ras G12R. In some embodiments, the mutant or variant K-Ras G12 is K-Ras G12A. In some embodiments, the K-Ras mutant cancer is a cancer having a mutant or variant K-Ras G13. In some embodiments, the mutant or variant K-Ras G13 is K-Ras G13D, K-Ras G13C, or any combination thereof. In some embodiments, the K-Ras mutant cancer is a cancer having a mutant or variant K-Ras Q61. In some embodiments, the mutant or variant K-Ras Q61 is K-Ras Q61H, K-Ras Q61R, or any combination thereof.
  • the K-Ras mutant cancer is a cancer having a mutant or variant K-Ras A146.
  • the mutant or variant K- Ras A146 is K-Ras A146T.
  • the K-Ras mutant cancer is a cancer having a mutant or variant K-Ras G12D, K-Ras G12V, K-Ras G12C, K-Ras G12R, K-Ras G12A, K-Ras G13D, K-Ras G13C, K-Ras Q61H, K-Ras Q61R, K-Ras A146T, or any combination thereof.
  • the cancer is a mutant B-Raf cancer.
  • the mutant B-Raf cancer comprises a B-Raf amplification.
  • a “B-Raf amplification” refers to a cancer or cancer cell comprising at least three B-Raf copies, at least four B-Raf copies, at least five B-Raf copies, at least six B-Raf copies, at least seven B-Raf copies, at least eight B- Raf copies, at least nine B-Raf copies, at least ten B-Raf copies, or more.
  • the mutant B-Raf cancer is a cancer having a mutant or variant B-Raf V600, also referred to as a “Class I B-Raf mutation.”
  • the mutant or variant B-Raf V600 is B-Raf V600E, V600K, V600D, V600R, or any combination thereof.
  • the mutant B-Raf cancer is a cancer having a mutant or variant B-Raf K601, B-Raf P367, B-Raf G464, B-Raf L485, B-Raf E586, B-Raf T588, B-Raf T599, B-Raf L597, B-Raf G469, or any combination thereof, which are also collectively referred to as “Class II B-Raf mutations.”
  • the mutant B-Raf K601 is B-Raf K601E, K601N, K601T, or any combination thereof.
  • the mutant B-Raf P367 is B-Raf P367L, P367S, or any combination thereof.
  • the mutant B-Raf G464 is B-Raf G464V, G464E, or any combination thereof.
  • the mutant B-Raf L485 is L485W.
  • the mutant B-Raf E586 is E586K.
  • the mutant B-Raf T599 is T588TT, T588TS, T599I, T599K, or any combination thereof.
  • the mutant B-Raf L597 is B-Raf L597Q, L597R, L597S, L597V, or any combination thereof.
  • the mutant B-Raf G469 is B-Raf G469A, G469V, G469R, or any combination thereof.
  • the mutant B-Raf cancer is a cancer having a mutant or variant B- Raf D287, B-Raf V459, B-Raf G466, B-Raf S467, B-Raf G469, B-Raf N581, B-Raf D595, B-Raf F595, B-Raf G596, or any combination thereof, which are also collectively referred to as “Class III B-Raf mutations.”
  • the mutant B-Raf D287 is B-Raf D287N.
  • the mutant B-Raf V459 is B-Raf B459L.
  • the mutant B-Raf G466 is B-Raf G466A, B-Raf G466E, B-Raf G466V, or any combination thereof.
  • the mutant B-Raf S467 is B-Raf S467L.
  • the mutant B-Raf G469 is B-Raf-G469E.
  • the mutant B-Raf N581 is B-Raf N581I, B-Raf N581S, B-Raf N581T, or any combination thereof.
  • the mutant B-Raf D595 is B-Raf D595A, B-Raf D595G, B-Raf D595H, B-Raf D595N, or any combination thereof.
  • the mutant B-Raf F595 is B-Raf F595L.
  • the mutant B- Raf G596 is B-Raf G596D, B-Raf G596R, or any combination thereof.
  • the mutant B-Raf cancer comprises a B-Raf amplification and at least one B-Raf mutant or variant B- Raf Class I, Class II, or Class III mutation.
  • the cancer is an N-Ras mutant cancer.
  • the N-Ras mutant cancer is an activated mutant N-Ras cancer.
  • the N-Ras mutant cancer is a cancer having a mutant or variant N-Ras G12.
  • the mutant or variant N-Ras G12 is N-Ras G12D, N-Ras G12V, N-Ras G12C, N-Ras G12A, N-Ras G12R, N-Ras G12S, or any combination thereof.
  • the mutant or variant N-Ras G12 is N-Ras G12D.
  • the mutant or variant N-Ras G12 is N-Ras is N-Ras G12V. In some embodiments, the mutant or variant N-Ras G12 is N-Ras G12C. In some embodiments, the mutant or variant N-Ras G12 is N-Ras G12R. In some embodiments, the mutant or variant N- Ras G12 is N-Ras is N-Ras G12A. In some embodiments, the mutant or variant N-Ras G12 is N- Ras G12S. In some embodiments, the N-Ras mutant cancer is a cancer having a mutant or variant N-Ras Q61.
  • the N-Ras mutant cancer is a cancer having a mutant or variant N-Ras G12D, N-Ras G12V, N-Ras G12C, N-Ras G12R, N-Ras G12A, N-Ras G12S, N-Ras Q61, or any combination thereof.
  • the cancer is a C-Raf mutant cancer.
  • the mutant C-Raf cancer comprises a C-Raf amplification.
  • a “C-Raf amplification” refers to a cancer cell comprising at least three C-Raf copies, at least four C-Raf copies, at least five C-Raf copies, at least six C-Raf copies, at least seven C-Raf copies, at least eight C-Raf copies, at least nine C-Raf copies, at least ten C-Raf copies, or more.
  • the mutant C-Raf cancer is a cancer having a mutant or variant C-Raf S427.
  • the mutant or variant C-Raf S427 is C-Raf S427G.
  • the mutant C-Raf cancer is a cancer having a mutant or variant C-Raf I448.
  • the mutant C-Raf I448 is C-Raf I448V.
  • the mutant C-Raf cancer comprises a C-Raf amplification and/or at least one C-Raf mutant or variant C-Raf. [00130]
  • the cancer is an NF1 and/or NF2 mutant cancer.
  • an “NF1 mutant cancer” refers to a cancer having a mutant or variant gene encoding the neurofibromin protein, and includes deletion mutations, loss-of-function mutations, microdeletion mutations, missense mutations, copy number loss mutations, and substitution mutations.
  • an “NF2 mutant cancer” refers to a cancer having a mutant or variant gene encoding the Merlin protein (also known as schwannomin protein), and includes deletion mutations, loss-of- function mutations, microdeletion mutations, missense mutations, copy number loss mutations, and substitution mutations.
  • the cancer is selected from non-small cell lung cancer (NSCLC), pancreatic cancer, colorectal cancer (CRC), uterine carcinoma, endometrial carcinoma, bladder cancer, head and neck cancer, thyroid cancer, melanoma, multiple myeloma, acute myeloid leukemia (AML), low-grade serous ovarian cancer, neurofibroma, and glioma.
  • NSCLC non-small cell lung cancer
  • CRC colorectal cancer
  • uterine carcinoma endometrial carcinoma
  • bladder cancer bladder cancer
  • head and neck cancer thyroid cancer
  • melanoma multiple myeloma
  • AML acute myeloid leukemia
  • low-grade serous ovarian cancer neurofibroma
  • neurofibroma neurofibroma
  • glioma glioma.
  • the cancer is a K-Ras mutant non-small cell lung cancer (NSCLC).
  • NSCLC non-small cell lung cancer
  • the cancer is a K-Ra
  • the cancer is a K-Ras mutant colorectal cancer. In some embodiments of the methods and uses described herein, the cancer is a K-Ras mutant uterine carcinoma. In some embodiments of the methods and uses described herein, the cancer is a K-Ras mutant endometrial carcinoma. In some embodiments of the methods and uses described herein, the cancer is a K-Ras mutant bladder cancer. In some embodiments of the methods and uses described herein, the cancer is a K-Ras mutant head and neck cancer. In some embodiments of the methods and uses described herein, the cancer is a K-Ras mutant thyroid cancer.
  • the cancer is a K-Ras mutant low-grade serous ovarian cancer. In some embodiments of the methods and uses described herein, the cancer is an N-Ras mutant melanoma. In some embodiments of the methods and uses described herein, the cancer is an N-Ras mutant multiple myeloma. In some embodiments of the methods and uses described herein, the cancer is an N-Ras mutant acute myeloid leukemia (AML). In some embodiments of the methods and uses described herein, the cancer is an N-Ras mutant Bladder cancer. In some embodiments of the methods and uses described herein, the cancer is a K-Ras mutant low-grade serous ovarian cancer.
  • AML acute myeloid leukemia
  • the cancer is an N-Ras mutant Bladder cancer.
  • the cancer is a B-Raf mutant non-small cell lung cancer (NSCLC). In some embodiments of the methods and uses described herein, the cancer is a C-Raf mutant bladder cancer. In some embodiments of the methods and uses described herein, the cancer is an NF1 mutant glioma. In some embodiments of the methods and uses described herein, the cancer is an NF1 mutant non- small cell lung cancer (NSCLC). In some embodiments of the methods and uses described herein, the cancer is an NF2 mutant neurofibroma.
  • NSCLC non-small cell lung cancer
  • the cancer is lung cancer, thyroid cancer, ovarian cancer, colorectal cancer, prostate cancer, cancer of the pancreas, cancer of the esophagus, liver cancer, breast cancer, skin cancer, or mesothelioma.
  • the cancer is mesothelioma, such as malignant mesothelioma.
  • a cancer includes, without limitation, leukemias (e.g., acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, acute myeloblastic leukemia, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute erythroleukemia, chronic leukemia, chronic myelocytic leukemia, chronic lymphocytic leukemia), polycythemia vera, lymphoma (e.g., Hodgkin’s disease or non-Hodgkin’s disease), Waldenstrom's macroglobulinemia, multiple myeloma, heavy chain disease, and solid tumors such as sarcomas and carcinomas (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angios
  • leukemias e
  • a cancer is glioma, astrocytoma, glioblastoma multiforme (GBM, also known as glioblastoma), medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, schwannoma, neurofibrosarcoma, meningioma, melanoma, neuroblastoma, or retinoblastoma.
  • a cancer is acoustic neuroma, astrocytoma (e.g.
  • GBM Glioblastoma
  • the cancer is a type found more commonly in children than adults, such as brain stem glioma, craniopharyngioma, ependymoma, juvenile pilocytic astrocytoma (JPA), medulloblastoma, optic nerve glioma, pineal tumor, primitive neuroectodermal tumors (PNET), or rhabdoid tumor.
  • the patient is an adult human. In some embodiments, the patient is a child or pediatric patient.
  • Cancer includes, in another embodiment, without limitation, mesothelioma, hepatobilliary (hepatic and billiary duct), bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, ovarian cancer, colon cancer, rectal cancer, cancer of the anal region, stomach cancer, gastrointestinal (gastric, colorectal, and duodenal), uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin’s Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, testicular cancer, chronic or acute leukemia, chronic myeloid leukemia, lymph
  • a cancer is a solid tumor, such as a sarcoma, carcinoma, or lymphoma.
  • Solid tumors generally comprise an abnormal mass of tissue that typically does not include cysts or liquid areas.
  • the cancer is selected from renal cell carcinoma, or kidney cancer; hepatocellular carcinoma (HCC) or hepatoblastoma, or liver cancer; melanoma; breast cancer; colorectal carcinoma, or colorectal cancer; colon cancer; rectal cancer; anal cancer; lung cancer, such as non-small cell lung cancer (NSCLC) or small cell lung cancer (SCLC); ovarian cancer, ovarian epithelial cancer, ovarian carcinoma, or fallopian tube cancer; papillary serous cystadenocarcinoma or uterine papillary serous carcinoma (UPSC); prostate cancer; testicular cancer; gallbladder cancer; hepatocholangiocarcinoma; soft tissue and bone synovial sarcoma; rhabdomyo
  • HCC hepatocellular
  • a cancer is hepatocellular carcinoma (HCC).
  • the cancer is hepatoblastoma.
  • the cancer is colon cancer.
  • the cancer is rectal cancer.
  • the cancer is ovarian cancer, or ovarian carcinoma.
  • the cancer is ovarian epithelial cancer.
  • the cancer is fallopian tube cancer.
  • the cancer is papillary serous cystadenocarcinoma.
  • the cancer is uterine papillary serous carcinoma (UPSC).
  • the cancer is hepatocholangiocarcinoma.
  • the cancer is soft tissue and bone synovial sarcoma. In some embodiments, the cancer is rhabdomyosarcoma. In some embodiments, the cancer is osteosarcoma. In some embodiments, the cancer is anaplastic thyroid cancer. In some embodiments, the cancer is adrenocortical carcinoma. In some embodiments, the cancer is pancreatic cancer, or pancreatic ductal carcinoma. In some embodiments, the cancer is pancreatic adenocarcinoma. In some embodiments, the cancer is glioma. In some embodiments, the cancer is malignant peripheral nerve sheath tumors (MPNST). In some embodiments, the cancer is neurofibromatosis-1 associated MPNST.
  • MPNST peripheral nerve sheath tumors
  • a cancer is Waldenstrom’s macroglobulinemia. In some embodiments, the cancer is medulloblastoma.
  • a cancer is a viral-associated cancer, including human immunodeficiency virus (HIV) associated solid tumors, human papilloma virus (HPV)-16 positive incurable solid tumors, and adult T-cell leukemia, which is caused by human T-cell leukemia virus type I (HTLV-I) and is a highly aggressive form of CD4+ T-cell leukemia characterized by clonal integration of HTLV-I in leukemic cells (See https://clinicaltrials.gov/ct2/show/study/ NCT02631746); as well as virus-associated tumors in gastric cancer, nasopharyngeal carcinoma, cervical cancer, vaginal cancer, vulvar cancer, squamous cell carcinoma of the head and neck, and Merkel cell carcinoma.
  • HCV human immunodeficiency virus
  • HPV human papilloma virus
  • the methods or uses described herein inhibit or reduce or arrest or ameliorate the growth or spread of a cancer or tumor.
  • the tumor is treated by arresting, reducing, or inhibiting further growth of the cancer or tumor.
  • the methods or uses described herein increase or potentiate or activate one or more immune responses to inhibit or reduce or arrest or ameliorate the growth or spread of a cancer or tumor.
  • the cancer or tumor is treated by reducing the size (e.g., volume or mass) of the cancer or tumor by at least 5%, at least 10%, at least 25%, at least 50%, at least 75%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% relative to the size of the cancer or tumor prior to treatment.
  • cancers or tumors are treated by reducing the quantity of the cancers or tumors in the patient by at least 5%, at least 10%, at least 25%, at least 50%, at least 75%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% relative to the quantity of cancers or tumors prior to treatment.
  • a patient treated using the methods or uses described herein exhibits progression-free survival of at least about one month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months, at least about one year, at least about eighteen months, at least about two years, at least about three years, at least about four years, or at least about five years after the treatment is initiated.
  • a patient treated using the methods or uses described herein exhibits an overall survival of at least about one month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months, at least about one year, at least about 14 months, at least about 16 months, at least about 18 months, at least about 20 months, at least about 22 months, at least about two years, at least about three years, at least about four years, or at least about five years after the treatment is initiated.
  • a patient treated using the methods or uses described herein exhibits an objective response rate (ORR) of at least about 15%, at least about 20%, at least about 25%, at least about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%.
  • ORR objective response rate
  • the compounds and compositions, according to the method of the present disclosure may be administered using any amount and any route of administration effective for inhibiting MEK and treating or lessening the severity of a disease, for example, as those described herein.
  • dosage unit form refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present disclosure will be decided by the attending physician within the scope of sound medical judgment.
  • the specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.
  • patient means an animal, preferably a mammal, and most preferably a human.
  • compositions of this disclosure can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the disease or disorder being treated.
  • the compounds of the disclosure may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art such as, for example, water or other solvents,
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • Injectable formulations can be sterilized, for example, by filtration through a bacterial- retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this disclosure with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol
  • the dosage form may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • embedding compositions examples include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
  • the active compounds can also be in micro-encapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art.
  • the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.
  • Dosage forms for topical or transdermal administration of a compound of this disclosure include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • the active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required.
  • Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this disclosure.
  • the present disclosure contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body.
  • Such dosage forms can be made by dissolving or dispensing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin.
  • the rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • Co-Administration with One or More Other Therapeutic Agent(s) [00155] Depending upon the particular condition, or disease, to be treated, additional therapeutic agents that are normally administered to treat that condition, can also be present in the compositions of this disclosure.
  • additional therapeutic agents that are normally administered to treat a particular disease, or condition, are known as "appropriate for the disease, or condition, being treated.”
  • the present disclosure provides a method of treating a disclosed disease or condition comprising administering to a patient in need thereof an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof and co-administering simultaneously or sequentially an effective amount of one or more additional therapeutic agents, such as those described herein.
  • the method includes co-administering one additional therapeutic agent.
  • the method includes co-administering two additional therapeutic agents.
  • the combination of the disclosed compound and the additional therapeutic agent or agents acts synergistically.
  • a compound of the current disclosure can be administered alone or in combination with one or more other therapeutic compounds, possible combination therapy taking the form of fixed combinations or the administration of a compound of the disclosure and one or more other therapeutic compounds being staggered or given independently of one another, or the combined administration of fixed combinations and one or more other therapeutic compounds.
  • One or more other therapeutic agent(s) can be administered separately from a compound or composition of the disclosure, as part of a multiple dosage regimen.
  • one or more other therapeutic agent(s) may be part of a single dosage form, mixed together with a compound of this disclosure in a single composition.
  • one or more other therapeutic agent(s) and a compound or composition of the disclosure can be administered simultaneously, sequentially or within a period of time from one another, for example within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours from one another.
  • one or more other therapeutic agent(s) and a compound or composition of the disclosure are administered as a multiple dosage regimen within greater than 24 hours apart.
  • the term “combination,” “combined,” and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this disclosure.
  • a compound of the present disclosure can be administered with one or more other therapeutic agent(s) simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form.
  • the present disclosure provides a single unit dosage form comprising a compound of the current disclosure, one or more other therapeutic agent(s), and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • the amount of a compound of the disclosure and one or more other therapeutic agent(s) (in those compositions which comprise an additional therapeutic agent as described above) that can be combined with the carrier materials to produce a single dosage form varies depending upon the host treated and the particular mode of administration.
  • a composition of the disclosure should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of a compound of the disclosure can be administered.
  • the one or more other therapeutic agent(s) and a compound of the disclosure can act synergistically. Therefore, the amount of the one or more other therapeutic agent(s) in such compositions may be less than that required in a monotherapy utilizing only that therapeutic agent.
  • a dosage of between 0.01 - 1,000 g/kg body weight/day of the one or more other therapeutic agent(s) can be administered.
  • the amount of one or more other therapeutic agent(s) present in the compositions of this disclosure may be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent.
  • the amount of one or more other therapeutic agent(s) in the presently disclosed compositions ranges from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent.
  • one or more other therapeutic agent(s) is administered at a dosage of about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, or about 95% of the amount normally administered for that agent.
  • the phrase "normally administered” means the amount an FDA approved therapeutic agent is approved for dosing per the FDA label insert.
  • the compounds of this disclosure, or pharmaceutical compositions thereof can also be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents and catheters.
  • vascular stents for example, have been used to overcome restenosis (re-narrowing of the vessel wall after injury).
  • patients using stents or other implantable devices risk clot formation or platelet activation. These unwanted effects may be prevented or mitigated by pre-coating the device with a pharmaceutically acceptable composition comprising a kinase inhibitor.
  • the one or more other therapeutic agent is a TEAD inhibitor.
  • the TEAD inhibitor is selected from those described in WO 2020/243415, the contents of which are herein incorporated by reference in their entirety.
  • the TEAD inhibitor is selected from those described in WO 2020/243423, the contents of which are herein incorporated by reference in their entirety.
  • the TEAD inhibitor is selected from those described in U.S. Patent No.11,274,082, the contents of which are herein incorporated by reference in their entirety.
  • the TEAD inhibitor is selected from those described in WO 2022/120353, the contents of which are herein incorporated by reference in their entirety. In certain embodiments, the TEAD inhibitor is selected from those described in WO 2022/120354, the contents of which are herein incorporated by reference in their entirety.
  • the second anti-cancer agent is a TEAD degrader. In certain embodiments, the TEAD degrader is selected from those described in WO 2022/120355, the contents of which are herein incorporated by reference in their entirety. [00166] TEAD inhibitors can be produced by organic synthesis methods known to one of ordinary skill in the art.
  • TEAD inhibitors can be prepared as described in Pobbati et al., "Targeting the Central Pocket in Human Transcription Factor TEAD as a Potential Cancer Therapeutic Strategy," Structure 2015, 23, 2076-2086; Gibault et al., “Targeting Transcriptional Enhanced Associate Domains (TEADs),” J. Med. Chem.
  • the one or more other therapeutic agent is an ERK5 inhibitor. In some embodiments, the one or more other therapeutic agent is an ERK5 inhibitor. In certain embodiments, the ERK5 inhibitor is selected from those described in WO 2022/051567, the contents of which are herein incorporated by reference in their entirety. In certain embodiments, the ERK5 inhibitor is selected from those described in WO 2022/051565, the contents of which are herein incorporated by reference in their entirety. In certain embodiments, the ERK5 inhibitor is selected from those described in WO 2022/051569, the contents of which are herein incorporated by reference in their entirety.
  • the ERK5 inhibitor is selected from those described in WO 2022/051568, the contents of which are herein incorporated by reference in their entirety.
  • one or more other therapeutic agent is a Poly ADP ribose polymerase (PARP) inhibitor.
  • PARP Poly ADP ribose polymerase
  • a PARP inhibitor is selected from olaparib (LYNPARZA®, AstraZeneca); rucaparib (RUBRACA®, Clovis Oncology); niraparib (ZEJULA®, Tesaro); talazoparib (MDV3800/BMN 673/LT00673, Medivation/Pfizer/Biomarin); veliparib (ABT-888, AbbVie); and BGB-290 (BeiGene, Inc.).
  • one or more other therapeutic agent is a histone deacetylase (HDAC) inhibitor.
  • HDAC histone deacetylase
  • an HDAC inhibitor is selected from vorinostat (ZOLINZA®, Merck); romidepsin (ISTODAX®, Celgene); panobinostat (FARYDAK®, Novartis); belinostat (BELEODAQ®, Spectrum Pharmaceuticals); entinostat (SNDX-275, Syndax Pharmaceuticals) (NCT00866333); and chidamide (EPIDAZA®, HBI-8000, Chipscreen Biosciences, China).
  • one or more other therapeutic agent is a CDK inhibitor, such as a CDK4/CDK6 inhibitor.
  • a CDK 4/6 inhibitor is selected from palbociclib (IBRANCE®, Pfizer); ribociclib (KISQALI®, Novartis); abemaciclib (Ly2835219, Eli Lilly); and trilaciclib (G1T28, G1 Therapeutics).
  • one or more other therapeutic agent is a phosphatidylinositol 3 kinase (PI3K) inhibitor.
  • a PI3K inhibitor is selected from idelalisib (ZYDELIG®, Gilead), alpelisib (BYL719, Novartis), taselisib (GDC-0032, Genentech/Roche); pictilisib (GDC-0941, Genentech/Roche); copanlisib (BAY806946, Bayer); duvelisib (formerly IPI-145, Infinity Pharmaceuticals); PQR309 (Piqur Therapeutics, Switzerland); and TGR1202 (formerly RP5230, TG Therapeutics).
  • one or more other therapeutic agent is a platinum-based therapeutic, also referred to as platins.
  • a platinum-based therapeutic is selected from cisplatin (PLATINOL®, Bristol-Myers Squibb); carboplatin (PARAPLATIN®, Bristol-Myers Squibb; also, Teva; Pfizer); oxaliplatin (ELOXITIN® Sanofi-Aventis); nedaplatin (AQUPLA®, Shionogi), picoplatin (Poniard Pharmaceuticals); and satraplatin (JM-216, Agennix).
  • one or more other therapeutic agent is a taxane compound, which causes disruption of microtubules, which are essential for cell division.
  • a taxane compound is selected from paclitaxel (TAXOL®, Bristol-Myers Squibb), docetaxel (TAXOTERE®, Sanofi-Aventis; DOCEFREZ®, Sun Pharmaceutical), albumin-bound paclitaxel (ABRAXANE®; Abraxis/Celgene), cabazitaxel (JEVTANA®, Sanofi-Aventis), and SID530 (SK Chemicals, Co.) (NCT00931008).
  • one or more other therapeutic agent is a nucleoside inhibitor, or a therapeutic agent that interferes with normal DNA synthesis, protein synthesis, cell replication, or will otherwise inhibit rapidly proliferating cells.
  • a nucleoside inhibitor is selected from trabectedin (guanidine alkylating agent, YONDELIS®, Janssen Oncology), mechlorethamine (alkylating agent, VALCHLOR®, Aktelion Pharmaceuticals); vincristine (ONCOVIN®, Eli Lilly; VINCASAR®, Teva Pharmaceuticals; MARQIBO®, Talon Therapeutics); temozolomide (prodrug to alkylating agent 5-(3-methyltriazen-1-yl)-imidazole-4-carboxamide (MTIC) TEMODAR®, Merck); cytarabine injection (ara-C, antimetabolic cytidine analog, Pfizer); lomustine (alkylating agent,
  • one or more other therapeutic agent is a kinase inhibitor or VEGF-R antagonist.
  • Approved VEGF inhibitors and kinase inhibitors useful in the present disclosure include: bevacizumab (AVASTIN®, Genentech/Roche) an anti-VEGF monoclonal antibody; ramucirumab (CYRAMZA®, Eli Lilly), an anti-VEGFR-2 antibody and ziv-aflibercept, also known as VEGF Trap (ZALTRAP®; Regeneron/Sanofi).
  • VEGFR inhibitors such as regorafenib (STIVARGA®, Bayer); vandetanib (CAPRELSA®, AstraZeneca); axitinib (INLYTA®, Pfizer); and lenvatinib (LENVIMA®, Eisai); Raf inhibitors, such as sorafenib (NEXAVAR®, Bayer AG and Onyx); dabrafenib (TAFINLAR®, Novartis); and vemurafenib (ZELBORAF®, Genentech/Roche); MEK inhibitors, such as cobimetanib (COTELLIC®, Exelexis/Genentech/Roche); trametinib (MEKINIST®, Novartis); Bcr-Abl tyrosine kinase inhibitors, such as imatinib (GLEEVEC®, Novartis); nilotinib (TASIGNA®, Novartis); dasatinib (
  • kinase inhibitors and VEGF-R antagonists that are in development and may be used in the present disclosure include tivozanib (Aveo Pharmaecuticals); vatalanib (Bayer/Novartis); lucitanib (Clovis Oncology); dovitinib (TKI258, Novartis); Chiauanib (Chipscreen Biosciences); CEP-11981 (Cephalon); linifanib (Abbott Laboratories); neratinib (HKI-272, Puma Biotechnology); radotinib (SUPECT®, IY5511, Il-Yang Pharmaceuticals, S.
  • one or more other therapeutic agent is an mTOR inhibitor, which inhibits cell proliferation, angiogenesis and glucose uptake.
  • an mTOR inhibitor is everolimus (AFINITOR®, Novartis); temsirolimus (TORISEL®, Pfizer); and sirolimus (RAPAMUNE®, Pfizer).
  • one or more other therapeutic agent is a proteasome inhibitor.
  • Approved proteasome inhibitors useful in the present disclosure include bortezomib (VELCADE®, Takeda); carfilzomib (KYPROLIS®, Amgen); and ixazomib (NINLARO®, Takeda).
  • one or more other therapeutic agent is a growth factor antagonist, such as an antagonist of platelet-derived growth factor (PDGF), or epidermal growth factor (EGF) or its receptor (EGFR).
  • Approved PDGF antagonists which may be used in the present disclosure include olaratumab (LARTRUVO®; Eli Lilly).
  • Approved EGFR antagonists which may be used in the present disclosure include cetuximab (ERBITUX®, Eli Lilly); necitumumab (PORTRAZZA®, Eli Lilly), panitumumab (VECTIBIX®, Amgen); and osimertinib (targeting activated EGFR, TAGRISSO®, AstraZeneca).
  • one or more other therapeutic agent is an aromatase inhibitor.
  • an aromatase inhibitor is selected from exemestane (AROMASIN®, Pfizer); anastazole (ARIMIDEX®, AstraZeneca) and letrozole (FEMARA®, Novartis).
  • one or more other therapeutic agent is an antagonist of the hedgehog pathway.
  • Approved hedgehog pathway inhibitors which may be used in the present disclosure include sonidegib (ODOMZO®, Sun Pharmaceuticals); and vismodegib (ERIVEDGE®, Genentech), both for treatment of basal cell carcinoma.
  • one or more other therapeutic agent is a folic acid inhibitor. Approved folic acid inhibitors useful in the present disclosure include pemetrexed (ALIMTA®, Eli Lilly).
  • one or more other therapeutic agent is a CC chemokine receptor 4 (CCR4) inhibitor.
  • CCR4 inhibitors being studied that may be useful in the present disclosure include mogamulizumab (POTELIGEO®, Kyowa Hakko Kirin, Japan).
  • one or more other therapeutic agent is an isocitrate dehydrogenase (IDH) inhibitor.
  • IDH inhibitors being studied which may be used in the present disclosure include AG120 (Celgene; NCT02677922); AG221 (Celgene, NCT02677922; NCT02577406); BAY1436032 (Bayer, NCT02746081); IDH305 (Novartis, NCT02987010).
  • one or more other therapeutic agent is an arginase inhibitor.
  • Arginase inhibitors being studied which may be used in the present disclosure include AEB1102 (pegylated recombinant arginase, Aeglea Biotherapeutics), which is being studied in Phase 1 clinical trials for acute myeloid leukemia and myelodysplastic syndrome (NCT02732184) and solid tumors (NCT02561234); and CB-1158 (Calithera Biosciences).
  • one or more other therapeutic agent is a glutaminase inhibitor.
  • Glutaminase inhibitors being studied which may be used in the present disclosure include CB-839 (Calithera Biosciences).
  • one or more other therapeutic agent is an antibody that binds to tumor antigens, that is, proteins expressed on the cell surface of tumor cells.
  • Approved antibodies that bind to tumor antigens which may be used in the present disclosure include rituximab (RITUXAN®, Genentech/BiogenIdec); ofatumumab (anti-CD20, ARZERRA®, GlaxoSmithKline); obinutuzumab (anti-CD20, GAZYVA®, Genentech), ibritumomab (anti- CD20 and Yttrium-90, ZEVALIN®, Spectrum Pharmaceuticals); daratumumab (anti-CD38, DARZALEX®, Janssen Biotech), dinutuximab (anti-glycolipid GD2, UNITUXIN®, United Therapeutics); trastuzumab (anti-HER2, HERCEPTIN®, Genentech); ado-trastuzumab em
  • one or more other therapeutic agent is a topoisomerase inhibitor.
  • Approved topoisomerase inhibitors useful in the present disclosure include irinotecan (ONIVYDE®, Merrimack Pharmaceuticals); topotecan (HYCAMTIN®, GlaxoSmithKline).
  • Topoisomerase inhibitors being studied which may be used in the present disclosure include pixantrone (PIXUVRI®, CTI Biopharma).
  • one or more other therapeutic agent is an inhibitor of anti- apoptotic proteins, such as BCL-2.
  • Approved anti-apoptotics which may be used in the present disclosure include venetoclax (VENCLEXTA®, AbbVie/Genentech); and blinatumomab (BLINCYTO®, Amgen).
  • Other therapeutic agents targeting apoptotic proteins which have undergone clinical testing and may be used in the present disclosure include navitoclax (ABT-263, Abbott), a BCL-2 inhibitor (NCT02079740).
  • one or more other therapeutic agent is an androgen receptor inhibitor.
  • Approved androgen receptor inhibitors useful in the present disclosure include enzalutamide (XTANDI®, Astellas/Medivation); approved inhibitors of androgen synthesis include abiraterone (ZYTIGA®, Centocor/Ortho); approved antagonist of gonadotropin-releasing hormone (GnRH) receptor (degaralix, FIRMAGON®, Ferring Pharmaceuticals).
  • one or more other therapeutic agent is a selective estrogen receptor modulator (SERM), which interferes with the synthesis or activity of estrogens.
  • SERMs useful in the present disclosure include raloxifene (EVISTA®, Eli Lilly).
  • one or more other therapeutic agent is an inhibitor of bone resorption.
  • An approved therapeutic which inhibits bone resorption is Denosumab (XGEVA®, Amgen), an antibody that binds to RANKL, prevents binding to its receptor RANK, found on the surface of osteoclasts, their precursors, and osteoclast-like giant cells, which mediates bone pathology in solid tumors with osseous metastases.
  • Other approved therapeutics that inhibit bone resorption include bisphosphonates, such as zoledronic acid (ZOMETA®, Novartis).
  • one or more other therapeutic agent is an inhibitor of interaction between the two primary p53 suppressor proteins, MDMX and MDM2.
  • Inhibitors of p53 suppression proteins being studied which may be used in the present disclosure include ALRN- 6924 (Aileron), a stapled peptide that equipotently binds to and disrupts the interaction of MDMX and MDM2 with p53.
  • ALRN-6924 is currently being evaluated in clinical trials for the treatment of AML, advanced myelodysplastic syndrome (MDS) and peripheral T-cell lymphoma (PTCL) (NCT02909972; NCT02264613).
  • one or more other therapeutic agent is an inhibitor of transforming growth factor-beta (TGF-beta or TGF- ⁇ ).
  • Inhibitors of TGF-beta proteins being studied which may be used in the present disclosure include NIS793 (Novartis), an anti-TGF-beta antibody being tested in the clinic for treatment of various cancers, including breast, lung, hepatocellular, colorectal, pancreatic, prostate and renal cancer (NCT 02947165).
  • the inhibitor of TGF-beta proteins is fresolimumab (GC1008; Sanofi-Genzyme), which is being studied for melanoma (NCT00923169); renal cell carcinoma (NCT00356460); and non-small cell lung cancer (NCT02581787).
  • the additional therapeutic agent is a TGF-beta trap, such as described in Connolly et al. (2012) Int’l J. Biological Sciences 8:964-978.
  • TGF-beta trap such as described in Connolly et al. (2012) Int’l J. Biological Sciences 8:964-978.
  • M7824 Merck KgaA - formerly MSB0011459X
  • NCT02699515 a bispecific, anti-PD- L1/TGF- ⁇ trap compound
  • NCT02517398 NCT02517398
  • M7824 is comprised of a fully human IgG1 antibody against PD-L1 fused to the extracellular domain of human TGF-beta receptor II, which functions as a TGF- ⁇ “trap.”
  • one or more other therapeutic agent is selected from glembatumumab vedotin-monomethyl auristatin E (MMAE) (Celldex), an anti-glycoprotein NMB (gpNMB) antibody (CR011) linked to the cytotoxic MMAE.
  • gpNMB is a protein overexpressed by multiple tumor types associated with cancer cells’ ability to metastasize.
  • one or more other therapeutic agents is an antiproliferative compound.
  • antiproliferative compounds include, but are not limited to aromatase inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase II inhibitors; microtubule active compounds; alkylating compounds; histone deacetylase inhibitors; compounds which induce cell differentiation processes; cyclooxygenase inhibitors; MMP inhibitors; mTOR inhibitors; antineoplastic antimetabolites; platin compounds; compounds targeting/decreasing a protein or lipid kinase activity and further anti-angiogenic compounds; compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase; gonadorelin agonists; anti-androgens; methionine aminopeptidase inhibitors; matrix metalloproteinase inhibitors; bisphosphonates; biological response modifiers; antiproliferative antibodies; heparanase inhibitors; inhibitors of Ras oncogenic isoforms; telomerase inhibitors; proteasome inhibitors; compounds used in
  • aromatase inhibitor as used herein relates to a compound which inhibits estrogen production, for instance, the conversion of the substrates androstenedione and testosterone to estrone and estradiol, respectively.
  • the term includes, but is not limited to steroids, especially atamestane, exemestane and formestane and, in particular, non-steroids, especially aminoglutethimide, roglethimide, pyridoglutethimide, trilostane, testolactone, ketokonazole, vorozole, fadrozole, anastrozole and letrozole.
  • Exemestane is marketed under the trade name AROMASINTM.
  • Formestane is marketed under the trade name LENTARONTM. Fadrozole is marketed under the trade name AFEMATM. Anastrozole is marketed under the trade name ARIMIDEXTM. Letrozole is marketed under the trade names FEMARATM or FEMArTM. Aminoglutethimide is marketed under the trade name ORIMETENTM.
  • a combination of the disclosure comprising a chemotherapeutic agent which is an aromatase inhibitor is particularly useful for the treatment of hormone receptor positive tumors, such as breast tumors.
  • the term "antiestrogen” as used herein relates to a compound which antagonizes the effect of estrogens at the estrogen receptor level.
  • Tamoxifen is marketed under the trade name NOLVADEXTM.
  • Raloxifene hydrochloride is marketed under the trade name EVISTATM.
  • Fulvestrant can be administered under the trade name FASLODEXTMFulvestrant can be administered under the trade name FaslodexTM.
  • a combination of the disclosure comprising a chemotherapeutic agent which is an antiestrogen is particularly useful for the treatment of estrogen receptor positive tumors, such as breast tumors.
  • anti-androgen as used herein relates to any substance which is capable of inhibiting the biological effects of androgenic hormones and includes, but is not limited to, bicalutamide (CASODEXTM).
  • gonadorelin agonist as used herein includes, but is not limited to abarelix, goserelin, and goserelin acetate. Goserelin can be administered under the trade name ZOLADEXTM.
  • topoisomerase I inhibitor includes, but is not limited to topotecan, gimatecan, irinotecan, camptothecian and its analogues, 9-nitrocamptothecin and the macromolecular camptothecin conjugate PNU-166148.
  • Irinotecan can be administered, e.g., in the form as it is marketed, e.g., under the trademark CAMPTOSARTM.
  • Topotecan is marketed under the trade name HYCAMPTINTM.
  • topoisomerase II inhibitor includes, but is not limited to the anthracyclines such as doxorubicin (including liposomal formulation, such as CAELYXTM), daunorubicin, epirubicin, idarubicin and nemorubicin, the anthraquinones mitoxantrone and losoxantrone, and the podophillotoxines etoposide and teniposide.
  • Etoposide is marketed under the trade name ETOPOPHOSTM.
  • Teniposide is marketed under the trade name VM 26-Bristol
  • Doxorubicin is marketed under the trade name ACRIBLASTINTM or ADRIAMYCINTM.
  • microtubule active agent relates to microtubule stabilizing, microtubule destabilizing compounds and microtublin polymerization inhibitors including, but not limited to taxanes, such as paclitaxel and docetaxel; vinca alkaloids, such as vinblastine or vinblastine sulfate, vincristine or vincristine sulfate, and vinorelbine; discodermolides; cochicine and epothilones and derivatives thereof.
  • Paclitaxel is marketed under the trade name TAXOLTM. Docetaxel is marketed under the trade name TAXOTERETM. Vinblastine sulfate is marketed under the trade name VINBLASTIN R.PTM. Vincristine sulfate is marketed under the trade name FARMISTINTM.
  • alkylating agent includes, but is not limited to, cyclophosphamide, ifosfamide, melphalan or nitrosourea (BCNU or Gliadel). Cyclophosphamide is marketed under the trade name CYCLOSTINTM. Ifosfamide is marketed under the trade name HOLOXANTM.
  • histone deacetylase inhibitors or "HDAC inhibitors” relates to compounds which inhibit the histone deacetylase and which possess antiproliferative activity. This includes, but is not limited to, suberoylanilide hydroxamic acid (SAHA).
  • antiproliferative activity This includes, but is not limited to, suberoylanilide hydroxamic acid (SAHA).
  • antiproliferative activity This includes, but is not limited to, suberoylanilide hydroxamic acid (SAHA).
  • antiproliferative activity includes, but is not limited to, suberoylanilide hydroxamic acid (SAHA).
  • antiproliferative activity includes, but is not limited to, suberoylanilide hydroxamic acid (SAHA).
  • antiproliferative activity includes, but is not limited to, suberoylanilide hydroxamic acid (SAHA).
  • antiproliferative activity includes, but is not limited to, suberoylanilide hydroxamic acid (SAHA).
  • Gemcitabine is marketed under the trade name GEMZARTM.
  • the term "platin compound" as used herein includes, but is not limited to, carboplatin, cis-platin, cisplatinum and oxaliplatin.
  • Carboplatin can be administered, e.g., in the form as it is marketed, e.g., under the trademark CARBOPLATTM.
  • Oxaliplatin can be administered, e.g., in the form as it is marketed, e.g. under the trademark ELOXATINTM.
  • the term "compounds targeting/decreasing a protein or lipid kinase activity; or a protein or lipid phosphatase activity; or further anti-angiogenic compounds” as used herein includes, but is not limited to, protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, such as a) compounds targeting, decreasing or inhibiting the activity of the platelet-derived growth factor-receptors (PDGFR), such as compounds which target, decrease or inhibit the activity of PDGFR, especially compounds which inhibit the PDGF receptor, such as an N-phenyl-2-pyrimidine-amine derivative, such as imatinib, SU101, SU6668 and GFB- 111; b) compounds targeting, decreasing or inhibiting the activity of the fibroblast growth factor- receptors (FGFR); c) compounds targeting, decreasing or inhibiting the activity of the insulin-like growth factor receptor I (I
  • PI3K inhibitor includes, but is not limited to compounds having inhibitory activity against one or more enzymes in the phosphatidylinositol-3-kinase family, including, but not limited to PI3K ⁇ , PI3K ⁇ , PI3K ⁇ , PI3K ⁇ , PI3K-C2 ⁇ , PI3K-C2 ⁇ , PI3K-C2 ⁇ , PI3K- C2 ⁇ , Vps34, p110- ⁇ , p110- ⁇ , p110- ⁇ , p110- ⁇ , p85- ⁇ , p85- ⁇ , p55- ⁇ , p150, p101, and p87.
  • PI3K inhibitors useful in this disclosure include but are not limited to ATU-027, SF-1126, DS- 7423, PBI-05204, GSK-2126458, ZSTK-474, buparlisib, pictrelisib, PF-4691502, BYL-719, dactolisib, XL-147, XL-765, and idelalisib.
  • Bcl-2 inhibitor includes, but is not limited to compounds having inhibitory activity against B-cell lymphoma 2 protein (Bcl-2), including but not limited to ABT-199, ABT-731, ABT-737, apogossypol, Ascenta’s pan-Bcl-2 inhibitors, curcumin (and analogs thereof), dual Bcl-2/Bcl-xL inhibitors (Infinity Pharmaceuticals/Novartis Pharmaceuticals), Genasense (G3139), HA14-1 (and analogs thereof; see WO2008118802), navitoclax (and analogs thereof, see US7390799), NH-1 (Shenayng Pharmaceutical University), obatoclax (and analogs thereof, see WO2004106328), S-001 (Gloria Pharmaceuticals), TW series compounds (Univ.
  • Bcl-2 inhibitor as used herein includes, but is not limited to compounds having inhibitory activity against Bruton’s Tyrosine Kinase (BTK), including, but not limited to AVL-292 and ibrutinib.
  • SYK inhibitor includes, but is not limited to compounds having inhibitory activity against spleen tyrosine kinase (SYK), including but not limited to PRT- 062070, R-343, R-333, Excellair, PRT-062607, and fostamatinib.
  • SYK spleen tyrosine kinase
  • Further examples of BTK inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this disclosure can be found in WO2008039218 and WO2011090760, the entirety of which are incorporated herein by reference.
  • SYK inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this disclosure can be found in WO2003063794, WO2005007623, and WO2006078846, the entirety of which are incorporated herein by reference.
  • PI3K inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this disclosure can be found in WO2004019973, WO2004089925, WO2007016176, US8138347, WO2002088112, WO2007084786, WO2007129161, WO2006122806, WO2005113554, and WO2007044729 the entirety of which are incorporated herein by reference.
  • JAK inhibitory compounds and conditions treatable by such compounds in combination with compounds of this disclosure can be found in WO2009114512, WO2008109943, WO2007053452, WO2000142246, and WO2007070514, the entirety of which are incorporated herein by reference.
  • Further anti-angiogenic compounds include compounds having another mechanism for their activity, e.g., unrelated to protein or lipid kinase inhibition e.g., thalidomide (THALOMIDTM) and TNP-470.
  • TAALOMIDTM thalidomide
  • proteasome inhibitors useful for use in combination with compounds of the disclosure include, but are not limited to bortezomib, disulfiram, epigallocatechin-3-gallate (EGCG), salinosporamide A, carfilzomib, ONX-0912, CEP-18770, and MLN9708.
  • Compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase are e.g., inhibitors of phosphatase 1, phosphatase 2A, or CDC25, such as okadaic acid or a derivative thereof.
  • Compounds which induce cell differentiation processes include, but are not limited to, retinoic acid, ⁇ - ⁇ - or ⁇ - tocopherol or ⁇ - ⁇ - or ⁇ -tocotrienol.
  • the term cyclooxygenase inhibitor as used herein includes, but is not limited to, Cox- 2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acid and derivatives, such as celecoxib ( [00223] CELEBREXTM), rofecoxib (VIOXXTM), etoricoxib, valdecoxib or a 5-alkyl-2- arylaminophenylacetic acid, such as 5-methyl-2-(2'-chloro-6'-fluoroanilino)phenyl acetic acid, lumiracoxib.
  • bisphosphonates includes, but is not limited to, etridonic, clodronic, tiludronic, pamidronic, alendronic, ibandronic, risedronic and zoledronic acid.
  • Etridonic acid is marketed under the trade name DIDRONELTM.
  • Clodronic acid is marketed under the trade name BONEFOSTM.
  • Tiludronic acid is marketed under the trade name SkelidTM.
  • Pamidronic acid is marketed under the trade name AREDIATM.
  • Alendronic acid is marketed under the trade name FOSAMAXTM.
  • Ibandronic acid is marketed under the trade name BONDRANATTM.
  • Risedronic acid is marketed under the trade name ACTONELTM.
  • Zoledronic acid is marketed under the trade name ZOMETATM.
  • mTOR inhibitors relates to compounds which inhibit the mammalian target of rapamycin (mTOR) and which possess antiproliferative activity such as sirolimus (RAPAMUNE®), everolimus (CERTICANTM), CCI-779 and ABT578.
  • heparanase inhibitor refers to compounds which target, decrease or inhibit heparin sulfate degradation. The term includes, but is not limited to, PI-88.
  • biological response modifier as used herein refers to a lymphokine or interferons.
  • inhibitor of Ras oncogenic isoforms such as H-Ras, K-Ras, or N-Ras
  • a "farnesyl transferase inhibitor” such as L-744832, DK8G557 or R115777 (ZARNESTRA TM).
  • telomerase inhibitor refers to compounds which target, decrease or inhibit the activity of telomerase. Compounds which target, decrease or inhibit the activity of telomerase are especially compounds which inhibit the telomerase receptor, such as telomestatin.
  • methionine aminopeptidase inhibitor refers to compounds which target, decrease or inhibit the activity of methionine aminopeptidase.
  • compounds which target, decrease or inhibit the activity of methionine aminopeptidase include, but are not limited to, bengamide or a derivative thereof.
  • proteasome inhibitor refers to compounds which target, decrease or inhibit the activity of the proteasome.
  • Compounds which target, decrease or inhibit the activity of the proteasome include, but are not limited to, Bortezomib (VELCADETM) and MLN 341.
  • matrix metalloproteinase inhibitor or (“MMP” inhibitor) as used herein includes, but is not limited to, collagen peptidomimetic and nonpeptidomimetic inhibitors, tetracycline derivatives, e.g., hydroxamate peptidomimetic inhibitor batimastat and its orally bioavailable analogue marimastat (BB-2516), prinomastat (AG3340), metastat (NSC 683551) BMS-279251, BAY 12-9566, TAA211, MMI270B or AAJ996.
  • MMP matrix metalloproteinase inhibitor
  • FMS-like tyrosine kinase inhibitors which are compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon, 1- ⁇ -D-arabinofuransylcytosine (ara-c) and bisulfan; and ALK inhibitors, which are compounds which target, decrease or inhibit anaplastic lymphoma kinase.
  • FMS-like tyrosine kinase receptors are especially compounds, proteins or antibodies which inhibit members of the Flt-3R receptor kinase family, such as PKC412, midostaurin, a staurosporine derivative, SU11248 and MLN518.
  • HSP90 inhibitors includes, but is not limited to, compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90; degrading, targeting, decreasing or inhibiting the HSP90 client proteins via the ubiquitin proteosome pathway.
  • Compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90 are especially compounds, proteins or antibodies which inhibit the ATPase activity of HSP90, such as 17-allylamino,17-demethoxygeldanamycin (17AAG), a geldanamycin derivative; other geldanamycin related compounds; radicicol and HDAC inhibitors.
  • antiproliferative antibodies includes, but is not limited to, trastuzumab (HERCEPTINTM), Trastuzumab-DM1, erbitux, bevacizumab (AVASTINTM), rituximab (RITUXAN ® ), PRO64553 (anti-CD40) and 2C4 Antibody.
  • antibodies is meant intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least 2 intact antibodies, and antibodies fragments so long as they exhibit the desired biological activity.
  • compounds of the current disclosure can be used in combination with standard leukemia therapies, especially in combination with therapies used for the treatment of AML.
  • compounds of the current disclosure can be administered in combination with, for example, farnesyl transferase inhibitors and/or other drugs useful for the treatment of AML, such as Daunorubicin, Adriamycin, Ara-C, VP-16, Teniposide, Mitoxantrone, Idarubicin, Carboplatinum and PKC412.
  • HDAC histone deacetylase
  • SAHA suberoylanilide hydroxamic acid
  • HDAC inhibitors include MS275, SAHA, FK228 (formerly FR901228), Trichostatin A and compounds disclosed in US 6,552,065 including, but not limited to, N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)-ethyl]- amino]methyl]phenyl]- 2E-2-propenamide, or a pharmaceutically acceptable salt thereof and N-hydroxy-3-[4-[(2- hydroxyethyl) ⁇ 2-(1H-indol-3-yl)ethyl]-amino]methyl]phenyl]-2E-2- propenamide, or a pharmaceutically acceptable salt thereof, especially the lactate salt.
  • Somatostatin receptor antagonists as used herein refer to compounds which target, treat or inhibit the somatostatin receptor such as octreotide, and SOM230.
  • Tumor cell damaging approaches refer to approaches such as ionizing radiation.
  • the term "ionizing radiation” referred to above and hereinafter means ionizing radiation that occurs as either electromagnetic rays (such as X-rays and gamma rays) or particles (such as alpha and beta particles). Ionizing radiation is provided in, but not limited to, radiation therapy and is known in the art. See Hellman, Principles of Radiation Therapy, Cancer, in Principles and Practice of Oncology, Devita et al., Eds., 4 th Edition, Vol.1, pp.248-275 (1993).
  • EDG binders and ribonucleotide reductase inhibitors.
  • EDG binders refers to a class of immunosuppressants that modulates lymphocyte recirculation, such as FTY720.
  • ribonucleotide reductase inhibitors refers to pyrimidine or purine nucleoside analogs including, but not limited to, fludarabine and/or cytosine arabinoside (ara-C), 6-thioguanine, 5-fluorouracil, cladribine, 6-mercaptopurine (especially in combination with ara-C against ALL) and/or pentostatin.
  • Ribonucleotide reductase inhibitors are especially hydroxyurea or 2-hydroxy-1H-isoindole-1 ,3-dione derivatives.
  • VEGF such as 1-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine or a pharmaceutically acceptable salt thereof, 1-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine succinate; ANGIOSTATINTM; ENDOSTATINTM; anthranilic acid amides; ZD4190; Zd6474; SU5416; SU6668; bevacizumab; or anti-VEGF antibodies or anti-VEGF receptor antibodies, such as rhuMAb and RHUFab, VEGF aptamer such as Macugon; FLT-4 inhibitors, FLT-3 inhibitors, VEGFR-2 IgGI antibody, Angiozyme (VEGF aptamer such as Macugon; FLT-4 inhibitors,
  • Photodynamic therapy refers to therapy which uses certain chemicals known as photosensitizing compounds to treat or prevent cancers. Examples of photodynamic therapy include treatment with compounds, such as VISUDYNETM and porfimer sodium.
  • Angiostatic steroids as used herein refers to compounds which block or inhibit angiogenesis, such as, e.g., anecortave, triamcinolone, hydrocortisone, 11- ⁇ -epihydrocotisol, cortexolone, 17 ⁇ -hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone and dexamethasone.
  • Implants containing corticosteroids refers to compounds, such as fluocinolone and dexamethasone.
  • Other chemotherapeutic compounds include, but are not limited to, plant alkaloids, hormonal compounds and antagonists; biological response modifiers, preferably lymphokines or interferons; antisense oligonucleotides or oligonucleotide derivatives; shRNA or siRNA; or miscellaneous compounds or compounds with other or unknown mechanism of action.
  • the structure of the active compounds identified by code numbers, generic or trade names may be taken from the actual edition of the standard compendium "The Merck Index" or from databases, e.g., Patents International (e.g., IMS World Publications).
  • one or more other therapeutic agent is an immuno-oncology agent.
  • an immuno-oncology agent refers to an agent which is effective to enhance, stimulate, and/or up-regulate immune responses in a subject.
  • the administration of an immuno-oncology agent with a compound of the disclosure has a synergic effect in treating a cancer.
  • An immuno-oncology agent can be, for example, a small molecule drug, an antibody, or a biologic or small molecule.
  • biologic immuno-oncology agents include, but are not limited to, cancer vaccines, antibodies, and cytokines.
  • an antibody is a monoclonal antibody. In some embodiments, a monoclonal antibody is humanized or human.
  • an immuno-oncology agent is (i) an agonist of a stimulatory (including a co-stimulatory) receptor or (ii) an antagonist of an inhibitory (including a co- inhibitory) signal on T cells, both of which result in amplifying antigen-specific T cell responses.
  • Certain of the stimulatory and inhibitory molecules are members of the immunoglobulin super family (IgSF).
  • B7 family includes B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6.
  • B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6 includes B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6.
  • TNF family of molecules that bind to cognate TNF receptor family members which includes CD40 and CD40L, OX-40, OX-40L, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, TACI, APRIL, BCMA, LT ⁇ R, LIGHT, DcR3, HVEM, VEGI/TL1A, TRAMP/DR3, EDAR, EDA1, XEDAR, EDA2, TNFR1, Lymphotoxin ⁇ /TNF ⁇ , TNFR2, TNF ⁇ , LT ⁇ R, Lymphotoxin ⁇ 1 ⁇ 2, FA
  • an immuno-oncology agent is a cytokine that inhibits T cell activation (e.g., IL-6, IL-10, TGF- ⁇ , VEGF, and other immunosuppressive cytokines) or a cytokine that stimulates T cell activation, for stimulating an immune response.
  • a combination of a compound of the disclosure and an immuno- oncology agent can stimulate T cell responses.
  • an immuno-oncology agent is: (i) an antagonist of a protein that inhibits T cell activation (e.g., immune checkpoint inhibitors) such as CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, Galectin 9, CEACAM-1, BTLA, CD69, Galectin-1, TIGIT, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, and TIM- 4; or (ii) an agonist of a protein that stimulates T cell activation such as B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD70, CD27, CD40, DR3 and CD28H.
  • T cell activation e.g., immune checkpoint inhibitors
  • an antagonist of a protein that inhibits T cell activation e.g.,
  • an immuno-oncology agent is an antagonist of inhibitory receptors on NK cells or an agonist of activating receptors on NK cells.
  • an immuno-oncology agent is an antagonist of KIR, such as lirilumab.
  • an immuno-oncology agent is an agent that inhibits or depletes macrophages or monocytes, including but not limited to CSF-1R antagonists such as CSF-1R antagonist antibodies including RG7155 (WO11/70024, WO11/107553, WO11/131407, WO13/87699, WO13/119716, WO13/132044) or FPA-008 (WO11/140249; WO13169264; WO14/036357).
  • CSF-1R antagonists such as CSF-1R antagonist antibodies including RG7155 (WO11/70024, WO11/107553, WO11/131407, WO13/87699, WO13/119716, WO13/132044) or FPA-008 (WO11/140249; WO13169264; WO14/036357).
  • an immuno-oncology agent is selected from agonistic agents that ligate positive costimulatory receptors, blocking agents that attenuate signaling through inhibitory receptors, antagonists, and one or more agents that increase systemically the frequency of anti-tumor T cells, agents that overcome distinct immune suppressive pathways within the tumor microenvironment (e.g., block inhibitory receptor engagement (e.g., PD-L1/PD-1 interactions), deplete or inhibit Tregs (e.g., using an anti-CD25 monoclonal antibody (e.g., daclizumab) or by ex vivo anti-CD25 bead depletion), inhibit metabolic enzymes such as IDO, or reverse/prevent T cell energy or exhaustion) and agents that trigger innate immune activation and/or inflammation at tumor sites.
  • block inhibitory receptor engagement e.g., PD-L1/PD-1 interactions
  • Tregs e.g., using an anti-CD25 monoclonal antibody (e.g., daclizumab) or by ex
  • an immuno-oncology agent is a CTLA-4 antagonist.
  • a CTLA-4 antagonist is an antagonistic CTLA-4 antibody.
  • an antagonistic CTLA-4 antibody is YERVOY (ipilimumab) or tremelimumab.
  • an immuno-oncology agent is a PD-1 antagonist.
  • a PD-1 antagonist is administered by infusion.
  • an immuno- oncology agent is an antibody or an antigen-binding portion thereof that binds specifically to a Programmed Death-1 (PD-1) receptor and inhibits PD-1 activity.
  • a PD-1 antagonist is an antagonistic PD-1 antibody.
  • an antagonistic PD-1 antibody is OPDIVO (nivolumab), KEYTRUDA (pembrolizumab), or MEDI-0680 (AMP-514; WO2012/145493).
  • an immuno-oncology agent may be pidilizumab (CT- 011).
  • an immuno-oncology agent is a recombinant protein composed of the extracellular domain of PD-L2 (B7-DC) fused to the Fc portion of IgG1, called AMP-224.
  • an immuno-oncology agent is a PD-L1 antagonist.
  • a PD-L1 antagonist is an antagonistic PD-L1 antibody.
  • a PD-L1 antibody is MPDL3280A (RG7446; WO2010/077634), durvalumab (MEDI4736), BMS- 936559 (WO2007/005874), and MSB0010718C (WO2013/79174).
  • an immuno-oncology agent is a LAG-3 antagonist.
  • a LAG-3 antagonist is an antagonistic LAG-3 antibody.
  • a LAG3 antibody is BMS-986016 (WO10/19570, WO14/08218), or IMP-731 or IMP-321 (WO08/132601, WO009/44273).
  • an immuno-oncology agent is a CD137 (4-1BB) agonist.
  • a CD137 (4-1BB) agonist is an agonistic CD137 antibody.
  • a CD137 antibody is urelumab or PF-05082566 (WO12/32433).
  • an immuno-oncology agent is a GITR agonist.
  • a GITR agonist is an agonistic GITR antibody.
  • a GITR antibody is BMS-986153, BMS-986156, TRX-518 (WO006/105021, WO009/009116), or MK- 4166 (WO11/028683).
  • an immuno-oncology agent is an indoleamine (2,3)- dioxygenase (IDO) antagonist.
  • an IDO antagonist is selected from epacadostat (INCB024360, Incyte); indoximod (NLG-8189, NewLink Genetics Corporation); capmanitib (INC280, Novartis); GDC-0919 (Genentech/Roche); PF-06840003 (Pfizer); BMS:F001287 (Bristol-Myers Squibb); Phy906/KD108 (Phytoceutica); an enzyme that breaks down kynurenine (Kynase, Ikena Oncology, formerly known as Kyn Therapeutics); and NLG-919 (WO09/73620, WO009/1156652, WO11/56652, WO12/142237).
  • an immuno-oncology agent is an OX40 agonist.
  • an OX40 agonist is an agonistic OX40 antibody.
  • an OX40 antibody is MEDI-6383 or MEDI-6469.
  • an immuno-oncology agent is an OX40L antagonist.
  • an OX40L antagonist is an antagonistic OX40 antibody.
  • an OX40L antagonist is RG-7888 (WO06/029879).
  • an immuno-oncology agent is a CD40 agonist.
  • a CD40 agonist is an agonistic CD40 antibody.
  • an immuno- oncology agent is a CD40 antagonist. In some embodiments, a CD40 antagonist is an antagonistic CD40 antibody. In some embodiments, a CD40 antibody is lucatumumab or dacetuzumab. [00262] In some embodiments, an immuno-oncology agent is a CD27 agonist. In some embodiments, a CD27 agonist is an agonistic CD27 antibody. In some embodiments, a CD27 antibody is varlilumab. [00263] In some embodiments, an immuno-oncology agent is MGA271 (to B7H3) (WO11/109400).
  • an immuno-oncology agent is abagovomab, adecatumumab, afutuzumab, alemtuzumab, anatumomab mafenatox, apolizumab, atezolimab, avelumab, blinatumomab, BMS-936559, catumaxomab, durvalumab, epacadostat, epratuzumab, indoximod, inotuzumab ozogamicin, intelumumab, ipilimumab, isatuximab, lambrolizumab, MED14736, MPDL3280A, nivolumab, obinutuzumab, ocaratuzumab, ofatumumab, olatatumab, pembrolizumab, pidilizumab, rituximab,
  • an immuno-oncology agent is an immunostimulatory agent.
  • antibodies blocking the PD-1 and PD-L1 inhibitory axis can unleash activated tumor- reactive T cells and have been shown in clinical trials to induce durable anti-tumor responses in increasing numbers of tumor histologies, including some tumor types that conventionally have not been considered immunotherapy sensitive. See, e.g., Okazaki, T. et al. (2013) Nat. Immunol.14, 1212–1218; Zou et al. (2016) Sci. Transl. Med.8.
  • the anti-PD-1 antibody nivolumab (OPDIVO ® , Bristol-Myers Squibb, also known as ONO-4538, MDX1106 and BMS-936558), has shown potential to improve the overall survival in patients with RCC who had experienced disease progression during or after prior anti-angiogenic therapy.
  • the immunomodulatory therapeutic specifically induces apoptosis of tumor cells.
  • Approved immunomodulatory therapeutics which may be used in the present disclosure include pomalidomide (POMALYST®, Celgene); lenalidomide (REVLIMID®, Celgene); ingenol mebutate (PICATO®, LEO Pharma).
  • an immuno-oncology agent is a cancer vaccine.
  • the cancer vaccine is selected from sipuleucel-T (PROVENGE®, Dendreon/Valeant Pharmaceuticals), which has been approved for treatment of asymptomatic, or minimally symptomatic metastatic castrate-resistant (hormone-refractory) prostate cancer; and talimogene laherparepvec (IMLYGIC®, BioVex/Amgen, previously known as T-VEC), a genetically modified oncolytic viral therapy approved for treatment of unresectable cutaneous, subcutaneous and nodal lesions in melanoma.
  • sipuleucel-T PROVENGE®, Dendreon/Valeant Pharmaceuticals
  • IMLYGIC® BioVex/Amgen, previously known as T-VEC
  • an immuno-oncology agent is selected from an oncolytic viral therapy such as pexastimogene devacirepvec (PexaVec/JX-594, SillaJen/formerly Jennerex Biotherapeutics), a thymidine kinase- (TK-) deficient vaccinia virus engineered to express GM-CSF, for hepatocellular carcinoma (NCT02562755) and melanoma (NCT00429312); pelareorep (REOLYSIN®, Oncolytics Biotech), a variant of respiratory enteric orphan virus (reovirus) which does not replicate in cells that are not RAS-activated, in numerous cancers, including colorectal cancer (NCT01622543); prostate cancer (NCT01619813); head and neck squamous cell cancer (NCT01166542); pancreatic adenocarcinoma (NCT00998322); and non-small cell lung cancer (NSCLC) (
  • an immuno-oncology agent is selected from JX-929 (SillaJen/formerly Jennerex Biotherapeutics), a TK- and vaccinia growth factor-deficient vaccinia virus engineered to express cytosine deaminase, which is able to convert the prodrug 5- fluorocytosine to the cytotoxic drug 5-fluorouracil; TG01 and TG02 (Targovax/formerly Oncos), peptide-based immunotherapy agents targeted for difficult-to-treat RAS mutations; and TILT-123 (TILT Biotherapeutics), an engineered adenovirus designated: Ad5/3-E2F-delta24-hTNF ⁇ -IRES- hIL20; and VSV-GP (ViraTherapeutics) a vesicular stomatitis virus (VSV) engineered to express the glycoprotein (GP) of lymphocytic choriomeningitis virus (LCMV), which can
  • an immuno-oncology agent is a T-cell engineered to express a chimeric antigen receptor, or CAR.
  • the T-cells engineered to express such chimeric antigen receptor are referred to as a CAR-T cells.
  • CARs have been constructed that consist of binding domains, which may be derived from natural ligands, single chain variable fragments (scFv) derived from monoclonal antibodies specific for cell-surface antigens, fused to endodomains that are the functional end of the T-cell receptor (TCR), such as the CD3-zeta signaling domain from TCRs, which is capable of generating an activation signal in T lymphocytes.
  • binding domains which may be derived from natural ligands, single chain variable fragments (scFv) derived from monoclonal antibodies specific for cell-surface antigens, fused to endodomains that are the functional end of the T-cell receptor (TCR), such as the CD3-zeta signaling domain from TCRs
  • the CAR-T cell is one of those described in U.S. Patent 8,906,682 (June et al.; hereby incorporated by reference in its entirety), which discloses CAR-T cells engineered to comprise an extracellular domain having an antigen binding domain (such as a domain that binds to CD19), fused to an intracellular signaling domain of the T cell antigen receptor complex zeta chain (such as CD3 zeta).
  • an antigen binding domain such as a domain that binds to CD19
  • CD3 zeta intracellular signaling domain of the T cell antigen receptor complex
  • an immunostimulatory agent is an activator of retinoic acid receptor-related orphan receptor ⁇ (ROR ⁇ t).
  • ROR ⁇ t is a transcription factor with key roles in the differentiation and maintenance of Type 17 effector subsets of CD4+ (Th17) and CD8+ (Tc17) T cells, as well as the differentiation of IL-17 expressing innate immune cell subpopulations such as NK cells.
  • an activator of ROR ⁇ t is LYC-55716 (Lycera), which is currently being evaluated in clinical trials for the treatment of solid tumors (NCT02929862).
  • an immunostimulatory agent is an agonist or activator of a toll- like receptor (TLR).
  • TLR toll- like receptor
  • Suitable activators of TLRs include an agonist or activator of TLR9 such as SD-101 (Dynavax).
  • SD-101 is an immunostimulatory CpG which is being studied for B-cell, follicular and other lymphomas (NCT02254772).
  • Agonists or activators of TLR8 which may be used in the present disclosure include motolimod (VTX-2337, VentiRx Pharmaceuticals) which is being studied for squamous cell cancer of the head and neck (NCT02124850) and ovarian cancer (NCT02431559).
  • immuno-oncology agents that can be used in the present disclosure include urelumab (BMS-663513, Bristol-Myers Squibb), an anti-CD137 monoclonal antibody; varlilumab (CDX-1127, Celldex Therapeutics), an anti-CD27 monoclonal antibody; BMS-986178 (Bristol- Myers Squibb), an anti-OX40 monoclonal antibody; lirilumab (IPH2102/BMS-986015, Innate Pharma, Bristol-Myers Squibb), an anti-KIR monoclonal antibody; monalizumab (IPH2201, Innate Pharma, AstraZeneca) an anti-NKG2A monoclonal antibody; andecaliximab (GS-5745, Gilead Sciences), an anti-MMP9 antibody; MK-4166 (Merck & Co.), an anti-GITR monoclonal antibody.
  • urelumab BMS-663513, Bristol-
  • an immunostimulatory agent is selected from elotuzumab, mifamurtide, an agonist or activator of a toll-like receptor, and an activator of ROR ⁇ t.
  • an immunostimulatory therapeutic is recombinant human interleukin 15 (rhIL-15). rhIL-15 has been tested in the clinic as a therapy for melanoma and renal cell carcinoma (NCT01021059 and NCT01369888) and leukemias (NCT02689453).
  • an immunostimulatory agent is recombinant human interleukin 12 (rhIL-12).
  • an IL-15 based immunotherapeutic is heterodimeric IL-15 (hetIL-15, Novartis/Admune), a fusion complex composed of a synthetic form of endogenous IL-15 complexed to the soluble IL-15 binding protein IL-15 receptor alpha chain (IL15:sIL-15RA), which has been tested in Phase 1 clinical trials for melanoma, renal cell carcinoma, non-small cell lung cancer and head and neck squamous cell carcinoma (NCT02452268).
  • a recombinant human interleukin 12 (rhIL-12) is NM-IL-12 (Neumedicines, Inc.), NCT02544724, or NCT02542124.
  • an immuno-oncology agent is selected from those descripted in Jerry L. Adams et al., “Big opportunities for small molecules in immuno-oncology,” Cancer Therapy 2015, Vol.14, pages 603-622, the content of which is incorporated herein by reference in its entirety.
  • an immuno-oncology agent is selected from the examples described in Table 1 of Jerry L. Adams et al.
  • an immuno-oncology agent is a small molecule targeting an immuno-oncology target selected from those listed in Table 2 of Jerry L. Adams et al.
  • an immuno-oncology agent is a small molecule agent selected from those listed in Table 2 of Jerry L. Adams et al.
  • an immuno-oncology agent is selected from the small molecule immuno-oncology agents described in Peter L. Toogood, “Small molecule immuno-oncology therapeutic agents,” Bioorganic & Medicinal Chemistry Letters 2018, Vol.28, pages 319-329, the content of which is incorporated herein by reference in its entirety.
  • an immuno-oncology agent is an agent targeting the pathways as described in Peter L. Toogood.
  • an immuno-oncology agent is selected from those described in Sandra L.
  • an immuno-oncology agent is a bispecific T cell engager (BITE®) antibody construct.
  • a bispecific T cell engager (BITE®) antibody construct is a CD19/CD3 bispecific antibody construct.
  • a bispecific T cell engager (BITE®) antibody construct is an EGFR/CD3 bispecific antibody construct.
  • a bispecific T cell engager (BITE®) antibody construct activates T cells.
  • a bispecific T cell engager (BITE®) antibody construct activates T cells, which release cytokines inducing upregulation of intercellular adhesion molecule 1 (ICAM-1) and FAS on bystander cells.
  • a bispecific T cell engager (BITE®) antibody construct activates T cells which result in induced bystander cell lysis.
  • the bystander cells are in solid tumors.
  • the bystander cells being lysed are in proximity to the BITE®-activated T cells.
  • the bystander cells comprises tumor-associated antigen (TAA) negative cancer cells.
  • the bystander cells comprise EGFR-negative cancer cells.
  • an immuno- oncology agent is an antibody which blocks the PD-L1/PD1 axis and/or CTLA4.
  • an immuno-oncology agent is an ex vivo expanded tumor-infiltrating T cell.
  • an immuno-oncology agent is a bispecific antibody construct or chimeric antigen receptors (CARs) that directly connect T cells with tumor-associated surface antigens (TAAs).
  • CARs chimeric antigen receptors
  • TAAs tumor-associated surface antigens
  • Exemplary Immune Checkpoint Inhibitors [00280]
  • an immuno-oncology agent is an immune checkpoint inhibitor as described herein. [00281] The term “checkpoint inhibitor” as used herein relates to agents useful in preventing cancer cells from avoiding the immune system of the patient.
  • T-cell exhaustion results from chronic exposure to antigens that has led to up-regulation of inhibitory receptors.
  • inhibitory receptors serve as immune checkpoints in order to prevent uncontrolled immune reactions.
  • PD-1 and co-inhibitory receptors such as cytotoxic T-lymphocyte antigen 4 (CTLA-4, B and T Lymphocyte Attenuator (BTLA; CD272), T cell Immunoglobulin and Mucin domain-3 (Tim-3), Lymphocyte Activation Gene-3 (Lag-3; CD223), and others are often referred to as a checkpoint regulators.
  • an immune checkpoint inhibitor is an antibody to PD-1.
  • PD-1 binds to the programmed cell death 1 receptor (PD-1) to prevent the receptor from binding to the inhibitory ligand PDL-1, thus overriding the ability of tumors to suppress the host anti-tumor immune response.
  • the checkpoint inhibitor is a biologic therapeutic or a small molecule.
  • the checkpoint inhibitor is a monoclonal antibody, a humanized antibody, a fully human antibody, a fusion protein or a combination thereof.
  • the checkpoint inhibitor inhibits a checkpoint protein selected from CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands or a combination thereof.
  • a checkpoint protein selected from CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands or a combination thereof.
  • the checkpoint inhibitor interacts with a ligand of a checkpoint protein selected from CTLA-4, PDL1, PDL2, PDl, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands or a combination thereof.
  • the checkpoint inhibitor is an immunostimulatory agent, a T cell growth factor, an interleukin, an antibody, a vaccine or a combination thereof.
  • the interleukin is IL-7 or IL-15.
  • the interleukin is glycosylated IL-7.
  • the vaccine is a dendritic cell (DC) vaccine.
  • DC dendritic cell
  • Checkpoint inhibitors include any agent that blocks or inhibits in a statistically significant manner, the inhibitory pathways of the immune system. Such inhibitors may include small molecule inhibitors or may include antibodies, or antigen binding fragments thereof, that bind to and block or inhibit immune checkpoint receptors or antibodies that bind to and block or inhibit immune checkpoint receptor ligands.
  • Illustrative checkpoint molecules that can be targeted for blocking or inhibition include, but are not limited to, CTLA-4, PDL1, PDL2, PD1, B7-H3, B7- H4, BTLA, HVEM, GAL9, LAG3, TIM3, VISTA, KIR, 2B4 (belongs to the CD2 family of molecules and is expressed on all NK, ⁇ , and memory CD8 + ( ⁇ ) T cells), CD160 (also referred to as BY55), CGEN-15049, CHK 1 and CHK2 kinases, A2aR, and various B-7 family ligands.
  • CTLA-4 CTLA-4, PDL1, PDL2, PD1, B7-H3, B7- H4, BTLA, HVEM, GAL9, LAG3, TIM3, VISTA, KIR, 2B4 (belongs to the CD2 family of molecules and is expressed on all NK, ⁇ , and memory CD8 + ( ⁇ ) T cells
  • CD160 also referred to as BY55
  • B7 family ligands include, but are not limited to, B7- 1, B7-2, B7-DC, B7-H1, B7-H2, B7-H3, B7- H4, B7-H5, B7-H6 and B7-H7.
  • Checkpoint inhibitors include antibodies, or antigen binding fragments thereof, other binding proteins, biologic therapeutics, or small molecules, that bind to and block or inhibit the activity of one or more of CTLA-4, PDL1, PDL2, PD1, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD 160 and CGEN-15049.
  • Illustrative immune checkpoint inhibitors include, but are not limited to, Tremelimumab (CTLA-4 blocking antibody), anti-OX40, PD-L1 monoclonal Antibody (Anti-B7-Hl; MEDI4736), MK-3475 (PD-1 blocker), Nivolumab (anti-PD1 antibody), CT-011 (anti-PD1 antibody), BY55 monoclonal antibody, AMP224 (anti-PDL1 antibody), BMS- 936559 (anti-PDL1 antibody), MPLDL3280A (anti-PDL1 antibody), MSB0010718C (anti-PDL1 antibody), and ipilimumab (anti-CTLA-4 checkpoint inhibitor).
  • CTLA-4 blocking antibody PD-L1 monoclonal Antibody
  • Anti-B7-Hl MEDI4736
  • MK-3475 PD-1 blocker
  • Nivolumab anti-PD1 antibody
  • CT-011 anti-PD1 antibody
  • BY55 monoclonal antibody AMP
  • Checkpoint protein ligands include, but are not limited to PD-L1, PD-L2, B7-H3, B7- H4, CD28, CD86 and TIM-3.
  • the immune checkpoint inhibitor is selected from a PD-1 antagonist, a PD-L1 antagonist, and a CTLA-4 antagonist.
  • the checkpoint inhibitor is selected from the group consisting of nivolumab (OPDIVO®), ipilimumab (YERVOY®), and pembrolizumab (KEYTRUDA®).
  • the checkpoint inhibitor is selected from nivolumab (anti-PD-1 antibody, OPDIVO®, Bristol-Myers Squibb); pembrolizumab (anti-PD-1 antibody, KEYTRUDA®, Merck); ipilimumab (anti-CTLA-4 antibody, YERVOY®, Bristol-Myers Squibb); durvalumab (anti-PD-L1 antibody, IMFINZI®, AstraZeneca); and atezolizumab (anti-PD-L1 antibody, TECENTRIQ®, Genentech).
  • nivolumab anti-PD-1 antibody, OPDIVO®, Bristol-Myers Squibb
  • pembrolizumab anti-PD-1 antibody, KEYTRUDA®, Merck
  • ipilimumab anti-CTLA-4 antibody, YERVOY®, Bristol-Myers Squibb
  • durvalumab anti-PD-L1 antibody, IMFINZI®,
  • the checkpoint inhibitor is selected from the group consisting of lambrolizumab (MK-3475), nivolumab (BMS-936558), pidilizumab (CT-011), AMP-224, MDX-1105, MEDI4736, MPDL3280A, BMS-936559, ipilimumab, lirlumab, IPH2101, pembrolizumab (KEYTRUDA®), and tremelimumab.
  • MK-3475 lambrolizumab
  • BMS-936558 nivolumab
  • CT-011 pidilizumab
  • AMP-224 pidilizumab
  • MDX-1105 MEDI4736
  • MPDL3280A MPDL3280A
  • BMS-936559 ipilimumab
  • lirlumab IPH2101, pembrolizumab (KEYTRUDA®)
  • tremelimumab tremelimum
  • an immune checkpoint inhibitor is REGN2810 (Regeneron), an anti-PD-1 antibody tested in patients with basal cell carcinoma (NCT03132636); NSCLC (NCT03088540); cutaneous squamous cell carcinoma (NCT02760498); lymphoma (NCT02651662); and melanoma (NCT03002376); pidilizumab (CureTech), also known as CT- 011, an antibody that binds to PD-1, in clinical trials for diffuse large B-cell lymphoma and multiple myeloma; avelumab (BAVENCIO®, Pfizer/Merck KGaA), also known as MSB0010718C), a fully human IgG1 anti-PD-L1 antibody, in clinical trials for non-small cell lung cancer, Merkel cell carcinoma, mesothelioma, solid tumors, renal cancer, ovarian cancer, bladder cancer, head and neck cancer, and gastric cancer; or P
  • Tremelimumab (CP-675,206; Astrazeneca) is a fully human monoclonal antibody against CTLA-4 that has been in studied in clinical trials for a number of indications, including: mesothelioma, colorectal cancer, kidney cancer, breast cancer, lung cancer and non-small cell lung cancer, pancreatic ductal adenocarcinoma, pancreatic cancer, germ cell cancer, squamous cell cancer of the head and neck, hepatocellular carcinoma, prostate cancer, endometrial cancer, metastatic cancer in the liver, liver cancer, large B-cell lymphoma, ovarian cancer, cervical cancer, metastatic anaplastic thyroid cancer, urothelial cancer, fallopian tube cancer, multiple myeloma, bladder cancer, soft tissue sarcoma, and melanoma.
  • AGEN-1884 (Agenus) is an anti-CTLA4 antibody that is being studied in Phase 1 clinical trials for advanced solid tumors (NCT02694822).
  • a checkpoint inhibitor is an inhibitor of T-cell immunoglobulin mucin containing protein-3 (TIM-3).
  • TIM-3 inhibitors that may be used in the present disclosure include TSR-022, LY3321367 and MBG453.
  • TSR-022 (Tesaro) is an anti-TIM-3 antibody which is being studied in solid tumors (NCT02817633).
  • LY3321367 (Eli Lilly) is an anti-TIM-3 antibody which is being studied in solid tumors (NCT03099109).
  • a checkpoint inhibitor is an inhibitor of T cell immunoreceptor with Ig and ITIM domains, or TIGIT, an immune receptor on certain T cells and NK cells.
  • TIGIT inhibitors that may be used in the present disclosure include BMS-986207 (Bristol-Myers Squibb), an anti-TIGIT monoclonal antibody (NCT02913313); OMP-313M32 (Oncomed); and anti-TIGIT monoclonal antibody (NCT03119428).
  • a checkpoint inhibitor is an inhibitor of Lymphocyte Activation Gene-3 (LAG-3).
  • LAG-3 inhibitors that may be used in the present disclosure include BMS- 986016 and REGN3767 and IMP321.
  • BMS-986016 (Bristol-Myers Squibb), an anti-LAG-3 antibody, is being studied in glioblastoma and gliosarcoma (NCT02658981).
  • REGN3767 (Regeneron), is also an anti-LAG-3 antibody, and is being studied in malignancies (NCT03005782).
  • IMP321 is an LAG-3-Ig fusion protein, being studied in melanoma (NCT02676869); adenocarcinoma (NCT02614833); and metastatic breast cancer (NCT00349934).
  • Checkpoint inhibitors that may be used in the present disclosure include OX40 agonists.
  • OX40 agonists that are being studied in clinical trials include PF-04518600/PF-8600 (Pfizer), an agonistic anti-OX40 antibody, in metastatic kidney cancer (NCT03092856) and advanced cancers and neoplasms (NCT02554812; NCT05082566); GSK3174998 (Merck), an agonistic anti-OX40 antibody, in Phase 1 cancer trials (NCT02528357); MEDI0562 (Medimmune/AstraZeneca), an agonistic anti-OX40 antibody, in advanced solid tumors (NCT02318394 and NCT02705482); MEDI6469, an agonistic anti-OX40 antibody (Medimmune/AstraZeneca), in patients with colorectal cancer (NCT02559024), breast cancer (NCT01862900), head and neck cancer (NCT02274155) and metastatic prostate cancer (NCT01303705); and BMS-986178 (Bristol-My
  • Checkpoint inhibitors that may be used in the present disclosure include CD137 (also called 4-1BB) agonists.
  • CD137 agonists that are being studied in clinical trials include utomilumab (PF-05082566, Pfizer) an agonistic anti-CD137 antibody, in diffuse large B-cell lymphoma (NCT02951156) and in advanced cancers and neoplasms (NCT02554812 and NCT05082566); urelumab (BMS-663513, Bristol-Myers Squibb), an agonistic anti-CD137 antibody, in melanoma and skin cancer (NCT02652455) and glioblastoma and gliosarcoma (NCT02658981); and CTX-471 (Compass Therapeutics), an agonistic anti-CD137 antibody in metastatic or locally advanced malignancies (NCT03881488).
  • Checkpoint inhibitors that may be used in the present disclosure include CD27 agonists.
  • CD27 agonists that are being studied in clinical trials include varlilumab (CDX-1127, Celldex Therapeutics) an agonistic anti-CD27 antibody, in squamous cell head and neck cancer, ovarian carcinoma, colorectal cancer, renal cell cancer, and glioblastoma (NCT02335918); lymphomas (NCT01460134); and glioma and astrocytoma (NCT02924038).
  • Checkpoint inhibitors that may be used in the present disclosure include glucocorticoid- induced tumor necrosis factor receptor (GITR) agonists.
  • GITR glucocorticoid- induced tumor necrosis factor receptor
  • GITR agonists that are being studied in clinical trials include TRX518 (Leap Therapeutics), an agonistic anti-GITR antibody, in malignant melanoma and other malignant solid tumors (NCT01239134 and NCT02628574); GWN323 (Novartis), an agonistic anti-GITR antibody, in solid tumors and lymphoma (NCT 02740270); INCAGN01876 (Incyte/Agenus), an agonistic anti-GITR antibody, in advanced cancers (NCT02697591 and NCT03126110); MK-4166 (Merck), an agonistic anti-GITR antibody, in solid tumors (NCT02132754) and MEDI1873 (Medimmune/AstraZeneca), an agonistic hexameric GITR-ligand molecule with a human IgG1 Fc domain, in advanced solid tumors (NCT02583165).
  • TRX518 Leap Therapeutics
  • Checkpoint inhibitors that may be used in the present disclosure include inducible T- cell co-stimulator (ICOS, also known as CD278) agonists.
  • ICOS agonists that are being studied in clinical trials include MEDI-570 (Medimmune), an agonistic anti-ICOS antibody, in lymphomas (NCT02520791); GSK3359609 (Merck), an agonistic anti-ICOS antibody, in Phase 1 (NCT02723955); JTX-2011 (Jounce Therapeutics), an agonistic anti-ICOS antibody, in Phase 1 (NCT02904226).
  • Checkpoint inhibitors that may be used in the present disclosure include killer IgG-like receptor (KIR) inhibitors.
  • KIR killer IgG-like receptor
  • KIR inhibitors that are being studied in clinical trials include lirilumab (IPH2102/BMS-986015, Innate Pharma/Bristol-Myers Squibb), an anti-KIR antibody, in leukemias (NCT01687387, NCT02399917, NCT02481297, NCT02599649), multiple myeloma (NCT02252263), and lymphoma (NCT01592370); IPH2101 (1-7F9, Innate Pharma) in myeloma (NCT01222286 and NCT01217203); and IPH4102 (Innate Pharma), an anti-KIR antibody that binds to three domains of the long cytoplasmic tail (KIR3DL2), in lymphoma (NCT02593045).
  • Checkpoint inhibitors that may be used in the present disclosure include CD47 inhibitors of interaction between CD47 and signal regulatory protein alpha (SIRPa).
  • CD47/SIRPa inhibitors that are being studied in clinical trials include ALX-148 (Alexo Therapeutics), an antagonistic variant of (SIRPa) that binds to CD47 and prevents CD47/SIRPa-mediated signaling, in phase 1 (NCT03013218); TTI-621 (SIRPa-Fc, Trillium Therapeutics), a soluble recombinant fusion protein created by linking the N-terminal CD47-binding domain of SIRPa with the Fc domain of human IgG1, acts by binding human CD47, and preventing it from delivering its “do not eat” signal to macrophages, is in clinical trials in Phase 1 (NCT02890368 and NCT02663518); CC-90002 (Celgene), an anti-CD47 antibody, in leukemias (NCT02641002); and Hu
  • Checkpoint inhibitors that may be used in the present disclosure include CD73 inhibitors.
  • CD73 inhibitors that are being studied in clinical trials include MEDI9447 (Medimmune), an anti-CD73 antibody, in solid tumors (NCT02503774); and BMS-986179 (Bristol-Myers Squibb), an anti-CD73 antibody, in solid tumors (NCT02754141).
  • Checkpoint inhibitors that may be used in the present disclosure include agonists of stimulator of interferon genes protein (STING, also known as transmembrane protein 173, or TMEM173).
  • STING stimulator of interferon genes protein
  • Agonists of STING that are being studied in clinical trials include MK-1454 (Merck), an agonistic synthetic cyclic dinucleotide, in lymphoma (NCT03010176); and ADU- S100 (MIW815, Aduro Biotech/Novartis), an agonistic synthetic cyclic dinucleotide, in Phase 1 (NCT02675439 and NCT03172936).
  • Checkpoint inhibitors that may be used in the present disclosure include CSF1R inhibitors.
  • CSF1R inhibitors that are being studied in clinical trials include pexidartinib (PLX3397, Plexxikon), a CSF1R small molecule inhibitor, in colorectal cancer, pancreatic cancer, metastatic and advanced cancers (NCT02777710) and melanoma, non-small cell lung cancer, squamous cell head and neck cancer, gastrointestinal stromal tumor (GIST) and ovarian cancer (NCT02452424); and IMC-CS4 (LY3022855, Lilly), an anti-CSF-1R antibody, in pancreatic cancer (NCT03153410), melanoma (NCT03101254), and solid tumors (NCT02718911); and BLZ945 (4-[2((1R,2R)-2-hydroxycyclohexylamino)-benzothiazol-6-yloxyl]-pyridine-2- carboxylic acid methylamide, Novartis), an orally available inhibitor of CSF1R, in advanced solid tumors
  • Checkpoint inhibitors that can be used in the present disclosure include NKG2A receptor inhibitors.
  • NKG2A receptor inhibitors that are being studied in clinical trials include monalizumab (IPH2201, Innate Pharma), an anti-NKG2A antibody, in head and neck neoplasms (NCT02643550) and chronic lymphocytic leukemia (NCT02557516).
  • the immune checkpoint inhibitor is selected from nivolumab, pembrolizumab, ipilimumab, avelumab, durvalumab, atezolizumab, or pidilizumab.
  • EXAMPLE 1 Synthesis of Certain Exemplary Compounds Synthesis of 3-[[2-Fluoro-3-(methylsulfamoylamino)phenyl]methyl]-7-pyrimidin-2-yloxy- 4H-1,3-benzoxazin-2-one (I-3) Step 1: 3-Amino-2-fluorobenzonitrile [00305] To a stirred solution of 2-fluoro-3-nitrobenzonitrile (2 g, 12.04 mmol, 1 eq) in EtOH (20 mL) and H 2 O (4 mL) was added NH 4 Cl (6.44 g, 120.40 mmol, 10 eq) and Fe (3.36 g, 60.20 mmol, 5 eq).
  • reaction mixture was stirred at 90 °C for 12 h under N2.
  • the reaction mixture was filtered through celite and the filtrate was concentrated to yield a residue which was diluted with H 2 O (100 mL) and extracted with EtOAc (100 mL ⁇ 3).
  • the combined organic layers were washed with brine (30 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to yield 3-amino-2-fluorobenzonitrile (1.4 g, crude) as a black brown solid.
  • Step 2 tert-Butyl N-tert-butoxycarbonyl-N-(3-cyano-2-fluoro-phenyl)carbamate
  • Boc 2 O 6.73 g, 30.85 mmol, 7.09 mL, 3 eq
  • DMAP 251.29 mg, 2.06 mmol, 0.2 eq
  • TEA 2.08 g, 20.57 mmol, 2.86 mL, 2 eq
  • Step 3 tert-Butyl N-[3-(aminomethyl)-2-fluoro-phenyl]-N-tert-butoxycarbonyl-carbamate [00307] To a solution of tert-butyl N-tert-butoxycarbonyl-N-(3-cyano-2-fluoro- phenyl)carbamate (1.5 g, 3.48 mmol, 78.0% purity, 1 eq) in MeOH (60 mL) was added Raney-Ni (298.02 mg, 3.48 mmol, 1 eq) and NH3.H 2 O (1.31 g, 10.44 mmol, 1.44 mL, 28%, 3 eq) under N2.
  • Step 4 2-Hydroxy-4-pyrimidin-2-yloxy-benzaldehyde [00308] To a stirred solution of 2,4-dihydroxybenzaldehyde (5 g, 36.20 mmol, 1 eq) in DMF (80 mL) was added Cs2CO3 (35.38 g, 108.60 mmol, 3 eq) and 2-bromopyrimidine (5.76 g, 36.20 mmol, 1 eq). The reaction mixture was stirred at 80 °C for 12 h under N2. The reaction mixture was diluted with H 2 O (300 mL) and extracted with EtOAc (500mL ⁇ 3).
  • Step 5 tert-Butyl N-tert-butoxycarbonyl-N-[2-fluoro-3-[[(2-hydroxy-4-pyrimidin-2-yloxy- phenyl)methylamino]methyl]phenyl]carbamate
  • 2-hydroxy-4-pyrimidin-2-yloxy-benzaldehyde 400 mg, 1.72 mmol, 93.0% purity, 1 eq
  • tert-butyl N-[3-(aminomethyl)-2-fluoro-phenyl]-N-tert-butoxycarbonyl- carbamate 585.70 mg, 1.72 mmol, 1 eq
  • MeOH 10 mL
  • AcOH 103.33 mg, 1.72 mmol, 98.41 ⁇ L, 1 eq
  • NaBH 3 CN 324.39 mg, 5.16 mmol, 3 eq
  • Step 6 tert-Butyl N-tert-butoxycarbonyl-N-[2-fluoro-3-[(2-oxo-7-pyrimidin-2-yloxy-4H-1,3- benzoxazin-3-yl)methyl]phenyl]carbamate
  • tert-butyl N-tert-butoxycarbonyl-N-[2-fluoro-3-[[(2-hydroxy- 4-pyrimidin-2-yloxy-phenyl)methylamino]methyl]phenyl]carbamate 80 mg, 136.15 ⁇ mol, 92% purity, 1 eq
  • CDI 66.23 mg, 408.45 ⁇ mol, 3 eq
  • Step 7 3-[(3-Amino-2-fluoro-phenyl)methyl]-7-pyrimidin-2-yloxy-4H-1,3-benzoxazin-2-one [00311]
  • tert-butyl N-tert-butoxycarbonyl-N-[2-fluoro-3-[(2-oxo-7- pyrimidin-2-yloxy-4H-1,3-benzoxazin-3-yl)methyl]phenyl]carbamate 50.00 mg, 83.84 ⁇ mol, 95.0% purity, 1 eq) in DCM (2.5 mL) was added TFA (0.5 mL). The reaction mixture was stirred at 25 °C for 1 h under N 2 .
  • Step 8 3-[[2-Fluoro-3-(methylsulfamoylamino)phenyl]methyl]-7-pyrimidin-2-yloxy-4H-1,3- benzoxazin-2-one [00312] To a stirred solution of 3-[(3-amino-2-fluoro-phenyl)methyl]-7-pyrimidin-2-yloxy-4H- 1,3-benzoxazin-2-one (35 mg, 72.86 ⁇ mol, 1 eq, TFA) in DCM (3 mL) was added DIEA (28.25 mg, 218.58 ⁇ mol, 38.07 ⁇ L, 3 eq) and METHYLSULFAMOYL CHLORIDE (8.50 mg, 65.57 ⁇ mol, 0.9 eq) under N2 atmosphere at 0 °C.
  • reaction mixture was stirred at 25 °C for 12 h under N 2 .
  • the reaction mixture was concentrated under reduced pressure to yield a residue which was purified by preparative HPLC (column: Phenomenex Luna C18150*25mm*10 ⁇ m; mobile phase: [water( NH4HCO3)-ACN]; B%: 22%-52%, 8min) followed by lyophilization to yield 3-[[2- fluoro-3-(methylsulfamoylamino)phenyl]methyl]-7-pyrimidin-2-yloxy-4H-1,3-benzoxazin-2-one (5.6 mg, 11.85 ⁇ mol, 16.3% yield, 97.2% purity) as a white solid.
  • Step 2 4-((((2-chloro-3-fluoropyridin-4-yl)methyl)amino)methyl)benzene-1,3-diol
  • [(2-chloro-3-fluoropyridin-4- yl)methyl][(2,4-dimethoxyphenyl)methyl]amine (31.0 g, 99 mmol, 1.0 eq) and DCE (400 mL) at room temperature.
  • BBr 3 (249.9 g, 997 mmol, 10.0 eq) dropwise over 30 min at 0°C.
  • Step 3 3-((2-chloro-3-fluoropyridin-4-yl)methyl)-7-hydroxy-3,4-dihydro-2H- benzo[e][1,3]oxazin-2-one [00317]
  • DCM 300 mL
  • Pyridine 50.4 g, 636 mmol, 10.0 eq
  • Step 4 3-((2-chloro-3-fluoropyridin-4-yl)methyl)-7-((3-fluoropyridin-2-yl)oxy)-3,4-dihydro- 2H-benzo[e][1,3]oxazin-2-one [00319]
  • 3-[(2-chloro-3-fluoropyridin- 4-yl)methyl]-7-hydroxy-4H-1,3-benzoxazin-2-one (15.0 g, 48 mmol, 1.0 eq)
  • 2,3-difluoropyridine (8.4 g, 72 mmol, 1.5 eq
  • Cs 2 CO 3 (47.5 g, 145 mmol, 3.0 eq)
  • DMF 200 mL
  • Step 5 3-((2-((diphenylmethylene)amino)-3-fluoropyridin-4-yl)methyl)-7-((3-fluoropyridin- 2-yl)oxy)-3,4-dihydro-2H-benzo[e][1,3]oxazin-2-one [00321]
  • 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-[(3-fluoropyridin-2- yl)oxy]-4H-1,3-benzoxazin-2-one (11.5 g, 28 mmol, 1.0 eq) and diphenylmethanimine (7.7 g, 42 mmol, 1.5 eq) in dioxane (200 mL) were added Pd 2 (dba) 3 (5.2 g, 5.6 mmol, 0.2 eq), XantPhos (6.6 g, 11.4 mmol, 0.4 eq) and Cs2CO
  • Step 7 3-( ⁇ 3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl ⁇ methyl)-7-[(3-fluoropyridin-2- yl)oxy]-4H-1,3-benzoxazin-2-one [00325]
  • DMF 50 mL
  • Pyridine (10.0 g, 127 mmol, 10.0 eq) at room temperature.
  • Step 2 [00329] A mixture of 1-(5-fluoro-2,4-dihydroxyphenyl)ethanone (120 g, 705.30 mmol, 1.00 equiv) and 1-(2-chloro-3-fluoropyridin-4-yl)methanamine (203.86 g, 1269.55 mmol, 1.80 equiv) in MeOH (1.20 L) was stirred for overnight at 80°C under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The precipitated solids were collected by filtration and washed with MeOH (3x100 mL).
  • the resulting mixture was stirred for 3 h at 100°C under nitrogen atmosphere.
  • the mixture was cooled to room temperature and diluted with EtOAc (1 L) and water (500 mL).
  • the precipitated solids were filtered off and the filtrate was layered.
  • the aqueous layer was extracted with EtOAc (3x500 mL). The combined organic layers were dried and concentrated under reduced pressure.
  • the resulting mixture was stirred for 1 h at 0°C.
  • the solution was poured slowly to water/ice (580 mL) with stirring.
  • the precipitated solids were collected by filtration and washed with water (3x100 mL).
  • the filter cake was dried in vacuum and purified by trituration with EtOAc (200 mL).
  • the racemic mixture (18.7 g) was finally separated by SFC with the following conditions (Column: CHIRALPAK IH, 3*25 cm, 5 ⁇ m; Mobile Phase A: CO 2 , Mobile Phase B: MeOH (0.1% 2M NH3-MeOH); Flow rate: 80 mL/min; Gradient: isocratic 30% B; Wave Length: 220 nm; Retention Time 1 (min): 5.1; Retention Time 2 (min): 6; Sample Solvent: MeOH (0.1% 2M NH3-MeOH); Injection Volume: 3 mL; Number of Runs: 140).
  • Step 10 To a stirred solution of 2-chloro-3-fluoropyridine-4-carbaldehyde (210 g, 1316.28 mmol, 1.00 equiv) in methanol (2100 mL) was added NaBH 4 (49.80 g, 1316.28 mmol, 1.00 equiv) in portions at 0°C under nitrogen atmosphere. The resulting mixture was stirred for additional 1 h at room temperature. The solution was diluted with water/ice (2000 mL) and extracted with EtOAc (5x800 mL). The combined organic layers were washed with brine (1x1000 mL), dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure.
  • Step 1 3-((2-chloro-3-fluoropyridin-4-yl)methyl)-4-methyl-7-((tetrahydro-2H-pyran-2-yl)oxy)- 3,4-dihydro-2H-benzo[e][1,3]oxazin-2-one [00339] To a stirred mixture of 4-methyl-7-(oxan-2-yloxy)-3,4-dihydro-1,3-benzoxazin-2-one (10.0 g, 37 mmol, 1.0 equiv) and cesium fluoride (17.3 g, 113 mmol, 3.0 equiv) in DMF (100.0 mL) was added 4-(bromomethyl)-2-chloro-3-fluoropyridine (12.8 g, 56 mmol, 1.5 equiv) at 0°C.
  • Step 4 7-[(5-bromo-1- ⁇ [2-(trimethylsilyl)ethoxy]methyl ⁇ -1,2,4-triazol-3-yl)oxy]-3-( ⁇ 3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl ⁇ methyl)-5-methyl-4H-1,3-benzoxazin-2-one [00342]
  • Step 1 3-((2-chloro-3-fluoropyridin-4-yl)methyl)-4-methyl-7-((tetrahydro-2H-pyran-2-yl)oxy)- 3,4-dihydro-2H-benzo[e][1,3]oxazin-2-one [00347]
  • a mixture of 4-methyl-7-(oxan-2-yloxy)- 3,4-dihydro-1,3-benzoxazin-2-one (6.00 g, 22.78 mmol, 1.0 eq) and CsF (10.38 g, 68.36 mmol, 3.0 eq) in DMF (60.0 mL).
  • Step 4 7-[(5-chloro-2- ⁇ [2-(trimethylsilyl)ethoxy]methyl ⁇ -1,2,4-triazol-3-yl)oxy]-3-( ⁇ 3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl ⁇ methyl)-4-methyl-4H-1,3-benzoxazin-2-one [00350] Into a 40 mL vial was placed a mixture of 3-( ⁇ 3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl ⁇ methyl)-7-hydroxy-4-methyl-4H-1,3-benzoxazin-2-one (800 mg, 2.02 mmol, 1.0 eq), 3,5-dichloro-1- ⁇ [2-(trimethylsilyl)ethoxy]methyl ⁇ -1,2,4-triazole (811 mg, 3.02 mmol, 1.5 eq), K 2 CO 3 (836 mg, 6.05 mmol, 3 eq)
  • Step 5 7-[(5-chloro-2H-1,2,4-triazol-3-yl)oxy]-3-( ⁇ 3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl ⁇ methyl)-4-methyl-4H-1,3-benzoxazin-2-one [00351]
  • Step 7 3,5-dichloro-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-1,2,4-triazole
  • Into a 40mL vial was added a solution of 3,5-dichloro-1H-1,2,4-triazole (1.20 g, 8.67 mmol, 1.0 eq) in ACN (12.0 mL), then SEM-Cl (2.18 g, 13.05 mmol, 1.5 eq) and K 2 CO 3 (3.61 g, 26.10 mmol, 3 eq) were added. The mixture was stirred for 1h at room temperature. The resulting mixture was diluted with EtOAc (50 mL) and water (50 mL) with stirring.
  • Step 1 3,5-dimethoxypyridine-2-carbonitrile O O
  • MeONa 152.0 g, 2826 mmol, 4.0 equiv
  • the resulting mixture was stirred for 1 h at 80°C.
  • the mixture was allowed to cool down to 5°C.
  • the precipitated solids were collected by filtration and washed with MeOH (1.0 L).
  • the filter cake was purified by trituration with 1M aqueous HCl (600 mL).
  • Step 4 2-( ⁇ [(2-chloro-3-fluoropyridin-4-yl)methyl]amino ⁇ methyl)pyridine-3,5-diol [00358] To a stirred solution of [(2-chloro-3-fluoropyridin-4-yl)methyl][(3,5- dimethoxypyridin-2-yl)methyl]amine (29.8 g, 95 mmol, 1.0 equiv) in DCE (400 mL) was added BBr3 (239.5 g, 956 mmol, 10.0 equiv) dropwise at room temperature.
  • Step 5 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-hydroxy-4H-pyrido[2,3-e][1,3]oxazin-2- one [00359]
  • 2-( ⁇ [(2-chloro-3-fluoropyridin-4- yl)methyl]amino ⁇ methyl)pyridine-3,5-diol (25.3 g, 89 mmol, 1.0 equiv) and TEA (45.1 g, 446 mmol, 5.0 equiv) in THF (520.0 mL) was added CDI (17.3 g, 107 mmol, 1.2 equiv) in portions at 0°C.
  • the crude product (30.0 g) was purified by Prep-HPLC with the following conditions (Column: Welch Xtimate 150-DAC; Mobile Phase A: Water (0.1%TFA), Mobile Phase B: ACN; Flow rate: 400 ml/min; Gradient: 5% B to 40% B in 38min, 40% to 40% B in 10min;) to afford 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-hydroxy-4H-pyrido[2,3-e][1,3]oxazin-2-one (13 g, 47%) as a brown solid.
  • ES-LCMS m/z 310 [M+H] + .
  • Step 6 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-[(3-fluoropyridin-2-yl)oxy]-4H-pyrido[2,3- e][1,3]oxazin-2-one [00360] To a stirred solution of 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-hydroxy-4H- pyrido[2,3-e][1,3]oxazin-2-one (7.2 g, 23 mmol, 1.0 equiv) and 3-fluoro-2-iodopyridine (10.4 g, 46 mmol, 2.0 equiv) in DMSO (200.0 mL) were added CuI (4.4 g, 23 mmol, 1.0 equiv), 2,2,6,6- tetramethylheptane-3,5-dione (6.4 g, 34 mmol, 1.5 equiv) and K3PO4 (14.8 g,
  • the mixture was stirred for 4 h at 100°C.
  • the resulting mixture was cooled to room temperature and diluted with EtOAc (400 mL) and H 2 O (500 ml). After filtered, the filter cake was washed with EtOAc (80 mL). The filtrate was layered, the aqueous layer was extracted with EtOAc (3 x 500 mL). The combined organic layers were washed with brine (500 mL), dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure.
  • Step 7 3-( ⁇ 2-[(diphenylmethylidene)amino]-3-fluoropyridin-4-yl ⁇ methyl)-7-[(3-fluoropyridin- 2-yl)oxy]-4H-pyrido[2,3-e][1,3]oxazin-2-one [00361] To a stirred solution of 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-[(3-fluoropyridin- 2-yl)oxy]-4H-pyrido[2,3-e][1,3]oxazin-2-one (2.7 g, 6.8 mmol, 1.0 equiv), Xantphos (0.79 g, 1.4 mmol, 0.2 equiv), Pd 2 (dba) 3 (0.93 g, 1.0 mmol, 0.15 equiv) and Cs 2 CO 3 (4.4 g, 13.6 mmol, 2.0 equiv) in dioxan
  • Step 8 3-[(2-amino-3-fluoropyridin-4-yl)methyl]-7-[(3-fluoropyridin-2-yl)oxy]-4H-pyrido[2,3- e][1,3]oxazin-2-one [00362]
  • Step 9 3-( ⁇ 3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl ⁇ methyl)-7-[(3-fluoropyridin-2- yl)oxy]-4H-pyrido[2,3-e][1,3]oxazin-2-one [00363] To a stirred solution of 3-[(2-amino-3-fluoropyridin-4-yl)methyl]-7-[(3-fluoropyridin- 2-yl)oxy]-4H-pyrido[2,3-e][1,3]oxazin-2-one (3.5 g, 9.1 mmol, 1.0 equiv) and pyridine (7.2 g, 90 mmol, 10.0 equiv) in DMF (35.0 mL) was added N-methylsulfamoyl chloride (3.3 g, 25 mmol, 28 equiv) dropwise at 0°C under nitrogen atmosphere The resulting mixture was stirred for
  • Step 1 3-(benzyloxy)-5-bromopyridine-2-carbonitrile [00366] To a stirred mixture of NaH (18.95 g, 789.45 mmol, 1.20 equiv) in THF (1.10 L), benzyl alcohol (81.10 g, 749.98 mmol, 1.14 equiv) was added dropwise at 0°C. The mixture was stirred for 1 h at room temperature. To the above mixture, a solution of 5-bromo-3-nitropyridine- 2-carbonitrile (150 g, 657.88 mmol, 1.0 equiv) in THF (1.00 L) was added dropwise over 30 min at 10 ⁇ 25°C.
  • Step 4 1-[3-(benzyloxy)-5-[(3-fluoropyridin-2-yl)oxy]pyridin-2-yl]ethanamine [00369] To a stirred mixture of 3-(benzyloxy)-5-[(3-fluoropyridin-2-yl)oxy]pyridine-2- carbonitrile (10.00 g, 31.12 mmol, 1.0 equiv) in THF (100 mL) was added bromo(methyl)magnesium (7.42 g, 62.24 mmol, 2.0 equiv) dropwise at -10 °C under nitrogen atmosphere.
  • the resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere.
  • the reaction was treatment by the addition of MeOH (100 mL) at 0°C.
  • MeOH 100 mL
  • NaBH4 0.94 g, 24.89 mmol, 0.8 equiv
  • the resulting mixture was stirred for additional 10 min at room temperature.
  • the reaction was quenched by the addition of HCl in MeOH (4M, 50 mL) at 0°C.
  • the resulting mixture was filtered, the filter cake was washed with MeOH (3 x 20 mL). The filtrate was concentrated under reduced pressure.
  • Step 5 2-(1-aminoethyl)-5-((3-fluoropyridin-2-yl)oxy)pyridin-3-ol
  • BBr3 20.67 g, 82.504 mmol, 4 equiv
  • Step 6 2-(1-((2-fluoro-3-nitrobenzyl)amino)ethyl)-5-((3-fluoropyridin-2-yl)oxy)pyridin-3-ol
  • 2-(1-aminoethyl)-5-[(3-fluoropyridin-2-yl)oxy]pyridin- 3-ol 300 mg, 1.20 mmol, 1.0 equiv
  • 2-fluoro-3-nitrobenzaldehyde 200 mg, 1.20 mmol, 1.0 equiv
  • DIEA 466 mg, 3.61 mmol, 3.0 equiv
  • MeOH 4.0 mL
  • Step 8 3-(3-amino-2-fluorobenzyl)-7-((3-fluoropyridin-2-yl)oxy)-4-methyl-3,4-dihydro-2H- pyrido[2,3-e][1,3]oxazin-2-one [00373] Into a 8 mL vial were added 3-[(2-fluoro-3-nitrophenyl)methyl]-7-[(3-fluoropyridin-2- yl)oxy]-4-methyl-4H-pyrido[2,3-e][1,3]oxazin-2-one (100 mg, 0.23 mmol, 1.0 equiv), Fe (130 mg, 2.33 mmol, 10.0 equiv) and CH 3 COOH (2.0 mL) at room temperature.
  • Step 9 3-( ⁇ 3-[(ethylsulfamoyl)amino]-2-fluorophenyl ⁇ methyl)-7-[(3-fluoropyridin-2-yl)oxy]-4- methyl-4H-pyrido[2,3-e][1,3]oxazin-2-one [00374] To a stirred solution of 3-[(3-amino-2-fluorophenyl)methyl]-7-[(3-fluoropyridin-2- yl)oxy]-4-methyl-4H-pyrido[2,3-e][1,3]oxazin-2-one (60 mg, 0.15 mmol, 1.0 equiv) and pyridine (71 mg, 0.91 mmol, 6.0 equiv) in DMF (2.0 mL) was added N-ethylsulfamoyl chloride (64 mg, 0.45 mmol, 3.0 equiv) dropwise at 0°C under N2 atmosphere.
  • EXAMPLE 2 SPR Assays MEK-RAF binding assay [00377] Compound effects on binding affinity of MEK to BRAF or CRAF were followed by surface plasmon resonance (SPR) with single-cycle kinetic analysis. GST-BRAF or GST-CRAF was immobilized onto a chip and increasing concentrations of His-tagged MEK1 were flowed over the sensor chip. 500 mM ATP was added to the running buffer. The dissociation constant for MEK1 binding to either BRAF or CRAF was calculated in the absence and presence of compound. Instruments were carried out on a Biacore 8K instrument. Sensorgrams were double-reference and DMSO corrected. Data was fit to a single-cycle kinetic model. [00378] Experimental results are provided in Table 2, below.
  • the compound numbers correspond to the compound numbers in Table 1.
  • the activity is classified as A-D, wherein A represents IC50 ⁇ 100 nM; B represents 100 nM ⁇ IC50 ⁇ 1 uM; C represents 1 ⁇ IC50 ⁇ 10 uM; and D represents IC50 ⁇ 10 uM. TABLE 2.
  • MEK-KSR binding assay Compound effects on binding affinity of MEK to KSR1 or KSR2 are followed by surface plasmon resonance (SPR) with single-cycle kinetic analysis.
  • SPR surface plasmon resonance
  • GST-KSR1 or GST-KSR2 is immobilized onto a chip and increasing concentrations of His-tagged MEK1 are flowed over the sensor chip. 500 mM ATP is added to the running buffer.
  • the dissociation constant for MEK1 binding to either BRAF or CRAF is calculated in the absence and presence of compound.
  • Instruments are carried out on a Biacore 8K instrument. Sensorgrams are double-reference and DMSO corrected. Data is fit to a single-cycle kinetic model.
  • EXAMPLE 3 Immunoprecipitation Assay [00380] Immunoprecipitation experiments were performed by plating about 450,000 HCT116 cells per well in 6-well plates. Cells were plated for 48 h so as to reach approximately 70% confluency before transfection. Then, 24 h after transfection, cells were treated with vehicle (0.1% DMSO) or a compound (about 200 nM) for 1 h. Cells were washed twice in cold PBS and then transferred to a pre-chilled tube in 0.6 ml of PBS solution. Cells were spun at 1,800g in a cold centrifuge for 10 min, and supernatant was aspirated.
  • vehicle 0.1% DMSO
  • a compound about 200 nM
  • pellets were resuspended in NP-40 buffer (50 mM Tris pH 7.8, 100 mM NaCl, 0.5% (v/v) NP-40, 10% (v/v) glycerol, 1 mM EDTA) supplemented with protease and phosphatase inhibitor cocktail (Thermo Fisher, 78440) and incubated on ice for 30 min. Lysates were centrifuged for 20 min at 2,100g, and supernatants are collected. Cleared lysates were quantified using BCA reagent (Pierce, 23225), with BSA as a standard.
  • FRET Fluorescence Resonance Energy Transfer
  • ERK1/2 phosphorylation levels were normalized to total ERK1/2 protein levels in each well.
  • IC 50 values were determined by fitting a variable slope, four parameters curve to the compound concentration to normalized Phospho-ERK1/2 relationship. The activity is classified as A-D, wherein A represents IC50 ⁇ 100 nM; B represents 100 nM ⁇ IC50 ⁇ 1 uM; C represents 1 ⁇ IC50 ⁇ 10 uM; and D represents IC50 ⁇ 10 uM. Table 4.
  • A-D Biological Assay for Inhibiting Mouse Xenograft Tumor Growth
  • B represents 100 nM ⁇ IC50 ⁇ 1 uM
  • C represents 1 ⁇ IC50 ⁇ 10 uM
  • D represents IC 50 ⁇ 10 uM.
  • EXAMPLE 4 Biological Assay for Inhibiting Mouse Xenograft Tumor Growth
  • Exemplary compounds having MEK inhibitory activity can be tested, alone or in combination with a second anti-cancer agent, such as a TEAD inhibitor or an ERK5 inhibitor, for ability to inhibit tumor growth in patient-derived mouse xenograft models of pancreatic cancer and lung cancer. Assay procedures are described below.
  • mice are inoculated subcutaneously in the right flank with a primary human tumor xenograft model tumor fragment (2-3 mm 3 in diameter) for tumor development.
  • mice are inoculated with a human lung adenocarcinoma tumor model (MSCLC, ADC model LU6424) that harbors a BRAF mutation (LU6424);
  • mice are inoculated with a human pancreatic tumor model (adenosquamous carcinoma model PA6258) with a K-Ras G12D mutation (PA6258).
  • mice When mean tumor volume reaches approximately 150- 200 mm 3 , animals are randomly allocated to appropriate treatment groups. Mice are treated with one of the following: (1) Vehicle control, (2) Exemplary MEK inhibitor alone, (3) Second anti- cancer agent alone, or (4) Combination of MEK inhibitor and second anti-cancer agent. Tumors are measured twice per week using calipers.
  • EXAMPLE 5 Mouse Pharmacokinetic Studies [00387] Exemplary compounds were tested to determine their pharmacokinetic properties in mice using cassette dosing protocols. Assay procedures and results are described below. Part I – Procedures for Mouse Casette Pharmacokinetics Studies [00388] Pharmacokinetics studies are conducted using cassette dosing protocols, with test compounds administered per mouse.
  • Treatment groups are 3 mice ( ⁇ 6-8 week old, ⁇ 20-30g balb/c male mice) per route of administration.
  • Compounds are formulated in 5% DMSO and 95% PEG400 with final concentrations of 0.5 mg/mL for IV and PO dosing.
  • Compounds are dosed at 2 mg/kg for IV dosing and 5 mg/kg for PO dosing.
  • Blood samples are withdrawn at 0.083, 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, and 24 hours following IV dosing, and 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, and 24 hours following PO dosing. Blood samples are centrifuged at 4,000 g for 5 minutes at 4 °C to obtain plasma samples.
  • Plasma samples are diluted with acetonitrile containing internal standard, vortexed for 30 seconds, then centrifuged at 4,000 rpm for 15 minutes at 4 °C. The supernatant is diluted with water, and compound concentration is determined by HPLC/MS/MS compared to calibration standards of 0.5-1,000 ng/mL.
  • EXAMPLE 6 In vivo efficacy studies using human MIA PaCa-2 xenografts in mice [00389] The antitumor activity of MEKi was evaluated in vivo using a human cell line derived xenografts grown from MIA PaCa-2 cell line in immunodeficient mice.
  • the tumor cell lines are maintained in vitro as monolayer culture in medium at 37°C in an atmosphere of 5% CO 2 in air.
  • the tumor cells are routinely sub-cultured before confluence by trypsin-EDTA treatment, not to exceed 4-5 passages.
  • the cells growing in an exponential growth phase are harvested for tumor inoculation.

Abstract

The present disclosure provides MEK inhibitors, compositions thereof, and methods of using the same.

Description

MEK INHIBITORS AND USES THEREOF CROSS REFERENCED TO RELATED APPLICATIONS [0001] This application claims the priority of US provisional application 63/269,140 filed March 10, 2022, US provisional application number 63/375,864 filed September 16, 2022, and US provisional application 63/479,118 filed January 9, 2023, each of which is hereby incorporated by reference in its entirety. TECHNICAL FIELD [0002] The present disclosure relates to compounds and methods useful for inhibiting mitogen- activated protein kinase/extracellular signal-regulated kinase (MEK). The disclosure also provides pharmaceutically acceptable compositions comprising compounds of the present disclosure and methods of using said compositions in the treatment of proliferative disorders, such as cancers. BACKGROUND [0003] Activation of the p42/44 MAPK signaling pathway comprising mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase (MEK)-ERK has been implicated in the pathogenesis and progression of various cancers. The MEK-ERK pathway is often activated by mutation of upstream factors, BRAF or Ras, or by the signals of constitutively activated cell-surface receptors. Inhibition of MEK can be a promising strategy for controlling the growth of tumors, for example, the tumors associated with MEK pathway signaling. SUMMARY [0004] It has now been found that compounds of the present disclosure, and pharmaceutically acceptable compositions thereof, are effective as MEK inhibitors. In one aspect, the instant disclosure provides a compound of Formula (I):
Figure imgf000002_0001
(I), or a pharmaceutically acceptable salt thereof, wherein each variable is as defined and described herein. [0005] Another aspect of the disclosure provides a method of treating a disorder mediated by MEK in a subject. The method comprises administering a therapeutically effective amount of a compound described herein to a subject in need thereof to treat the disorder mediated by MEK, as further described in the detailed description. [0006] Another aspect of the disclosure provides a method of inhibiting MEK activity. The method comprises contacting MEK or a KSR-MEK complex or a RAF-MEK complex with an effective amount of a compound described herein, as further described in the detailed description. [0007] In some aspects, compounds of the present disclosure, and pharmaceutically acceptable compositions thereof, are useful for treating proliferative disorders, such as the cancers as described herein. DETAILED DESCRIPTION [0008] Compounds of the present disclosure, and pharmaceutical compositions thereof, are useful as MEK inhibitors. Without wishing to be bound by any particular theory, it is believed that the compounds of the present disclosure may directly engage kinase suppressor of Ras (KSR) at the MEK interface, and that KSR may remodel the prototypical allosteric pocket of the MEK inhibitor in the KSR-MEK complex. Without wishing to be bound by any particular theory, it is believed that the compounds of the present disclosure may bind the KSR-MEK complex and may disrupt the related RAF–MEK complex. 1. Definitions: [0009] When introducing elements of the different aspects as described herein, the articles "a," "an," "the," and "said" are intended to mean that there are one or more of the elements. The terms "comprising," "including," and "having" are intended to be inclusive and mean that there may be additional elements other than the listed elements. [0010] Compounds of the present disclosure include those described generally herein, and are further illustrated by the classes, subclasses, and species disclosed herein. As used herein, the following definitions shall apply unless otherwise indicated. For purposes of this disclosure, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75th Ed. Additionally, general principles of organic chemistry are described in “Organic Chemistry”, Thomas Sorrell, University Science Books, Sausalito: 1999, and “March’s Advanced Organic Chemistry”, 5th Ed., Ed.: Smith, M.B. and March, J., John Wiley & Sons, New York: 2001, the entire contents of which are hereby incorporated by reference.
[0011] The term “aliphatic” or “aliphatic group”, as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as "carbocycle," “cycloaliphatic” or “cycloalkyl”), that has a single point of attachment to the rest of the molecule. Unless otherwise specified, aliphatic groups contain 1-6 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-5 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms. In some embodiments, “cycloaliphatic” (or “carbocycle” or “cycloalkyl”) refers to a monocyclic C3-C6 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule. Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl.
[0012] As used herein, the term “bicyclic ring” or “bicyclic ring system” refers to any bicyclic ring system, i.e. , carbocyclic or heterocyclic, saturated or having one or more units of unsaturation, having one or more atoms in common between the two rings of the ring system. Thus, the term includes any permissible ring fusion, such as ortho-fused or spirocyclic. As used herein, the term “heterobicyclic” is a subset of “bicyclic” that requires that one or more heteroatoms are present in one or both rings of the bicycle. Such heteroatoms may be present at ring junctions and are optionally substituted and may be selected from nitrogen (including N oxides) oxygen sulfur (including oxidized forms such as sulfones and sulfonates), phosphorus (including oxidized forms such as phosphates), boron, etc. In some embodiments, a bicyclic group has 7-12 ring members and 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. As used herein, the term “bridged bicyclic” refers to any bicyclic ring system, i.e., carbocyclic or heterocyclic, saturated or partially unsaturated, having at least one bridge. As defined by IUPAC, a “bridge” is an unbranched chain of atoms or an atom or a valence bond connecting two bridgeheads, where a “bridgehead” is any skeletal atom of the ring system which is bonded to three or more skeletal atoms (excluding hydrogen). In some embodiments, a bridged bicyclic group has 7-12 ring members and 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Such bridged bicyclic groups are well known in the art and include those groups set forth below where each group is attached to the rest of the molecule at any substitutable carbon or nitrogen atom. Unless otherwise specified, a bridged bicyclic group is optionally substituted with one or more substituents as set forth for aliphatic groups. Additionally or alternatively, any substitutable nitrogen of a bridged bicyclic group is optionally substituted. Exemplary bicyclic rings include:
Figure imgf000005_0001
Exemplary bridged bicyclics include:
Figure imgf000005_0002
Figure imgf000006_0001
[0013] The term “lower alkyl” refers to a C1-4 straight or branched alkyl group. Exemplary lower alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and tert-butyl. [0014] The term “lower haloalkyl” refers to a C1-4 straight or branched alkyl group that is substituted with one or more halogen atoms. [0015] The term “heteroatom” means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR+ (as in N-substituted pyrrolidinyl)). [0016] The term “unsaturated”, as used herein, means that a moiety has one or more units of unsaturation. [0017] As used herein, the term “bivalent C1-8 (or C1-6) saturated or unsaturated, straight or branched, hydrocarbon chain”, refers to bivalent alkylene, alkenylene, and alkynylene chains that are straight or branched as defined herein. [0018] The term “alkylene” refers to a bivalent alkyl group. An “alkylene chain” is a polymethylene group, i.e., –(CH2)n–, wherein n is a positive integer, preferably from 1 to 6, from 1 to 4, from 1 to 3, from 1 to 2, or from 2 to 3. A substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group. [0019] The term “alkenylene” refers to a bivalent alkenyl group. A substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group. [0020] As used herein, the term “cyclopropylenyl” refers to a bivalent cyclopropyl group of the following structure:
Figure imgf000007_0001
. [0021] The term “halogen” means F, Cl, Br, or I. [0022] The term “aryl” used alone or as part of a larger moiety as in “aralkyl,” “aralkoxy,” or “aryloxyalkyl,” refers to monocyclic or bicyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members. The term “aryl” may be used interchangeably with the term “aryl ring.” In certain embodiments of the present invention, “aryl” refers to an aromatic ring system which includes, but not limited to, phenyl, biphenyl, naphthyl, anthracyl and the like, which may bear one or more substituents. Also included within the scope of the term “aryl,” as it is used herein, is a group in which an aromatic ring is fused to one or more non–aromatic rings, such as indanyl, phthalimidyl, naphthimidyl, phenanthridinyl, or tetrahydronaphthyl, and the like. [0023] The terms “heteroaryl” and “heteroar–,” used alone or as part of a larger moiety, e.g., “heteroaralkyl,” or “heteroaralkoxy,” refer to groups having 5 to 10 ring atoms, preferably 5, 6, or 9 ring atoms; having 6, 10, or 14 ^ electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms. The term “heteroatom” refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen. Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl. The terms “heteroaryl” and “heteroar–”, as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring. Nonlimiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H–quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[2,3–b]–1,4–oxazin–3(4H)–one. A heteroaryl group may be mono– or bicyclic. The term “heteroaryl” may be used interchangeably with the terms “heteroaryl ring,” “heteroaryl group,” or “heteroaromatic,” any of which terms include rings that are optionally substituted. The term “heteroaralkyl” refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted. [0024] As used herein, the terms “heterocycle,” “heterocyclyl,” “heterocyclic radical,” and “heterocyclic ring” are used interchangeably and refer to a stable 5– to 7–membered monocyclic or 7–10–membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above. When used in reference to a ring atom of a heterocycle, the term "nitrogen" includes a substituted nitrogen. As an example, in a saturated or partially unsaturated ring having 0–3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N (as in 3,4–dihydro– 2H–pyrrolyl), NH (as in pyrrolidinyl), or +NR (as in N–substituted pyrrolidinyl). [0025] A heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted. Examples of such saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenyl, pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and quinuclidinyl. The terms “heterocycle,” “heterocyclyl,” “heterocyclyl ring,” “heterocyclic group,” “heterocyclic moiety,” and “heterocyclic radical,” are used interchangeably herein, and also include groups in which a heterocyclyl ring is fused to one or more aryl, heteroaryl, or cycloaliphatic rings, such as indolinyl, 3H–indolyl, chromanyl, phenanthridinyl, or tetrahydroquinolinyl. A heterocyclyl group may be mono– or bicyclic. The term “heterocyclylalkyl” refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted. [0026] As used herein, the term “partially unsaturated” refers to a ring moiety that includes at least one double or triple bond. The term “partially unsaturated” is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined. [0027] As described herein, compounds of the invention may contain “optionally substituted” moieties. In general, the term “substituted,” whether preceded by the term “optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an “optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds. The term “stable,” as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein. [0028] Each optional substituent on a substitutable carbon is a monovalent substituent independently selected from halogen; –(CH2)0–4Rº; –(CH2)0–4ORº; -O(CH2)0-4Ro, –O–(CH2)0– 4C(O)OR°; –(CH2)0–4CH(ORº)2; –(CH2)0–4SRº; –(CH2)0–4Ph, which may be substituted with R°; –(CH2)0–4O(CH2)0–1Ph which may be substituted with R°; –CH=CHPh, which may be substituted with R°; –(CH2)0–4O(CH2)0–1-pyridyl which may be substituted with R°; –NO2; –CN; – N3; -(CH2)0–4N(Rº)2; –(CH2)0–4N(Rº)C(O)Rº; –N(Rº)C(S)Rº; –(CH2)0– 4N(Rº)C(O)NRº2; -N(Rº)C(S)NRº2; –(CH2)0–4N(Rº)C(O)ORº; – N(Rº)N(Rº)C(O)Rº; -N(Rº)N(Rº)C(O)NRº2; -N(Rº)N(Rº)C(O)ORº; –(CH2)0–4C(O)Rº; – C(S)Rº; –(CH2)0–4C(O)ORº; –(CH2)0–4C(O)SRº; -(CH2)0–4C(O)OSiRº3; –(CH2)0–4OC(O)Rº; – OC(O)(CH2)0–4SR–, SC(S)SR°; –(CH2)0–4SC(O)Rº; –(CH2)0–4C(O)NRº2; –C(S)NRº2; –C(S)SR°; –SC(S)SR°, -(CH2)0–4OC(O)NRº2; -C(O)N(ORº)Rº; –C(O)C(O)Rº; –C(O)CH2C(O)Rº; – C(NORº)Rº; -(CH2)0–4SSRº; –(CH2)0–4S(O)2Rº; –(CH2)0–4S(O)2ORº; –(CH2)0–4OS(O)2Rº; – S(O)2NRº2; –S(O)(NR°)R°; –S(O)2N=C(NRº2)2; -(CH2)0–4S(O)Rº; -N(Rº)S(O)2NRº2; – N(Rº)S(O)2Rº; –N(ORº)Rº; –C(NH)NRº2; –P(O)2Rº; -P(O)Rº2; -OP(O)Rº2; –OP(O)(ORº)2; SiRº3; –(C1–4 straight or branched alkylene)O–N(Rº)2; or –(C1–4 straight or branched alkylene)C(O)O–N(Rº)2. [0029] Each Rº is independently hydrogen, C1–6 aliphatic, –CH2Ph, –O(CH2)0–1Ph, -CH2-(5-6 membered heteroaryl ring), or a 5–6–membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R ^, taken together with their intervening atom(s), form a 3–12–membered saturated, partially unsaturated, or aryl mono– or bicyclic ring having 0– 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which may be substituted by a divalent substituent on a saturated carbon atom of R ^ selected from =O and =S; or each R ^ is optionally substituted with a monovalent substituent independently selected from halogen, – (CH2)0–2R , –(haloR ), –(CH2)0–2OH, –(CH2)0–2OR , –(CH2)0–2CH(OR )2; -O(haloR ), –CN, –N3, –(CH2)0–2C(O)R , –(CH2)0–2C(O)OH, –(CH2)0–2C(O)OR , –(CH2)0–2SR , –(CH2)0–2SH, –(CH2)0– 2NH2, –(CH2)0–2NHR , –(CH2)0–2NR 2, –NO2, –SiR 3, –OSiR 3, -C(O)SR , –(C1–4 straight or branched alkylene)C(O)OR , or –SSR . [0030] Each R is independently selected from C1–4 aliphatic, –CH2Ph, –O(CH2)0–1Ph, or a 5– 6–membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein each R is unsubstituted or where preceded by halo is substituted only with one or more halogens; or wherein an optional substituent on a saturated carbon is a divalent substituent independently selected from =O, =S, =NNR*2, =NNHC(O)R*, =NNHC(O)OR*, =NNHS(O)2R*, =NR*, =NOR*, –O(C(R*2))2–3O–, or – S(C(R* 2))2–3S–, or a divalent substituent bound to vicinal substitutable carbons of an “optionally substituted” group is –O(CR*2)2–3O–, wherein each independent occurrence of R* is selected from hydrogen, C1–6 aliphatic or an unsubstituted 5–6–membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. [0031] When R* is C1–6 aliphatic, R* is optionally substituted with halogen, – R , -(haloR ), -OH, –OR , –O(haloR ), –CN, –C(O)OH, –C(O)OR , –NH2, –NHR , –NR 2, or – NO2, wherein each R is independently selected from C1–4 aliphatic, –CH2Ph, –O(CH2)0–1Ph, or a 5–6–membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein each R is unsubstituted or where preceded by halo is substituted only with one or more halogens. [0032] An optional substituent on a substitutable nitrogen is independently –R, –NR 2, – C(O)R, –C(O)OR, –C(O)C(O)R, –C(O)CH2C(O)R, -S(O)2R, -S(O)2NR2, –C(S)NR2, – C(NH)NR2, or –N(R)S(O)2R; wherein each R is independently hydrogen, C1–6 aliphatic, unsubstituted –OPh, or an unsubstituted 5–6–membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, two independent occurrences of R, taken together with their intervening atom(s) form an unsubstituted 3–12–membered saturated, partially unsaturated, or aryl mono– or bicyclic ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; wherein when R is C1–6 aliphatic, R is optionally substituted with halogen, –R , -(haloR ), -OH, –OR , –O(haloR ), – CN, –C(O)OH, –C(O)OR , –NH2, –NHR , –NR 2, or –NO2, wherein each R is independently selected from C1–4 aliphatic, –CH2Ph, –O(CH2)0–1Ph, or a 5–6–membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein each R is unsubstituted or where preceded by halo is substituted only with one or more halogens. [0033] In some embodiments, "alkyl" as used alone or as part of a group, refers to saturated monovalent hydrocarbon radicals having straight or branched hydrocarbon chains or, in the event that at least 3 carbon atoms are present, cyclic hydrocarbons or combinations thereof and contains 1 to 20 carbon atoms (C1-C20 alkyl), 1 to 10 carbon atoms (C1-C10 alkyl), 1 to 8 carbon atoms (C1- C8 alkyl), 1 to 6 carbon atoms (C1-C4 alkyl), or 1 to 4 carbon atoms (C1-C4 alkyl). In some embodiments, alkyl radicals include methyl, ethyl, propyl, isopropyl, n- butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isoamyl, hexyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like. [0034] In some embodiments, “alkoxy” refers to —O-(alkyl), wherein “alkyl” is as defined above. [0035] In some embodiments, "cycloalkyl" as used alone or in combination, means a saturated or partially saturated monocyclic, bicyclic or tricyclic alkyl radical wherein each cyclic moiety contains from 3 to 8 carbon atoms, or from 3 to 6 carbon atoms. In some embodiments, cycloalkyl radicals include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like. [0036] In some embodiments, halogen or halo is F, Cl, Br, or I. [0037] As used herein, the term “pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1–19, incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of this disclosure include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2– hydroxy–ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2–naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3–phenylpropionate, phosphate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p–toluenesulfonate, undecanoate, valerate salts, and the like. [0038] Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(C1-C4alkyl)4 salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate. [0039] Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the disclosure. Unless otherwise stated, all tautomeric forms of the compounds of the disclosure are within the scope of the disclosure. Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this disclosure. Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present disclosure. [0040] As used herein, the term “provided compound” refers to any MEK inhibitor genus, subgenus, and/or species set forth herein. [0041] As used herein, the terms “inhibitor” or “MEK inhibitor” or “MEK antagonist” are defined as a compound that binds to and/or inhibits MEK with measurable affinity. In some embodiments, inhibition in the presence of a MEK inhibitor or a MEK antagonist is observed in a dose-dependent manner. In some embodiments, the measured signal (e.g., signaling activity or biological activity) is at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, or at least about 100% lower than the signal measured with a negative control under comparable conditions. The potency of an inhibitor is usually defined by its IC50 value (half maximal inhibitory concentration or concentration required to inhibit 50% of the agonist response). The lower the IC50 value the greater the potency of the antagonist and the lower the concentration that is required to inhibit the maximum biological response. In certain embodiments, an inhibitor has an IC50 and/or binding constant of less than about 100 ^M, less than about 50 ^M, less than about 1 ^M, less than about 500 nM, less than about 100 nM, less than about 10 nM, or less than about 1 nM. [0042] The terms “measurable affinity” and “measurably inhibit,” as used herein, means a measurable change or inhibition in MEK activity between a sample comprising a compound of the present disclosure, or composition thereof, and MEK, and an equivalent sample comprising MEK, in the absence of said compound, or composition thereof. [0043] As used herein, the term “effective amount” refers to the amount of a compound sufficient to effect beneficial or desired results (e.g., a therapeutic, ameliorative, inhibitory, or preventative result). An effective amount can be administered in one or more administrations, applications, or dosages and is not intended to be limited to a particular formulation or administration route. [0044] As used herein, the term “treating” includes any effect, e.g., lessening, reducing, modulating, ameliorating or eliminating, that results in the improvement of the condition, disease, disorder, and the like, or ameliorating a symptom thereof. In some embodiments, treatment can be administered after one or more symptoms have developed. In other embodiments, treatment can be administered in the absence of symptoms. For example, treatment can be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment can also be continued after symptoms have resolved, for example, to prevent or delay their recurrence. [0045] The phrases “disorder mediated by MEK” or “disease mediated by MEK” or “MEK- associated disease or disorder,” as used herein, refer to diseases or disorders associated with, or mediated by MEK or MEK activity. A non-limiting example of a MEK-associated disease or disorder is a MEK-associated cancer. [0046] As used herein, the term “pharmaceutical composition” refers to the combination of an active agent with a carrier, inert or active, making the composition especially suitable for diagnostic or therapeutic use in vivo or ex vivo. [0047] As used herein, the term “pharmaceutically acceptable carrier” refers to any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, emulsions (e.g., such as an oil/water or water/oil emulsions), and various types of wetting agents. The compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see e.g., Martin, Remington’s Pharmaceutical Sciences, 15th Ed., Mack Publ. Co., Easton, PA [1975]. [0048] As used herein, a “non-ATP competitive” or “ATP non-competitive” MEK inhibitor refers to an inhibitor of MEK that does not bind in the ATP pocket of MEK, or does not displace ATP from the MEK active site, and can form direct contacts when co-bound to the MEK-ATP complex. Non-ATP competitive inhibition by a compound of the invention can be confirmed by art-recognized methods such as enzymology studies, competition assays, biophysical methods, including X-ray co-crystallography. An exemplary non-ATP competitive inhibitor of the invention inhibits recombinant MEK1 or MEK2 with an IC50 of from about 1 nM to about 50 µM. In some embodiments, an exemplary non-ATP competitive inhibitor of the invention inhibits recombinant MEK1 or MEK2 with an IC50 of about 1 nM to about 1000 nM, about 1 µM to about 50 µM, about 1 µM to about 25 µM, about 1 µM to about 10 µM, about 500 nM to about 5 µM, or about 10 nM to about 500 nM. [0049] An “inhibitor pocket”, as used herein, refers to a structure formed at the interface of the interaction between MEK and KSR or BRAF or CRAF with which an inhibitor of the invention is engaged. [0050] The term “trapping,” as used herein, refers to when an inhibitor of the present disclosure binds to and stabilizes the CRAF-MEK complex in an inactive conformation. [0051] As used herein, a compound of the invention “allosterically binds an inhibitor pocket” when a compound binds outside the active site, including, for example, outside or adjacent to the ATP-binding site of a kinase. [0052] As used herein, an “inhibitor-inhibitor pocket complex” describes a species in which an inhibitor of the invention allosterically binds an inhibitor pocket formed at an interaction interface between human MEK (MEK1 or MEK2) and human Kinase Suppressor of Ras (KSR1, KSR2, or the KSR homolog BRAF or CRAF) adjacent to ATP in a physiological complex between MEK and KSR. [0053] As used herein, when a moiety on an inhibitor “engages” an amino acid residue of MEK and/or KSR and/or BRAF or CRAF in an inhibitor pocket, this interaction is detectable by X-ray crystallography, or similar structural methods, such as cryo-electron microscopy, NMR, or in silico docking, including fragment binding and computational simulations, which demonstrates that the interaction defining the engagement is a separation between the inhibitor moiety and the amino acid residue of not more than about 8 Å, including, for example, from about 2 Å to about 5 Å, or from 5 Å to about 8 Å. The distances provided herein allow for the implicit inclusion of hydrogen atoms; however, hydrogen atoms were not included in the present crystallographic models, which is appropriate unless crystals diffract to very high resolutions (i.e., better than 1.5 Angstroms). Literature surveys of drug-receptor atom pairs across all structures in the protein data bank have used 4-5 Angstrom distance cutoffs (PMID 29308120, 26517868, 19221587) to evaluate reasonable small molecule hydrophobic bonding interactions and have found that intermolecular carbon-carbon interactions similar to the trametinib-KSR contacts are among the most highly represented drug-receptor atom pairs within the protein data bank. With respect to the interactions between the inhibitors of the invention and the MEK-KSR and/or BRAF or CRAF complex, a 4 Angstrom contact is reasonable based on the nature of the trametinib-KSR interaction and precedence of known drug-receptor complexes. This contact is within the range of known contacts as defined by several independent groups (PMID 29308120, 26517868, 19221587). 2. Description of Exemplary Embodiments: [0054] In one aspect, the present disclosure provides a compound of Formula (I):
Figure imgf000016_0001
or a pharmaceutically acceptable salt thereof, wherein: Ring A and Ring B are independently an optionally substituted ring selected from a phenyl ring, a 5-6 membered heteroaromatic ring having 1-4 heteroatoms independently selected from N, O, or S, or a 8-11 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from N, O, or S; X1 is C(R21) or N; X2 is C(R22) or N; X3 is C(R23) or N; R3 and R4 are independently halogen, R, -OR, -N(R)2, -N(R)-C(O)-R, or -C(O)-N(R)2; or R3 and R4, taken together with the atom to which they attach, form C=O, or an optionally substituted 3-8 membered carbocyclic ring, or an optionally substituted 3-8 membered heterocyclic ring having 1-3 heteroatoms independently selected from N, O, or S; each of R21, R22, and R23 is independently halogen, R, -OR, -N(R)2, -N(R)-C(O)-R, or - C(O)-N(R)2; L1 is -C(R5)(R6)- or -NR-; each of R5 and R6 is independently halogen, R, -OR, or -N(R)2; or R5 and R6, together with the atom to which they attach, form an optionally substituted 3-8 membered carbocyclic ring or an optionally substituted 3-8 membered heterocyclic ring having 1-3 heteroatoms independently selected from N, O, or S; and each R is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S. [0055] As defined generally above, Ring A is an optionally substituted ring selected from a phenyl ring, a 5-6 membered heteroaromatic ring having 1-4 heteroatoms independently selected from N, O, or S, or a 8-11 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from N, O, or S. In some embodiments, Ring A is an optionally substituted ring selected from a phenyl ring, a 5 or 6 membered heteroaromatic ring having 1, 2, 3, or 4 heteroatoms independently selected from N, O, or S, or a 8, 9, 10, or 11 membered bicyclic heteroaromatic ring having 1, 2, 3, 4, or 5 heteroatoms independently selected from N, O, or S. In some embodiments, Ring A is an optionally substituted ring selected from a phenyl ring or a 6 membered heteroaromatic ring having 1-4 heteroatoms independently selected from N. In some embodiments, Ring A is an optionally substituted ring selected from a phenyl ring or a 6 membered heteroaromatic ring having 1, 2, 3, or 4 heteroatoms independently selected from N. In some embodiments, Ring A is an optionally substituted ring selected form a phenyl ring or a 5 or 6 membered heteroaromatic having 1, 2, 3, or 4 heteroatoms independent selected from N. In some embodiments, Ring A is pyridinyl, pyrimidinyl, pyrazinyl, or pyrazolyl. [0056] In some embodiments, Ring
Figure imgf000017_0001
is independently halogen, R, -OR, or -N(R)2; and R8 is halogen, R, -OR, -N(R)2,
Figure imgf000017_0003
Figure imgf000017_0002
each of R10 and R11 is independently H, optionally substituted C1-6 aliphatic, optionally substituted 3-8 membered carbocyclyl, optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S, optionally substituted phenyl, or optionally substituted 5-6 membered heteroaryl having 1-4 heteroatoms independently selected from N, O, or S, or R10 and R11 taken together with the N atom to which they attach, form optionally substituted 3-8 membered heterocyclyl having 0, 1, or 2 heteroatoms independently selected from N, O, or S in addition to the N atom to which R10 and R11 attach; and R12 is H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S. [0057] In some embodiments, each R7 is independently halogen, R, or -OR. In some embodiments, R7 is independently H, or optionally substituted C1-C6 aliphatic. In some embodiments, R7 is independently H, halogen, or C1-C6 aliphatic. In some embodiments, R7 is unsubstituted C1-C6 alkoxy, or optionally substituted C1-C6 alkoxy. In some embodiments, each R7 is independently halogen. In some embodiments, R7 is independently H or fluoro. In some embodiments, R7 is fluoro. In some embodiments, R7 is H. [0058] In some embodiments, R8 is
Figure imgf000018_0001
, , or
Figure imgf000018_0002
. [0059] In some embodiments, R8 is
Figure imgf000018_0003
, , or
Figure imgf000018_0004
; each of R10 and R11 is independently H, C1-C6 aliphatic, or 3-8 membered heterocyclyl having 1, 2, or 3 N atoms; and R12 is H, optionally substituted C1-C6 aliphatic, or optionally substituted 3-8 membered carbocyclyl. In some embodiments, R8 is unsubstituted C1-C6 alkoxy, or optionally substituted C1-C6 alkoxy. [0060] In some embodiments, each of R10 and R11 is independently hydrogen or C1-C6 alkyl. [0061] In some embodiments, R12 is hydrogen or C1-C6 alkyl. In some embodiments, R12 is C1-C6 alkyl. In some embodiments, R12 is hydrogen, methyl, ethyl, propyl, butyl, or hexyl. [0062] As defined generally above, Ring B is an optionally substituted ring selected from a phenyl ring, a 5-6 membered heteroaromatic ring having 1-4 heteroatoms independently selected from N, O, or S, or a 8-11 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from N, O, or S. [0063] In some embodiments, Ring B is an optionally substituted ring selected from a phenyl ring or a 6 membered heteroaromatic ring having 1-4 N atoms. In some embodiments, Ring B is optionally substituted pyridinyl or optionally substituted pyrimidinyl. In some embodiments, Ring B is
Figure imgf000019_0001
, , , or
Figure imgf000019_0002
, and R1 is halogen, R, -OR, or -N(R)2. In some embodiments, R1 is hydrogen or halogen. In some embodiments, R1 is fluoro. In some embodiments, R1 is R. In some embodiments, R1 is unsubstituted, or optionally substituted C1-C6 alkyl. In some embodiments, R1 is unsubstituted C1-C6 alkoxy, or optionally substituted C1-C6 alkoxy. [0064] As defined generally above, X1 is C(R21) or N. In some embodiments, X1 is C(R21). In some embodiments, X1 is N. In some embodiments, X1 is C(H). [0065] As defined generally above, X2 is C(R22) or N. In some embodiments, X2 is C(R22). In some embodiments, X2 is N. In some embodiments, X2 is C(H). In some embodiments, X2 is C(R22), wherein R22 is halogen. [0066] As defined generally above, X3 is C(R23) or N. In some embodiments, X3 is C(R23). In some embodiments, X3 is N. In some embodiments, X3 is C(H). In some embodiments, X3 is C(R23), wherein R23 is halogen. [0067] In some embodiments, R21 is halogen. In some embodiments, R21 is R. In some embodiments, R21 is H. In some embodiments, R21 is optionally substituted C1-C6 aliphatic. In some embodiments, R21 is optionally substituted C1-C6 alkyl. In some embodiments, R21 is -OR. In some embodiments, R21 is optionally substituted C1-C6 alkoxy. In some embodiments, R21 is - N(R)2. In some embodiments, R21 is -N(R)-C(O)-R. In some embodiments, R21 is -C(O)-N(R)2. [0068] In some embodiments, R22 is halogen. In some embodiments, R22 is R. In some embodiments, R22 is H. In some embodiments, R22 is optionally substituted C1-C6 aliphatic. In some embodiments, R22 is optionally substituted C1-C6 alkyl. In some embodiments, R22 is -OR. In some embodiments, R22 is optionally substituted C1-C6 alkoxy. In some embodiments, R22 is - N(R)2. In some embodiments, R22 is -N(R)-C(O)-R. In some embodiments, R22 is -C(O)-N(R)2. [0069] In some embodiments, R23 is halogen. In some embodiments, R23 is R. In some embodiments, R23 is H. In some embodiments, R23 is optionally substituted C1-C6 aliphatic. In some embodiments, R23 is optionally substituted C1-C6 alkyl. In some embodiments, R23 is -OR. In some embodiments, R23 is optionally substituted C1-C6 alkoxy. In some embodiments, R23 is - N(R)2. In some embodiments, R23 is -N(R)-C(O)-R. In some embodiments, R23 is -C(O)-N(R)2. [0070] As defined generally above, R3 and R4 are independently halogen, R, -OR, -N(R)2, - N(R)-C(O)-R, or -C(O)-N(R)2; or R3 and R4, taken together with the atom to which they attach, form C(=O), or an optionally substituted 3-8 membered carbocyclic ring, or an optionally substituted 3-8 membered heterocyclic ring having 1-3 heteroatoms independently selected from N, O, or S. In some embodiments, R3 and R4 are independently hydrogen or R, or R3 and R4, taken together with the atom to which they attach, form C(=O), each of R is independently optionally substituted C1-C6 aliphatic. In some embodiments, R3 and R4 are independently hydrogen or R, or R3 and R4, taken together with the atom to which they attach, form C(=O), each of R is independently C1-C6 aliphatic. In some embodiments, R3 is unsubstituted C1-C6 alkoxy, or optionally substituted C1-C6 alkoxy. In some embodiments, R4 is unsubstituted C1-C6 alkoxy, or optionally substituted C1-C6 alkoxy. In some embodiments, R3 and R4 are independently hydrogen, -CH3, -CH2N(CH3)2, C(=O), or –(C3H5). In some embodiments, R3 and R4 are independently hydrogen, -CH3, -CH2N(CH3)2, or –(C3H5). [0071] In some embodiments, R3 and R4 are independently R; or R3 and R4, taken together with the atom to which they attach, form C(=O), or an optionally substituted 3-8 membered carbocyclic ring, or an optionally substituted 3-8 membered heterocyclic ring having 1-3 heteroatoms independently selected from N, O, or S. In some embodiments, R3 and R4 are independently R; or R3 and R4, taken together with the atom to which they attach, form C(=O), or an optionally substituted 3-8 membered carbocyclic ring. In some embodiments, R3 and R4 are independently R; and each R is independently H, optionally substituted C1-6 aliphatic, or optionally substituted 3-8 membered carbocyclyl. In some embodiments, R3 and R4 are independently R; and each R is independently H, methyl, or cyclopropyl. In some embodiments, R3 and R4 are independently hydrogen, -CH3, or -CH2N(CH3)2. In some embodiments, R3 and R4 are independently R; and each R is independently C1-C6 aliphatic. In some embodiments, R3 and R4 are independently hydrogen or CH3. [0072] In some embodiments, R3 is -T-N(R)2, wherein T is a bivalent C1-8 saturated or unsaturated, straight or branched, hydrocarbon chain wherein one, two or three methylene units of the chain are optionally and independently replaced by -Cy-, –C(R)2–, –S–, –N(R)–, –O–, –C(O)– , –OC(O)–, –C(O)O–, –C(O)N(R)–, –N(R)C(O)–, –S(O)–, –S(O)2– or –N(R)CH2C(O)–; and -Cy- is an optionally substituted 3-8 membered bivalent carbocyclic ring, or an optionally substituted 3-8 membered bivalent heterocyclic ring having 1, 2, or 3 heteroatoms independently selected from N, O, or S. [0073] In some embodiments, R4 is -T-N(R)2, wherein T is a bivalent C1-8 saturated or unsaturated, straight or branched, hydrocarbon chain wherein one, two or three methylene units of the chain are optionally and independently replaced by -Cy-, –C(R)2–, –S–, –N(R)–, –O–, –C(O)– , –OC(O)–, –C(O)O–, –C(O)N(R)–, –N(R)C(O)–, –S(O)–, –S(O)2– or –N(R)CH2C(O)–; and -Cy- is an optionally substituted 3-8 membered bivalent carbocyclic ring, or an optionally substituted 3-8 membered bivalent heterocyclic ring having 1, 2, or 3 heteroatoms independently selected from N, O, or S. [0074] As defined generally above, L1 is -C(R5)(R6)-, wherein each of R5 and R6 is independently halogen, R, -OR, or -N(R)2; or R5 and R6, together with the atom to which they attach, form an optionally substituted 3-8 membered carbocyclic ring or an optionally substituted 3-8 membered heterocyclic ring having 1-3 heteroatoms independently selected from N, O, or S. In some embodiments, R5 is hydrogen, and R6 is R. In some embodiments, R6 is hydrogen. In some embodiments, R5 and R6 are independently optionally substituted C1-C6 aliphatic. In some embodiments, R5 and R6 are independently methyl, ethyl, propyl, butyl, or hexyl. In some embodiments, R5 is hydrogen, and R6 is methyl. In some embodiments, R5 and R6 are independently halogen or R. In some embodiments, R5 and R6 are independently hydrogen, halogen, or C1-C6 aliphatic. [0075] In some embodiments, L1 is -CH2-. In some embodiments, L1 is -C(H)(CH3)-. [0076] In some embodiments, L1 is -NR-, wherein R is as defined and described in embodiments herein. In some embodiments, L1 is -NH-. In some embodiments, L1 is -N(CH3)-. [0077] As defined generally above, each R is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S. In some embodiments, R is H. In some embodiments, R is unsubstituted, or optionally substituted C1-C6 aliphatic. In some embodiments, R is unsubstituted, or optionally substituted C1-C6 alkyl. In some embodiments, R is unsubstituted, or optionally substituted 3-8 membered carbocyclyl. In some embodiments, R is unsubstituted, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S. [0078] In some embodiments, the present disclosure provides a compound of Formula (IA):
Figure imgf000022_0001
or a pharmaceutically acceptable salt thereof, wherein each variable is as defined above and described in embodiments herein, both singly and in combination. [0079] In some embodiments, the present disclosure provides a compound of Formula (IA), or a pharmaceutically acceptable salt thereof, wherein: X1 is C(R21) or N; X2 is C(R22) or N; X3 is C(R23) or N; R3 and R4 are independently R or -OR; or R3 and R4, taken together with the atom to which they attach, form C(=O), or an optionally substituted 3-8 membered carbocyclic ring, or an optionally substituted 3-8 membered heterocyclic ring having 1-3 heteroatoms independently selected from N, O, or S; each of R21, R22, and R23 is independently halogen, R, -OR, -N(R)2, -N(R)-C(O)-R, or - C(O)-N(R)2; L1 is -C(R5)(R6)-; each of R5 and R6 is independently H, halogen, R, or -OR; and each R is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S; R7 is independently halogen, R, or -OR; R8 is halogen, R, -OR, -N(R)2,
Figure imgf000023_0001
, , or ; each of R10 and R11 is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S, optionally substituted phenyl, or optionally substituted 5-6 membered heteroaryl having 1-4 heteroatoms independently selected from N, O, or S, or R10 and R11 taken together with the N atom to which they attach, form optionally substituted 3-8 membered heterocyclyl having 0, 1, or 2 heteroatoms independently selected from N, O, or S in addition to the N atom to which R10 and R11 attach; and R12 is H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S. [0080] In some embodiments, the present disclosure provides a compound of Formula (IB):
Figure imgf000023_0002
(IB) or a pharmaceutically acceptable salt thereof, wherein each variable is as defined above and described in embodiments herein, both singly and in combination. [0081] In some embodiments, the present disclosure provides a compound of Formula (IB), or a pharmaceutically acceptable salt thereof, wherein: X1 is C(R21) or N; X2 is C(R22) or N; X3 is C(R23) or N; R3 and R4 are independently R or -OR; or R3 and R4, taken together with the atom to which they attach, form C(=O), or an optionally substituted 3-8 membered carbocyclic ring, or an optionally substituted 3-8 membered heterocyclic ring having 1-3 heteroatoms independently selected from N, O, or S; each of R21, R22, and R23 is independently halogen, R, -OR, -N(R)2, -N(R)-C(O)-R, or - C(O)-N(R)2; L1 is -C(R5)(R6)-; each of R5 and R6 is independently H, halogen, R, or -OR; each R is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S; R7 is independently halogen, R, or -OR; R8 is halogen, R, -OR, -N(R)2,
Figure imgf000024_0002
, , or
Figure imgf000024_0003
each of R10 and R11 is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S, optionally substituted phenyl, or optionally substituted 5-6 membered heteroaryl having 1-4 heteroatoms independently selected from N, O, or S, or R10 and R11 taken together with the N atom to which they attach, form optionally substituted 3-8 membered heterocyclyl having 0, 1, or 2 heteroatoms independently selected from N, O, or S in addition to the N atom to which R10 and R11 attach; and R12 is H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S. [0082] In some embodiments, the present disclosure provides a compound of Formula (IC):
Figure imgf000024_0001
or a pharmaceutically acceptable salt thereof, wherein each variable is as defined above and described in embodiments herein, both singly and in combination. [0083] In some embodiments, the present disclosure provides a compound of Formula (IC), or a pharmaceutically acceptable salt thereof, wherein: X1 is C(R21) or N; X2 is C(R22) or N; X3 is C(R23) or N; R3 and R4 are independently R or -OR; or R3 and R4, taken together with the atom to which they attach, form C(=O), or an optionally substituted 3-8 membered carbocyclic ring, or an optionally substituted 3-8 membered heterocyclic ring having 1-3 heteroatoms independently selected from N, O, or S; each of R21, R22, and R23 is independently halogen, R, -OR, -N(R)2, -N(R)-C(O)-R, or - C(O)-N(R)2; L1 is -C(R5)(R6)-; each of R5 and R6 is independently H, halogen, R, or -OR; each R is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S; R7 is independently halogen, R, or -OR; R8 is halogen, R, -OR, -N(R)2,
Figure imgf000025_0001
, , or
Figure imgf000025_0002
; each of R10 and R11 is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S, optionally substituted phenyl, or optionally substituted 5-6 membered heteroaryl having 1-4 heteroatoms independently selected from N, O, or S, or R10 and R11 taken together with the N atom to which they attach, form optionally substituted 3-8 membered heterocyclyl having 0, 1, or 2 heteroatoms independently selected from N, O, or S in addition to the N atom to which R10 and R11 attach; and R12 is H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S. [0084] In another aspect, the present disclosure provides a compound of Formula (ID):
Figure imgf000026_0001
or a pharmaceutically acceptable salt thereof, wherein each variable is as defined above and described in embodiments herein, both singly and in combination. [0085] In some embodiments, the present disclosure provides a compound of Formula (ID), or a pharmaceutically acceptable salt thereof, wherein: X1 is C(R21) or N; X2 is C(R22) or N; X3 is C(R23) or N; R3 and R4 are independently R or -OR; or R3 and R4, taken together with the atom to which they attach, form C(=O), or an optionally substituted 3-8 membered carbocyclic ring, or an optionally substituted 3-8 membered heterocyclic ring having 1-3 heteroatoms independently selected from N, O, or S; each of R21, R22, and R23 is independently halogen, R, -OR, -N(R)2, -N(R)-C(O)-R, or - C(O)-N(R)2; L1 is -C(R5)(R6)-; each of R5 and R6 is independently H, halogen, R, or -OR; R1 is H or halogen; each R is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S; R7 is independently halogen, R, -OR, or -N(R)2; R8 is halogen, R, -OR, -N(R)2,
Figure imgf000027_0002
, or
Figure imgf000027_0003
each of R10 and R11 is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S, optionally substituted phenyl, or optionally substituted 5-6 membered heteroaryl having 1-4 heteroatoms independently selected from N, O, or S, or R10 and R11 taken together with the N atom to which they attach, form optionally substituted 3-8 membered heterocyclyl having 0, 1, or 2 heteroatoms independently selected from N, O, or S in addition to the N atom to which R10 and R11 attach; and R12 is H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S. [0086] In some embodiments, the present disclosure provides a compound of Formulae (IC- 1), (IC-2), (IC-3), (IC-4) or (IC-5):
Figure imgf000027_0001
(IC-3)
Figure imgf000028_0001
(IC-5) or a pharmaceutically acceptable salt thereof, wherein each variable is as defined above and described in embodiments herein, both singly and in combination. [0087] In some embodiments, the present disclosure provides a compound of Formula (IC-1), or a pharmaceutically acceptable salt thereof, wherein: R1 is H or halogen; each of R3 and R4 is independently H or optionally substituted C1-C6 aliphatic; R22 is H or halogen; R7 is H or halogen; each of R10 and R11 is independently H or optionally substituted C1-C6 aliphatic; and R12 is H or optionally substituted C1-C6 aliphatic. [0088] In some embodiments, the present disclosure provides a compound of Formula (IC-2), or a pharmaceutically acceptable salt thereof, wherein: R1 is H or halogen; R3 is H or optionally substituted C1-C6 alkyl; R22 is H or halogen; R7 is halogen; and each of R10 and R11 is independently H or optionally substituted C1-C6 alkyl. [0089] In some embodiments, the present disclosure provides a compound of Formula (IC-3), or a pharmaceutically acceptable salt thereof, wherein: R3 is H or optionally substituted C1-C6 alkyl; R22 is H or halogen; and each of R10 and R11 is independently H or optionally substituted C1-C6 alkyl. [0090] In some embodiments, the present disclosure provides a compound of Formula (IC-4), or a pharmaceutically acceptable salt thereof, wherein: each of R3 and R4 is independently H or optionally substituted C1-C6 aliphatic; R22 is H or halogen; and each of R10 and R11 is independently H or optionally substituted C1-C6 aliphatic; and R12 is H or optionally substituted C1-C6 aliphatic. [0091] In some embodiments, the present disclosure provides a compound of Formula (IC-5), or a pharmaceutically acceptable salt thereof, wherein: R1 is H or halogen; each of R3 and R4 is independently H or optionally substituted C1-C6 aliphatic; R22 is H or halogen; and R7 is H or halogen. [0092] In another aspect, the present disclosure provides a compound of Formula (IE):
Figure imgf000029_0001
or a pharmaceutically acceptable salt thereof, wherein each variable is as defined above and described in embodiments herein, both singly and in combination. [0093] In some embodiments, the present disclosure provides a compound of Formula (IE), or a pharmaceutically acceptable salt thereof, wherein: X1 is C(R21) or N; X2 is C(R22) or N; X3 is C(R23) or N; R3 and R4 are independently R or -OR; or R3 and R4, taken together with the atom to which they attach, form C(=O), or an optionally substituted 3-8 membered carbocyclic ring, or an optionally substituted 3-8 membered heterocyclic ring having 1-3 heteroatoms independently selected from N, O, or S; each of R21, R22, and R23 is independently halogen, R, -OR, -N(R)2, -N(R)-C(O)-R, or - C(O)-N(R)2; L1 is -C(R5)(R6)-; each of R5 and R6 is independently H, halogen, R, or -OR; R1 is H, halogen, optionally substituted C1-C6 aliphatic; each R is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S; R7 is independently halogen, R, -OR, or -N(R)2; R8 is halogen, R, -OR, -N(R)2,
Figure imgf000030_0002
, , or
Figure imgf000030_0003
; each of R10 and R11 is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S, optionally substituted phenyl, or optionally substituted 5-6 membered heteroaryl having 1-4 heteroatoms independently selected from N, O, or S, or R10 and R11 taken together with the N atom to which they attach, form optionally substituted 3-8 membered heterocyclyl having 0, 1, or 2 heteroatoms independently selected from N, O, or S in addition to the N atom to which R10 and R11 attach; and R12 is H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S. [0094] In another aspect, the present disclosure provides a compound of Formula (IF):
Figure imgf000030_0001
or a pharmaceutically acceptable salt thereof, wherein each variable is as defined above and described in embodiments herein, both singly and in combination. [0095] In some embodiments, the present disclosure provides a compound of Formula (IF), or a pharmaceutically acceptable salt thereof, wherein: X1 is C(R21) or N; X2 is C(R22) or N; X3 is C(R23) or N; R3 and R4 are independently R or -OR; or R3 and R4, taken together with the atom to which they attach, form C(=O), or an optionally substituted 3-8 membered carbocyclic ring, or an optionally substituted 3-8 membered heterocyclic ring having 1-3 heteroatoms independently selected from N, O, or S; each of R21, R22, and R23 is independently halogen, R, -OR, -N(R)2, -N(R)-C(O)-R, or - C(O)-N(R)2; L1 is -C(R5)(R6)-; each of R5 and R6 is independently H, halogen, R, or -OR; R1 is H, halogen, or optionally substituted C1-C6 aliphatic; each R is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S; R7 is independently halogen, R, -OR, or -N(R)2; R8 is halogen, R, -OR, -N(R)2,
Figure imgf000031_0001
, or
Figure imgf000031_0002
; each of R10 and R11 is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S, optionally substituted phenyl, or optionally substituted 5-6 membered heteroaryl having 1-4 heteroatoms independently selected from N, O, or S, or R10 and R11 taken together with the N atom to which they attach, form optionally substituted 3-8 membered heterocyclyl having 0, 1, or 2 heteroatoms independently selected from N, O, or S in addition to the N atom to which R10 and R11 attach; and R12 is H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S. [0096] In some embodiments, the present disclosure provides a compound of Formula (II):
Figure imgf000032_0001
or a pharmaceutically acceptable salt thereof, wherein W is S or S(O)2, and each of X1, X2, X3, R3, R4, L1, Ring A and Ring B is as defined above and described in embodiments herein, both singly and in combination.
Figure imgf000032_0002
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
I-83
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
[0097] In some embodiments, the present disclosure provides a compound set forth in Table 1 above, or a pharmaceutically acceptable salt thereof. 3. Uses, Formulation, and Administration: Pharmaceutically acceptable compositions [0098] According to another embodiment, the disclosure provides a pharmaceutical composition comprising a compound of this disclosure or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle. The amount of compound in compositions of this disclosure is such that is effective to measurably inhibit MEK, or a variant or mutant thereof, in a biological sample or in a patient. In certain embodiments, the amount of compound in compositions of this disclosure is such that is effective to measurably inhibit MEK, or a variant or mutant thereof, in a biological sample or in a patient. In certain embodiments, a composition of this disclosure is formulated for administration to a patient in need of such composition. In some embodiments, a composition of this disclosure is formulated for oral administration to a patient. [0099] As used herein, the terms “subject” and “patient” are used interchangeably and refer to organisms to be treated by the methods of the present disclosure. Such organisms preferably include, but are not limited to, mammals (e.g., murines, simians, equines, bovines, porcines, canines, felines, and the like), and, most preferably, includes humans. [00100] The term “pharmaceutically acceptable carrier, adjuvant, or vehicle” refers to a non- toxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of this disclosure include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropylene-block polymers, polyethylene glycol and wool fat. [00101] A “pharmaceutically acceptable derivative” means any non-toxic salt, ester, salt of an ester or other derivative of a compound of this disclosure that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this disclosure or an active metabolite or residue thereof. [00102] As used herein, the term "active metabolite or residue thereof" means that a metabolite or residue thereof also inhibits MEK, or a variant or mutant thereof. [00103] Compositions of the present disclosure can be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term "parenteral" as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this disclosure may be aqueous or oleaginous suspension. These suspensions can be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that can be employed are water, Ringer’s solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. [00104] For this purpose, any bland fixed oil can be employed including synthetic mono- or di- glycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions can also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used surfactants, such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation. [00105] Pharmaceutically acceptable compositions of this disclosure can be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents can also be added. [00106] Alternatively, pharmaceutically acceptable compositions of this disclosure can be administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols. [00107] Pharmaceutically acceptable compositions of this disclosure can also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs. [00108] Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches can also be used. [00109] For topical applications, provided pharmaceutically acceptable compositions can be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers. Carriers for topical administration of compounds of this disclosure include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, provided pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water. [00110] For ophthalmic use, provided pharmaceutically acceptable compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride. Alternatively, for ophthalmic uses, the pharmaceutically acceptable compositions can be formulated in an ointment such as petrolatum. [00111] Pharmaceutically acceptable compositions of this disclosure can also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well- known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents. [00112] Most preferably, pharmaceutically acceptable compositions of this disclosure are formulated for oral administration. Such formulations may be administered with or without food. In some embodiments, pharmaceutically acceptable compositions of this disclosure are administered without food. In other embodiments, pharmaceutically acceptable compositions of this disclosure are administered with food. [00113] In some embodiments, the pharmaceutical compositions of this disclosure are brain- penetrant or CNS-penetrant or provide brain exposure. As used herein, the terms “brain-penetrant, “CNS-penetrant, “ or “brain exposure” refers that the compounds and pharmaceutical compositions of this disclosure are capable of crossing the blood brain barrier (BBB), and thus are useful for treating a brain or CNS disease, condition, injury or disorder. In some embodiments, a brain or CNS disease, condition, injury or disorder is a neurodegenerative diseases, neuronal injury, stroke, genetic disorders, psychiatric disorders, developmental disorders, inflammation, infection or damage, and brain cancers, spinal cord injury (SCI) and traumatic brain injury (TBI). In certain embodiments, a brain disorder is selected from epilepsy, meningitis, encephalitis including HIV Encephalitis, progressive multifocal leukoencephalopathy, neuromyelitis optica, multiple sclerosis, late-stage neurological trypanosomiasis, amyotrophic lateral sclerosis (ALS), progressive bulbar palsy (PBP), primary lateral sclerosis (PLS), progressive muscular atrophy (PMA), Alzheimer's disease, Parkinson's disease, Huntington's disease, De Vivo disease, and any type of tumor, cancer or hyperproliferative disease in the brain or CNS. [00114] In some embodiments, a brain or CNS disease, condition, injury or disorder is a neurological disorder which affects the CNS and/or which has an etiology in the CNS, which includes, but is not limited to, neuropathy, amyloidosis, cancer, an ocular disease or disorder, viral or microbial infection, inflammation, ischemia, neurodegenerative disease, seizure, behavioral disorders, and a lysosomal storage disease. For the purposes of this application, the CNS will be understood to include the eye, which is normally sequestered from the rest of the body by the blood-retina barrier. Specific examples of neurological disorders include, but are not limited to, neurodegenerative diseases (including, but not limited to, Lewy body disease, postpoliomyelitis syndrome, Shy-Draeger syndrome, olivopontocerebellar atrophy, Parkinson's disease, multiple system atrophy, striatonigral degeneration, tauopathies (including, but not limited to, Alzheimer disease and supranuclear palsy), prion diseases (including, but not limited to, bovine spongiform encephalopathy, scrapie, Creutzfeldt- Jakob syndrome, kuru, Gerstmann-Straussler-Scheinker disease, chronic wasting disease, and fatal familial insomnia), bulbar palsy, motor neuron disease, and nervous system hetero degenerative disorders (including, but not limited to, Canavan disease, Huntington's disease, neuronal ceroid- lipofuscinosis, Alexander's disease, Tourette's syndrome, Menkes kinky hair syndrome, Cockayne syndrome, Halervorden-Spatz syndrome, lafora disease, Rett syndrome, hepatolenticular degeneration, Lesch-Nyhan syndrome, and Unverricht- Lundborg syndrome), dementia (including, but not limited to, Pick's disease, and spinocerebellar ataxia), cancer (e.g. of the CNS and/or brain, including brain metastases resulting from cancer elsewhere in the body). [00115] The “central nervous system” or “CNS” refers to the complex of nerve tissues that control bodily function, and includes the brain and spinal cord. [00116] The amount of compounds of the present disclosure that can be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration. Preferably, provided compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions. [00117] It should also be understood that a specific dosage and treatment regimen for any particular patient depends upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated. The amount of a compound of the present disclosure in the composition also depends upon the particular compound in the composition. Uses of Compounds and Pharmaceutically Acceptable Compositions [00118] In some embodiments, the present disclosure provides a method of using a compound as described herein for treating a disease or disorder associated with MEK. In some embodiments, a disease or disorder associated with MEK is a proliferative disorder. In some embodiments, a disease or disorder associated with MEK is a cancer. In some embodiments, a disease or disorder associated with MEK is a cancer as described herein. [00119] In some aspects and embodiments, provided herein are methods of treating, reducing the severity of, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more symptoms thereof, of a disease or disorder characterized by or associated with increased MEK expression and/or increased MEK activity, comprising the step of administering to a patient in need thereof a therapeutically effective amount of a compound of the present disclosure, or pharmaceutically acceptable composition thereof. In some aspects and embodiments, provided herein are methods of treating, reducing the severity of, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more symptoms thereof of a disease or disorder in which inhibition or antagonizing of MEK activity is beneficial, comprising the step of administering to a patient in need thereof a therapeutically effective amount of a compound of the present disclosure, or pharmaceutically acceptable composition thereof. [00120] Accordingly, in some aspects and embodiments, the present disclosure provides a method for treating one or more disorders, diseases, and/or conditions wherein the disorder, disease, or condition includes, but is not limited to, a cellular proliferative disorder, comprising administering to a patient in need thereof, a MEK inhibitor compound as described herein, or a pharmaceutical salt or composition thereof. In some embodiments, the cellular proliferative disorder is cancer. In some embodiments, the cancer is characterized by increased MEK expression and/or increased MEK activity, i.e., “increased activated MEK.” [00121] As used herein, the terms "increased," "elevated," or "enhanced," are used interchangeably and encompass any measurable increase in a biological function and/or biological activity and/or a concentration. For example, an increase can be by at least about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, about 100%, about 2-fold, about 3-fold, about 4- fold, about 5-fold, about 6-fold, about 7-fold, about 8-fold, about 9-fold, about 10-fold, about 20- fold, about 25-fold, about 50-fold, about 100-fold, or higher, relative to a control or baseline amount of a function, or activity, or concentration. [00122] As used herein, the terms "increased expression" and/or "increased activity" of a substance, such as MEK, in a sample or cancer or patient, refers to an increase in the amount of the substance, such as MEK, of about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, about 100%, about 2-fold, about 3-fold, about 4-fold, about 5-fold, about 6-fold, about 7-fold, about 8-fold, about 9-fold, about 10-fold, about 20-fold, about 25-fold, about 50- fold, about 100-fold, or higher, relative to the amount of the substance, such as MEK, in a control sample or control samples, such as an individual or group of individuals who are not suffering from the disease or disorder (e.g., cancer) or an internal control, as determined by techniques known in the art. A subject can also be determined to have an "increased expression" or "increased activity" of MEK if the expression and/or activity of MEK is increased by one standard deviation, two standard deviations, three standard deviations, four standard deviations, five standard deviations, or more, relative to the mean (average) or median amount of MEK in a control group of samples or a baseline group of samples or a retrospective analysis of patient samples. As practiced in the art, such control or baseline expression levels can be previously determined, or measured prior to the measurement in the sample or cancer or subject, or can be obtained from a database of such control samples. Cancer [00123] In some embodiments, the present disclosure provides a method for treating or preventing or reducing the risk of a cancer in patient comprising administering to the patient a compound of the disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof. [00124] A "cancer," as used herein, refers a broad group of various diseases characterized by the uncontrolled growth of abnormal cells in the body. Unregulated cell division and growth divide and grow results in the formation of malignant tumors that invade neighboring tissues and can also metastasize to distant parts of the body through the lymphatic system or bloodstream. [00125] The cancer or proliferative disorder or tumor to be treated using the compounds and methods and uses described herein include, but are not limited to, a hematological cancer, a lymphoma, a myeloma, a leukemia, a neurological cancer, skin cancer, breast cancer, a prostate cancer, a colorectal cancer, lung cancer, head and neck cancer, a gastrointestinal cancer, a liver cancer, a pancreatic cancer, a genitourinary cancer, a bone cancer, renal cancer, and a vascular cancer. [00126] In some embodiments, the cancer is a K-Ras mutant cancer. In some embodiments, the K-Ras mutant cancer is an activated mutant K-Ras cancer. In some embodiments, the K-Ras mutant cancer is a cancer having a mutant or variant K-Ras G12. In some embodiments, the mutant or variant K-Ras G12 is K-Ras G12D, K-Ras G12V, K-Ras G12C, K-Ras G12R, K-Ras G12A, or any combination thereof. In some embodiments, the mutant or variant K-Ras G12 is K-Ras G12D. In some embodiments, the mutant or variant K-Ras G12 is K-Ras G12V. In some embodiments, the mutant or variant K-Ras G12 is K-Ras G12C. In some embodiments, the mutant or variant K- Ras G12 is K-Ras G12R. In some embodiments, the mutant or variant K-Ras G12 is K-Ras G12A. In some embodiments, the K-Ras mutant cancer is a cancer having a mutant or variant K-Ras G13. In some embodiments, the mutant or variant K-Ras G13 is K-Ras G13D, K-Ras G13C, or any combination thereof. In some embodiments, the K-Ras mutant cancer is a cancer having a mutant or variant K-Ras Q61. In some embodiments, the mutant or variant K-Ras Q61 is K-Ras Q61H, K-Ras Q61R, or any combination thereof. In some embodiments, the K-Ras mutant cancer is a cancer having a mutant or variant K-Ras A146. In some embodiments, the mutant or variant K- Ras A146 is K-Ras A146T. In some embodiments, the K-Ras mutant cancer is a cancer having a mutant or variant K-Ras G12D, K-Ras G12V, K-Ras G12C, K-Ras G12R, K-Ras G12A, K-Ras G13D, K-Ras G13C, K-Ras Q61H, K-Ras Q61R, K-Ras A146T, or any combination thereof. [00127] In some embodiments, the cancer is a mutant B-Raf cancer. In some embodiments, the mutant B-Raf cancer comprises a B-Raf amplification. As used herein, a “B-Raf amplification” refers to a cancer or cancer cell comprising at least three B-Raf copies, at least four B-Raf copies, at least five B-Raf copies, at least six B-Raf copies, at least seven B-Raf copies, at least eight B- Raf copies, at least nine B-Raf copies, at least ten B-Raf copies, or more. In some embodiments, the mutant B-Raf cancer is a cancer having a mutant or variant B-Raf V600, also referred to as a “Class I B-Raf mutation.” In some embodiments, the mutant or variant B-Raf V600 is B-Raf V600E, V600K, V600D, V600R, or any combination thereof. In some embodiments, the mutant B-Raf cancer is a cancer having a mutant or variant B-Raf K601, B-Raf P367, B-Raf G464, B-Raf L485, B-Raf E586, B-Raf T588, B-Raf T599, B-Raf L597, B-Raf G469, or any combination thereof, which are also collectively referred to as “Class II B-Raf mutations.” In some embodiments, the mutant B-Raf K601 is B-Raf K601E, K601N, K601T, or any combination thereof. In some embodiments, the mutant B-Raf P367 is B-Raf P367L, P367S, or any combination thereof. In some embodiments, the mutant B-Raf G464 is B-Raf G464V, G464E, or any combination thereof. In some embodiments, the mutant B-Raf L485 is L485W. In some embodiments, the mutant B-Raf E586 is E586K. In some embodiments, the mutant B-Raf T599 is T588TT, T588TS, T599I, T599K, or any combination thereof. In some embodiments, the mutant B-Raf L597 is B-Raf L597Q, L597R, L597S, L597V, or any combination thereof. In some embodiments, the mutant B-Raf G469 is B-Raf G469A, G469V, G469R, or any combination thereof. In some embodiments, the mutant B-Raf cancer is a cancer having a mutant or variant B- Raf D287, B-Raf V459, B-Raf G466, B-Raf S467, B-Raf G469, B-Raf N581, B-Raf D595, B-Raf F595, B-Raf G596, or any combination thereof, which are also collectively referred to as “Class III B-Raf mutations.” In some embodiments, the mutant B-Raf D287 is B-Raf D287N. In some embodiments, the mutant B-Raf V459 is B-Raf B459L. In some embodiments, the mutant B-Raf G466 is B-Raf G466A, B-Raf G466E, B-Raf G466V, or any combination thereof. In some embodiments, the mutant B-Raf S467 is B-Raf S467L. In some embodiments, the mutant B-Raf G469 is B-Raf-G469E. In some embodiments, the mutant B-Raf N581 is B-Raf N581I, B-Raf N581S, B-Raf N581T, or any combination thereof. In some embodiments, the mutant B-Raf D595 is B-Raf D595A, B-Raf D595G, B-Raf D595H, B-Raf D595N, or any combination thereof. In some embodiments, the mutant B-Raf F595 is B-Raf F595L. In some embodiments, the mutant B- Raf G596 is B-Raf G596D, B-Raf G596R, or any combination thereof. In some embodiments, the mutant B-Raf cancer comprises a B-Raf amplification and at least one B-Raf mutant or variant B- Raf Class I, Class II, or Class III mutation. [00128] In some embodiments, the cancer is an N-Ras mutant cancer. In some embodiments, the N-Ras mutant cancer is an activated mutant N-Ras cancer. In some embodiments, the N-Ras mutant cancer is a cancer having a mutant or variant N-Ras G12. In some embodiments, the mutant or variant N-Ras G12 is N-Ras G12D, N-Ras G12V, N-Ras G12C, N-Ras G12A, N-Ras G12R, N-Ras G12S, or any combination thereof. In some embodiments, the mutant or variant N-Ras G12 is N-Ras G12D. In some embodiments, the mutant or variant N-Ras G12 is N-Ras is N-Ras G12V. In some embodiments, the mutant or variant N-Ras G12 is N-Ras G12C. In some embodiments, the mutant or variant N-Ras G12 is N-Ras G12R. In some embodiments, the mutant or variant N- Ras G12 is N-Ras is N-Ras G12A. In some embodiments, the mutant or variant N-Ras G12 is N- Ras G12S. In some embodiments, the N-Ras mutant cancer is a cancer having a mutant or variant N-Ras Q61. In some embodiments, the N-Ras mutant cancer is a cancer having a mutant or variant N-Ras G12D, N-Ras G12V, N-Ras G12C, N-Ras G12R, N-Ras G12A, N-Ras G12S, N-Ras Q61, or any combination thereof. [00129] In some embodiments, the cancer is a C-Raf mutant cancer. In some embodiments, the mutant C-Raf cancer comprises a C-Raf amplification. As used herein, a “C-Raf amplification” refers to a cancer cell comprising at least three C-Raf copies, at least four C-Raf copies, at least five C-Raf copies, at least six C-Raf copies, at least seven C-Raf copies, at least eight C-Raf copies, at least nine C-Raf copies, at least ten C-Raf copies, or more. In some embodiments, the mutant C-Raf cancer is a cancer having a mutant or variant C-Raf S427. In some embodiments, the mutant or variant C-Raf S427 is C-Raf S427G. In some embodiments, the mutant C-Raf cancer is a cancer having a mutant or variant C-Raf I448. In some embodiments, the mutant C-Raf I448 is C-Raf I448V. In some embodiments, the mutant C-Raf cancer comprises a C-Raf amplification and/or at least one C-Raf mutant or variant C-Raf. [00130] In some embodiments, the cancer is an NF1 and/or NF2 mutant cancer. As used herein, an “NF1 mutant cancer” refers to a cancer having a mutant or variant gene encoding the neurofibromin protein, and includes deletion mutations, loss-of-function mutations, microdeletion mutations, missense mutations, copy number loss mutations, and substitution mutations. As used herein, an “NF2 mutant cancer” refers to a cancer having a mutant or variant gene encoding the Merlin protein (also known as schwannomin protein), and includes deletion mutations, loss-of- function mutations, microdeletion mutations, missense mutations, copy number loss mutations, and substitution mutations. [00131] In some embodiments of the methods and uses described herein, the cancer is selected from non-small cell lung cancer (NSCLC), pancreatic cancer, colorectal cancer (CRC), uterine carcinoma, endometrial carcinoma, bladder cancer, head and neck cancer, thyroid cancer, melanoma, multiple myeloma, acute myeloid leukemia (AML), low-grade serous ovarian cancer, neurofibroma, and glioma. [00132] In some embodiments of the methods and uses described herein, the cancer is a K-Ras mutant non-small cell lung cancer (NSCLC). In some embodiments of the methods and uses described herein, the cancer is a K-Ras mutant pancreatic cancer. In some embodiments of the methods and uses described herein, the cancer is a K-Ras mutant colorectal cancer. In some embodiments of the methods and uses described herein, the cancer is a K-Ras mutant uterine carcinoma. In some embodiments of the methods and uses described herein, the cancer is a K-Ras mutant endometrial carcinoma. In some embodiments of the methods and uses described herein, the cancer is a K-Ras mutant bladder cancer. In some embodiments of the methods and uses described herein, the cancer is a K-Ras mutant head and neck cancer. In some embodiments of the methods and uses described herein, the cancer is a K-Ras mutant thyroid cancer. In some embodiments of the methods and uses described herein, the cancer is a K-Ras mutant low-grade serous ovarian cancer. In some embodiments of the methods and uses described herein, the cancer is an N-Ras mutant melanoma. In some embodiments of the methods and uses described herein, the cancer is an N-Ras mutant multiple myeloma. In some embodiments of the methods and uses described herein, the cancer is an N-Ras mutant acute myeloid leukemia (AML). In some embodiments of the methods and uses described herein, the cancer is an N-Ras mutant Bladder cancer. In some embodiments of the methods and uses described herein, the cancer is a K-Ras mutant low-grade serous ovarian cancer. In some embodiments of the methods and uses described herein, the cancer is a B-Raf mutant non-small cell lung cancer (NSCLC). In some embodiments of the methods and uses described herein, the cancer is a C-Raf mutant bladder cancer. In some embodiments of the methods and uses described herein, the cancer is an NF1 mutant glioma. In some embodiments of the methods and uses described herein, the cancer is an NF1 mutant non- small cell lung cancer (NSCLC). In some embodiments of the methods and uses described herein, the cancer is an NF2 mutant neurofibroma. [00133] In some embodiments of the methods and uses described herein, the cancer is lung cancer, thyroid cancer, ovarian cancer, colorectal cancer, prostate cancer, cancer of the pancreas, cancer of the esophagus, liver cancer, breast cancer, skin cancer, or mesothelioma. In some embodiments, the cancer is mesothelioma, such as malignant mesothelioma. [00134] In some embodiments, a cancer includes, without limitation, leukemias (e.g., acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, acute myeloblastic leukemia, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute erythroleukemia, chronic leukemia, chronic myelocytic leukemia, chronic lymphocytic leukemia), polycythemia vera, lymphoma (e.g., Hodgkin’s disease or non-Hodgkin’s disease), Waldenstrom's macroglobulinemia, multiple myeloma, heavy chain disease, and solid tumors such as sarcomas and carcinomas (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing’s tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, uterine cancer, testicular cancer, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, glioblastoma multiforme (GBM, also known as glioblastoma), medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, schwannoma, neurofibrosarcoma, meningioma, melanoma, neuroblastoma, and retinoblastoma). [00135] In some embodiments, a cancer is glioma, astrocytoma, glioblastoma multiforme (GBM, also known as glioblastoma), medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, schwannoma, neurofibrosarcoma, meningioma, melanoma, neuroblastoma, or retinoblastoma. [00136] In some embodiments, a cancer is acoustic neuroma, astrocytoma (e.g. Grade I – Pilocytic Astrocytoma, Grade II – Low-grade Astrocytoma, Grade III – Anaplastic Astrocytoma, or Grade IV – Glioblastoma (GBM)), chordoma, CNS lymphoma, craniopharyngioma, brain stem glioma, ependymoma, mixed glioma, optic nerve glioma, subependymoma, medulloblastoma, meningioma, metastatic brain tumor, oligodendroglioma, pituitary tumors, primitive neuroectodermal (PNET) tumor, or schwannoma. In some embodiments, the cancer is a type found more commonly in children than adults, such as brain stem glioma, craniopharyngioma, ependymoma, juvenile pilocytic astrocytoma (JPA), medulloblastoma, optic nerve glioma, pineal tumor, primitive neuroectodermal tumors (PNET), or rhabdoid tumor. In some embodiments, the patient is an adult human. In some embodiments, the patient is a child or pediatric patient. [00137] Cancer includes, in another embodiment, without limitation, mesothelioma, hepatobilliary (hepatic and billiary duct), bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, ovarian cancer, colon cancer, rectal cancer, cancer of the anal region, stomach cancer, gastrointestinal (gastric, colorectal, and duodenal), uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin’s Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, testicular cancer, chronic or acute leukemia, chronic myeloid leukemia, lymphocytic lymphomas, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, non-Hodgkins’s lymphoma, spinal axis tumors, brain stem glioma, pituitary adenoma, adrenocortical cancer, gall bladder cancer, multiple myeloma, cholangiocarcinoma, fibrosarcoma, neuroblastoma, retinoblastoma, or a combination of one or more of the foregoing cancers. [00138] In some embodiments, a cancer is a solid tumor, such as a sarcoma, carcinoma, or lymphoma. Solid tumors generally comprise an abnormal mass of tissue that typically does not include cysts or liquid areas. In some embodiments, the cancer is selected from renal cell carcinoma, or kidney cancer; hepatocellular carcinoma (HCC) or hepatoblastoma, or liver cancer; melanoma; breast cancer; colorectal carcinoma, or colorectal cancer; colon cancer; rectal cancer; anal cancer; lung cancer, such as non-small cell lung cancer (NSCLC) or small cell lung cancer (SCLC); ovarian cancer, ovarian epithelial cancer, ovarian carcinoma, or fallopian tube cancer; papillary serous cystadenocarcinoma or uterine papillary serous carcinoma (UPSC); prostate cancer; testicular cancer; gallbladder cancer; hepatocholangiocarcinoma; soft tissue and bone synovial sarcoma; rhabdomyosarcoma; osteosarcoma; chondrosarcoma; Ewing sarcoma; anaplastic thyroid cancer; adrenocortical carcinoma; pancreatic cancer; pancreatic ductal carcinoma or pancreatic adenocarcinoma; gastrointestinal/stomach (GIST) cancer; lymphoma; squamous cell carcinoma of the head and neck (SCCHN); salivary gland cancer; glioma, or brain cancer; neurofibromatosis-1 associated malignant peripheral nerve sheath tumors (MPNST); Waldenstrom’s macroglobulinemia; or medulloblastoma. [00139] In some embodiments, a cancer is hepatocellular carcinoma (HCC). In some embodiments, the cancer is hepatoblastoma. In some embodiments, the cancer is colon cancer. In some embodiments, the cancer is rectal cancer. In some embodiments, the cancer is ovarian cancer, or ovarian carcinoma. In some embodiments, the cancer is ovarian epithelial cancer. In some embodiments, the cancer is fallopian tube cancer. In some embodiments, the cancer is papillary serous cystadenocarcinoma. In some embodiments, the cancer is uterine papillary serous carcinoma (UPSC). In some embodiments, the cancer is hepatocholangiocarcinoma. In some embodiments, the cancer is soft tissue and bone synovial sarcoma. In some embodiments, the cancer is rhabdomyosarcoma. In some embodiments, the cancer is osteosarcoma. In some embodiments, the cancer is anaplastic thyroid cancer. In some embodiments, the cancer is adrenocortical carcinoma. In some embodiments, the cancer is pancreatic cancer, or pancreatic ductal carcinoma. In some embodiments, the cancer is pancreatic adenocarcinoma. In some embodiments, the cancer is glioma. In some embodiments, the cancer is malignant peripheral nerve sheath tumors (MPNST). In some embodiments, the cancer is neurofibromatosis-1 associated MPNST. In some embodiments, the cancer is Waldenstrom’s macroglobulinemia. In some embodiments, the cancer is medulloblastoma. [00140] In some embodiments, a cancer is a viral-associated cancer, including human immunodeficiency virus (HIV) associated solid tumors, human papilloma virus (HPV)-16 positive incurable solid tumors, and adult T-cell leukemia, which is caused by human T-cell leukemia virus type I (HTLV-I) and is a highly aggressive form of CD4+ T-cell leukemia characterized by clonal integration of HTLV-I in leukemic cells (See https://clinicaltrials.gov/ct2/show/study/ NCT02631746); as well as virus-associated tumors in gastric cancer, nasopharyngeal carcinoma, cervical cancer, vaginal cancer, vulvar cancer, squamous cell carcinoma of the head and neck, and Merkel cell carcinoma. (See https://clinicaltrials.gov/ct2/show/study/NCT02488759; see also https://clinicaltrials.gov/ct2/show/study/NCT0240886; https://clinicaltrials.gov/ct2/show/ NCT02426892) [00141] In some embodiments, the methods or uses described herein inhibit or reduce or arrest or ameliorate the growth or spread of a cancer or tumor. In some embodiments, the tumor is treated by arresting, reducing, or inhibiting further growth of the cancer or tumor. In some embodiments, the methods or uses described herein increase or potentiate or activate one or more immune responses to inhibit or reduce or arrest or ameliorate the growth or spread of a cancer or tumor. In some embodiments, the cancer or tumor is treated by reducing the size (e.g., volume or mass) of the cancer or tumor by at least 5%, at least 10%, at least 25%, at least 50%, at least 75%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% relative to the size of the cancer or tumor prior to treatment. In some embodiments, cancers or tumors are treated by reducing the quantity of the cancers or tumors in the patient by at least 5%, at least 10%, at least 25%, at least 50%, at least 75%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% relative to the quantity of cancers or tumors prior to treatment. [00142] In some embodiments, a patient treated using the methods or uses described herein exhibits progression-free survival of at least about one month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months, at least about one year, at least about eighteen months, at least about two years, at least about three years, at least about four years, or at least about five years after the treatment is initiated. In some embodiments, a patient treated using the methods or uses described herein exhibits an overall survival of at least about one month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months, at least about one year, at least about 14 months, at least about 16 months, at least about 18 months, at least about 20 months, at least about 22 months, at least about two years, at least about three years, at least about four years, or at least about five years after the treatment is initiated. [00143] In some embodiments, a patient treated using the methods or uses described herein exhibits an objective response rate (ORR) of at least about 15%, at least about 20%, at least about 25%, at least about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%. [00144] The compounds and compositions, according to the method of the present disclosure, may be administered using any amount and any route of administration effective for inhibiting MEK and treating or lessening the severity of a disease, for example, as those described herein. 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 or condition, the particular agent, its mode of administration, and the like. Compounds of the disclosure are preferably formulated in dosage unit form for ease of administration and uniformity of dosage. The expression “dosage unit form” as used herein refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present disclosure will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts. The term “patient”, as used herein, means an animal, preferably a mammal, and most preferably a human. [00145] Pharmaceutically acceptable compositions of this disclosure can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the disease or disorder being treated. In certain embodiments, the compounds of the disclosure may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect. [00146] Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents. [00147] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer’s solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables. [00148] Injectable formulations can be sterilized, for example, by filtration through a bacterial- retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use. [00149] In order to prolong the effect of a compound of the present disclosure, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide- polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues. [00150] Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this disclosure with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound. [00151] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents. [00152] Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like. [00153] The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. [00154] Dosage forms for topical or transdermal administration of a compound of this disclosure include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this disclosure. Additionally, the present disclosure contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel. Co-Administration with One or More Other Therapeutic Agent(s) [00155] Depending upon the particular condition, or disease, to be treated, additional therapeutic agents that are normally administered to treat that condition, can also be present in the compositions of this disclosure. As used herein, additional therapeutic agents that are normally administered to treat a particular disease, or condition, are known as "appropriate for the disease, or condition, being treated." [00156] In some embodiments, the present disclosure provides a method of treating a disclosed disease or condition comprising administering to a patient in need thereof an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof and co-administering simultaneously or sequentially an effective amount of one or more additional therapeutic agents, such as those described herein. In some embodiments, the method includes co-administering one additional therapeutic agent. In some embodiments, the method includes co-administering two additional therapeutic agents. In some embodiments, the combination of the disclosed compound and the additional therapeutic agent or agents acts synergistically. [00157] A compound of the current disclosure can be administered alone or in combination with one or more other therapeutic compounds, possible combination therapy taking the form of fixed combinations or the administration of a compound of the disclosure and one or more other therapeutic compounds being staggered or given independently of one another, or the combined administration of fixed combinations and one or more other therapeutic compounds. [00158] One or more other therapeutic agent(s) can be administered separately from a compound or composition of the disclosure, as part of a multiple dosage regimen. Alternatively, one or more other therapeutic agent(s) may be part of a single dosage form, mixed together with a compound of this disclosure in a single composition. If administered as a multiple dosage regime, one or more other therapeutic agent(s) and a compound or composition of the disclosure can be administered simultaneously, sequentially or within a period of time from one another, for example within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours from one another. In some embodiments, one or more other therapeutic agent(s) and a compound or composition of the disclosure are administered as a multiple dosage regimen within greater than 24 hours apart. [00159] As used herein, the term "combination," "combined," and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this disclosure. For example, a compound of the present disclosure can be administered with one or more other therapeutic agent(s) simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form. Accordingly, the present disclosure provides a single unit dosage form comprising a compound of the current disclosure, one or more other therapeutic agent(s), and a pharmaceutically acceptable carrier, adjuvant, or vehicle. [00160] The amount of a compound of the disclosure and one or more other therapeutic agent(s) (in those compositions which comprise an additional therapeutic agent as described above) that can be combined with the carrier materials to produce a single dosage form varies depending upon the host treated and the particular mode of administration. Preferably, a composition of the disclosure should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of a compound of the disclosure can be administered. [00161] In those compositions which comprise one or more other therapeutic agent(s), the one or more other therapeutic agent(s) and a compound of the disclosure can act synergistically. Therefore, the amount of the one or more other therapeutic agent(s) in such compositions may be less than that required in a monotherapy utilizing only that therapeutic agent. In such compositions a dosage of between 0.01 - 1,000 g/kg body weight/day of the one or more other therapeutic agent(s) can be administered. [00162] The amount of one or more other therapeutic agent(s) present in the compositions of this disclosure may be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent. Preferably the amount of one or more other therapeutic agent(s) in the presently disclosed compositions ranges from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent. In some embodiments, one or more other therapeutic agent(s) is administered at a dosage of about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, or about 95% of the amount normally administered for that agent. As used herein, the phrase "normally administered" means the amount an FDA approved therapeutic agent is approved for dosing per the FDA label insert. [00163] The compounds of this disclosure, or pharmaceutical compositions thereof, can also be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents and catheters. Vascular stents, for example, have been used to overcome restenosis (re-narrowing of the vessel wall after injury). However, patients using stents or other implantable devices risk clot formation or platelet activation. These unwanted effects may be prevented or mitigated by pre-coating the device with a pharmaceutically acceptable composition comprising a kinase inhibitor. Implantable devices coated with a compound of this disclosure are another embodiment of the present disclosure. Exemplary Other Therapeutic Agents [00164] In some embodiments, the one or more other therapeutic agent is a TEAD inhibitor. In certain embodiments, the TEAD inhibitor is selected from those described in WO 2020/243415, the contents of which are herein incorporated by reference in their entirety. In certain embodiments, the TEAD inhibitor is selected from those described in WO 2020/243423, the contents of which are herein incorporated by reference in their entirety. In certain embodiments, the TEAD inhibitor is selected from those described in U.S. Patent No.11,274,082, the contents of which are herein incorporated by reference in their entirety. In certain embodiments, the TEAD inhibitor is selected from those described in WO 2022/120353, the contents of which are herein incorporated by reference in their entirety. In certain embodiments, the TEAD inhibitor is selected from those described in WO 2022/120354, the contents of which are herein incorporated by reference in their entirety. [00165] In certain embodiments, the second anti-cancer agent is a TEAD degrader. In certain embodiments, the TEAD degrader is selected from those described in WO 2022/120355, the contents of which are herein incorporated by reference in their entirety. [00166] TEAD inhibitors can be produced by organic synthesis methods known to one of ordinary skill in the art. Additionally, certain TEAD inhibitors can be prepared as described in Pobbati et al., "Targeting the Central Pocket in Human Transcription Factor TEAD as a Potential Cancer Therapeutic Strategy," Structure 2015, 23, 2076-2086; Gibault et al., "Targeting Transcriptional Enhanced Associate Domains (TEADs)," J. Med. Chem. 2018, 61, 5057-5072; Bum-Erdene et al., "Small-Molecule Covalent Modification of Conserved Cysteine Leads to Allosteric Inhibition of the TEADoYap Protein-Protein Interaction," Cell Chemical Biology 2019, 26, 1-12; Holden et al., "Small Molecule Dysregulation of TEAD Lipidation Induces a Dominant- Negative Inhibition of HippoPathway Signaling," Cell Reports 2020, 31, 107809; WO 2017/053706, WO 2017/111076, WO 2018/204532, WO 2018/235926, US 20190010136, WO 2019/040380, WO 2019/113236, WO 2019/222431, WO 2019/232216, WO 2020/051099, WO 2020/081572, WO 2020/097389, WO 2020/190774, WO 2020/214734, PCT/US2020/35098, and PCT/US2020/35111, the contents of each of which are herein incorporated by reference in its entirety. [00167] In some embodiments, the one or more other therapeutic agent is an ERK5 inhibitor. In some embodiments, the one or more other therapeutic agent is an ERK5 inhibitor. In certain embodiments, the ERK5 inhibitor is selected from those described in WO 2022/051567, the contents of which are herein incorporated by reference in their entirety. In certain embodiments, the ERK5 inhibitor is selected from those described in WO 2022/051565, the contents of which are herein incorporated by reference in their entirety. In certain embodiments, the ERK5 inhibitor is selected from those described in WO 2022/051569, the contents of which are herein incorporated by reference in their entirety. In certain embodiments, the ERK5 inhibitor is selected from those described in WO 2022/051568, the contents of which are herein incorporated by reference in their entirety. [00168] In some embodiments, one or more other therapeutic agent is a Poly ADP ribose polymerase (PARP) inhibitor. In some embodiments, a PARP inhibitor is selected from olaparib (LYNPARZA®, AstraZeneca); rucaparib (RUBRACA®, Clovis Oncology); niraparib (ZEJULA®, Tesaro); talazoparib (MDV3800/BMN 673/LT00673, Medivation/Pfizer/Biomarin); veliparib (ABT-888, AbbVie); and BGB-290 (BeiGene, Inc.). [00169] In some embodiments, one or more other therapeutic agent is a histone deacetylase (HDAC) inhibitor. In some embodiments, an HDAC inhibitor is selected from vorinostat (ZOLINZA®, Merck); romidepsin (ISTODAX®, Celgene); panobinostat (FARYDAK®, Novartis); belinostat (BELEODAQ®, Spectrum Pharmaceuticals); entinostat (SNDX-275, Syndax Pharmaceuticals) (NCT00866333); and chidamide (EPIDAZA®, HBI-8000, Chipscreen Biosciences, China). [00170] In some embodiments, one or more other therapeutic agent is a CDK inhibitor, such as a CDK4/CDK6 inhibitor. In some embodiments, a CDK 4/6 inhibitor is selected from palbociclib (IBRANCE®, Pfizer); ribociclib (KISQALI®, Novartis); abemaciclib (Ly2835219, Eli Lilly); and trilaciclib (G1T28, G1 Therapeutics). [00171] In some embodiments, one or more other therapeutic agent is a phosphatidylinositol 3 kinase (PI3K) inhibitor. In some embodiments, a PI3K inhibitor is selected from idelalisib (ZYDELIG®, Gilead), alpelisib (BYL719, Novartis), taselisib (GDC-0032, Genentech/Roche); pictilisib (GDC-0941, Genentech/Roche); copanlisib (BAY806946, Bayer); duvelisib (formerly IPI-145, Infinity Pharmaceuticals); PQR309 (Piqur Therapeutics, Switzerland); and TGR1202 (formerly RP5230, TG Therapeutics). [00172] In some embodiments, one or more other therapeutic agent is a platinum-based therapeutic, also referred to as platins. Platins cause cross-linking of DNA, such that they inhibit DNA repair and/or DNA synthesis, mostly in rapidly reproducing cells, such as cancer cells. [00173] In some embodiments, a platinum-based therapeutic is selected from cisplatin (PLATINOL®, Bristol-Myers Squibb); carboplatin (PARAPLATIN®, Bristol-Myers Squibb; also, Teva; Pfizer); oxaliplatin (ELOXITIN® Sanofi-Aventis); nedaplatin (AQUPLA®, Shionogi), picoplatin (Poniard Pharmaceuticals); and satraplatin (JM-216, Agennix). [00174] In some embodiments, one or more other therapeutic agent is a taxane compound, which causes disruption of microtubules, which are essential for cell division. In some embodiments, a taxane compound is selected from paclitaxel (TAXOL®, Bristol-Myers Squibb), docetaxel (TAXOTERE®, Sanofi-Aventis; DOCEFREZ®, Sun Pharmaceutical), albumin-bound paclitaxel (ABRAXANE®; Abraxis/Celgene), cabazitaxel (JEVTANA®, Sanofi-Aventis), and SID530 (SK Chemicals, Co.) (NCT00931008). [00175] In some embodiments, one or more other therapeutic agent is a nucleoside inhibitor, or a therapeutic agent that interferes with normal DNA synthesis, protein synthesis, cell replication, or will otherwise inhibit rapidly proliferating cells. [00176] In some embodiments, a nucleoside inhibitor is selected from trabectedin (guanidine alkylating agent, YONDELIS®, Janssen Oncology), mechlorethamine (alkylating agent, VALCHLOR®, Aktelion Pharmaceuticals); vincristine (ONCOVIN®, Eli Lilly; VINCASAR®, Teva Pharmaceuticals; MARQIBO®, Talon Therapeutics); temozolomide (prodrug to alkylating agent 5-(3-methyltriazen-1-yl)-imidazole-4-carboxamide (MTIC) TEMODAR®, Merck); cytarabine injection (ara-C, antimetabolic cytidine analog, Pfizer); lomustine (alkylating agent, CEENU®, Bristol-Myers Squibb; GLEOSTINE®, NextSource Biotechnology); azacitidine (pyrimidine nucleoside analog of cytidine, VIDAZA®, Celgene); omacetaxine mepesuccinate (cephalotaxine ester) (protein synthesis inhibitor, SYNRIBO®; Teva Pharmaceuticals); asparaginase Erwinia chrysanthemi (enzyme for depletion of asparagine, ELSPAR®, Lundbeck; ERWINAZE®, EUSA Pharma); eribulin mesylate (microtubule inhibitor, tubulin-based antimitotic, HALAVEN®, Eisai); cabazitaxel (microtubule inhibitor, tubulin-based antimitotic, JEVTANA®, Sanofi-Aventis); capacetrine (thymidylate synthase inhibitor, XELODA®, Genentech); bendamustine (bifunctional mechlorethamine derivative, believed to form interstrand DNA cross-links, TREANDA®, Cephalon/Teva); ixabepilone (semi-synthetic analog of epothilone B, microtubule inhibitor, tubulin-based antimitotic, IXEMPRA®, Bristol-Myers Squibb); nelarabine (prodrug of deoxyguanosine analog, nucleoside metabolic inhibitor, ARRANON®, Novartis); clorafabine (prodrug of ribonucleotide reductase inhibitor, competitive inhibitor of deoxycytidine, CLOLAR®, Sanofi-Aventis); and trifluridine and tipiracil (thymidine- based nucleoside analog and thymidine phosphorylase inhibitor, LONSURF®, Taiho Oncology). [00177] In some embodiments, one or more other therapeutic agent is a kinase inhibitor or VEGF-R antagonist. Approved VEGF inhibitors and kinase inhibitors useful in the present disclosure include: bevacizumab (AVASTIN®, Genentech/Roche) an anti-VEGF monoclonal antibody; ramucirumab (CYRAMZA®, Eli Lilly), an anti-VEGFR-2 antibody and ziv-aflibercept, also known as VEGF Trap (ZALTRAP®; Regeneron/Sanofi). VEGFR inhibitors, such as regorafenib (STIVARGA®, Bayer); vandetanib (CAPRELSA®, AstraZeneca); axitinib (INLYTA®, Pfizer); and lenvatinib (LENVIMA®, Eisai); Raf inhibitors, such as sorafenib (NEXAVAR®, Bayer AG and Onyx); dabrafenib (TAFINLAR®, Novartis); and vemurafenib (ZELBORAF®, Genentech/Roche); MEK inhibitors, such as cobimetanib (COTELLIC®, Exelexis/Genentech/Roche); trametinib (MEKINIST®, Novartis); Bcr-Abl tyrosine kinase inhibitors, such as imatinib (GLEEVEC®, Novartis); nilotinib (TASIGNA®, Novartis); dasatinib (SPRYCEL®, BristolMyersSquibb); bosutinib (BOSULIF®, Pfizer); and ponatinib (INCLUSIG®, Ariad Pharmaceuticals); Her2 and EGFR inhibitors, such as gefitinib (IRESSA®, AstraZeneca); erlotinib (TARCEEVA®, Genentech/Roche/Astellas); lapatinib (TYKERB®, Novartis); afatinib (GILOTRIF®, Boehringer Ingelheim); osimertinib (targeting activated EGFR, TAGRISSO®, AstraZeneca); and brigatinib (ALUNBRIG®, Ariad Pharmaceuticals); c-Met and VEGFR2 inhibitors, such as cabozanitib (COMETRIQ®, Exelexis); and multikinase inhibitors, such as sunitinib (SUTENT®, Pfizer); pazopanib (VOTRIENT®, Novartis); ALK inhibitors, such as crizotinib (XALKORI®, Pfizer); ceritinib (ZYKADIA®, Novartis); and alectinib (ALECENZa®, Genentech/Roche); Bruton’s tyrosine kinase inhibitors, such as ibrutinib (IMBRUVICA®, Pharmacyclics/Janssen); and Flt3 receptor inhibitors, such as midostaurin (RYDAPT®, Novartis). [00178] Other kinase inhibitors and VEGF-R antagonists that are in development and may be used in the present disclosure include tivozanib (Aveo Pharmaecuticals); vatalanib (Bayer/Novartis); lucitanib (Clovis Oncology); dovitinib (TKI258, Novartis); Chiauanib (Chipscreen Biosciences); CEP-11981 (Cephalon); linifanib (Abbott Laboratories); neratinib (HKI-272, Puma Biotechnology); radotinib (SUPECT®, IY5511, Il-Yang Pharmaceuticals, S. Korea); ruxolitinib (JAKAFI®, Incyte Corporation); PTC299 (PTC Therapeutics); CP-547,632 (Pfizer); foretinib (Exelexis, GlaxoSmithKline); quizartinib (Daiichi Sankyo) and motesanib (Amgen/Takeda). [00179] In some embodiments, one or more other therapeutic agent is an mTOR inhibitor, which inhibits cell proliferation, angiogenesis and glucose uptake. In some embodiments, an mTOR inhibitor is everolimus (AFINITOR®, Novartis); temsirolimus (TORISEL®, Pfizer); and sirolimus (RAPAMUNE®, Pfizer). [00180] In some embodiments, one or more other therapeutic agent is a proteasome inhibitor. Approved proteasome inhibitors useful in the present disclosure include bortezomib (VELCADE®, Takeda); carfilzomib (KYPROLIS®, Amgen); and ixazomib (NINLARO®, Takeda). [00181] In some embodiments, one or more other therapeutic agent is a growth factor antagonist, such as an antagonist of platelet-derived growth factor (PDGF), or epidermal growth factor (EGF) or its receptor (EGFR). Approved PDGF antagonists which may be used in the present disclosure include olaratumab (LARTRUVO®; Eli Lilly). Approved EGFR antagonists which may be used in the present disclosure include cetuximab (ERBITUX®, Eli Lilly); necitumumab (PORTRAZZA®, Eli Lilly), panitumumab (VECTIBIX®, Amgen); and osimertinib (targeting activated EGFR, TAGRISSO®, AstraZeneca). [00182] In some embodiments, one or more other therapeutic agent is an aromatase inhibitor. In some embodiments, an aromatase inhibitor is selected from exemestane (AROMASIN®, Pfizer); anastazole (ARIMIDEX®, AstraZeneca) and letrozole (FEMARA®, Novartis). [00183] In some embodiments, one or more other therapeutic agent is an antagonist of the hedgehog pathway. Approved hedgehog pathway inhibitors which may be used in the present disclosure include sonidegib (ODOMZO®, Sun Pharmaceuticals); and vismodegib (ERIVEDGE®, Genentech), both for treatment of basal cell carcinoma. [00184] In some embodiments, one or more other therapeutic agent is a folic acid inhibitor. Approved folic acid inhibitors useful in the present disclosure include pemetrexed (ALIMTA®, Eli Lilly). [00185] In some embodiments, one or more other therapeutic agent is a CC chemokine receptor 4 (CCR4) inhibitor. CCR4 inhibitors being studied that may be useful in the present disclosure include mogamulizumab (POTELIGEO®, Kyowa Hakko Kirin, Japan). [00186] In some embodiments, one or more other therapeutic agent is an isocitrate dehydrogenase (IDH) inhibitor. IDH inhibitors being studied which may be used in the present disclosure include AG120 (Celgene; NCT02677922); AG221 (Celgene, NCT02677922; NCT02577406); BAY1436032 (Bayer, NCT02746081); IDH305 (Novartis, NCT02987010). [00187] In some embodiments, one or more other therapeutic agent is an arginase inhibitor. Arginase inhibitors being studied which may be used in the present disclosure include AEB1102 (pegylated recombinant arginase, Aeglea Biotherapeutics), which is being studied in Phase 1 clinical trials for acute myeloid leukemia and myelodysplastic syndrome (NCT02732184) and solid tumors (NCT02561234); and CB-1158 (Calithera Biosciences). [00188] In some embodiments, one or more other therapeutic agent is a glutaminase inhibitor. Glutaminase inhibitors being studied which may be used in the present disclosure include CB-839 (Calithera Biosciences). [00189] In some embodiments, one or more other therapeutic agent is an antibody that binds to tumor antigens, that is, proteins expressed on the cell surface of tumor cells. Approved antibodies that bind to tumor antigens which may be used in the present disclosure include rituximab (RITUXAN®, Genentech/BiogenIdec); ofatumumab (anti-CD20, ARZERRA®, GlaxoSmithKline); obinutuzumab (anti-CD20, GAZYVA®, Genentech), ibritumomab (anti- CD20 and Yttrium-90, ZEVALIN®, Spectrum Pharmaceuticals); daratumumab (anti-CD38, DARZALEX®, Janssen Biotech), dinutuximab (anti-glycolipid GD2, UNITUXIN®, United Therapeutics); trastuzumab (anti-HER2, HERCEPTIN®, Genentech); ado-trastuzumab emtansine (anti-HER2, fused to emtansine, KADCYLA®, Genentech); and pertuzumab (anti-HER2, PERJETA®, Genentech); and brentuximab vedotin (anti-CD30-drug conjugate, ADCETRIS®, Seattle Genetics). [00190] In some embodiments, one or more other therapeutic agent is a topoisomerase inhibitor. Approved topoisomerase inhibitors useful in the present disclosure include irinotecan (ONIVYDE®, Merrimack Pharmaceuticals); topotecan (HYCAMTIN®, GlaxoSmithKline). Topoisomerase inhibitors being studied which may be used in the present disclosure include pixantrone (PIXUVRI®, CTI Biopharma). [00191] In some embodiments, one or more other therapeutic agent is an inhibitor of anti- apoptotic proteins, such as BCL-2. Approved anti-apoptotics which may be used in the present disclosure include venetoclax (VENCLEXTA®, AbbVie/Genentech); and blinatumomab (BLINCYTO®, Amgen). Other therapeutic agents targeting apoptotic proteins which have undergone clinical testing and may be used in the present disclosure include navitoclax (ABT-263, Abbott), a BCL-2 inhibitor (NCT02079740). [00192] In some embodiments, one or more other therapeutic agent is an androgen receptor inhibitor. Approved androgen receptor inhibitors useful in the present disclosure include enzalutamide (XTANDI®, Astellas/Medivation); approved inhibitors of androgen synthesis include abiraterone (ZYTIGA®, Centocor/Ortho); approved antagonist of gonadotropin-releasing hormone (GnRH) receptor (degaralix, FIRMAGON®, Ferring Pharmaceuticals). [00193] In some embodiments, one or more other therapeutic agent is a selective estrogen receptor modulator (SERM), which interferes with the synthesis or activity of estrogens. Approved SERMs useful in the present disclosure include raloxifene (EVISTA®, Eli Lilly). [00194] In some embodiments, one or more other therapeutic agent is an inhibitor of bone resorption. An approved therapeutic which inhibits bone resorption is Denosumab (XGEVA®, Amgen), an antibody that binds to RANKL, prevents binding to its receptor RANK, found on the surface of osteoclasts, their precursors, and osteoclast-like giant cells, which mediates bone pathology in solid tumors with osseous metastases. Other approved therapeutics that inhibit bone resorption include bisphosphonates, such as zoledronic acid (ZOMETA®, Novartis). [00195] In some embodiments, one or more other therapeutic agent is an inhibitor of interaction between the two primary p53 suppressor proteins, MDMX and MDM2. Inhibitors of p53 suppression proteins being studied which may be used in the present disclosure include ALRN- 6924 (Aileron), a stapled peptide that equipotently binds to and disrupts the interaction of MDMX and MDM2 with p53. ALRN-6924 is currently being evaluated in clinical trials for the treatment of AML, advanced myelodysplastic syndrome (MDS) and peripheral T-cell lymphoma (PTCL) (NCT02909972; NCT02264613). [00196] In some embodiments, one or more other therapeutic agent is an inhibitor of transforming growth factor-beta (TGF-beta or TGF-β). Inhibitors of TGF-beta proteins being studied which may be used in the present disclosure include NIS793 (Novartis), an anti-TGF-beta antibody being tested in the clinic for treatment of various cancers, including breast, lung, hepatocellular, colorectal, pancreatic, prostate and renal cancer (NCT 02947165). In some embodiments, the inhibitor of TGF-beta proteins is fresolimumab (GC1008; Sanofi-Genzyme), which is being studied for melanoma (NCT00923169); renal cell carcinoma (NCT00356460); and non-small cell lung cancer (NCT02581787). Additionally, in some embodiments, the additional therapeutic agent is a TGF-beta trap, such as described in Connolly et al. (2012) Int’l J. Biological Sciences 8:964-978. One therapeutic compound currently in clinical trials for treatment of solid tumors is M7824 (Merck KgaA - formerly MSB0011459X), which is a bispecific, anti-PD- L1/TGF-β trap compound (NCT02699515); and (NCT02517398). M7824 is comprised of a fully human IgG1 antibody against PD-L1 fused to the extracellular domain of human TGF-beta receptor II, which functions as a TGF-β “trap.” [00197] In some embodiments, one or more other therapeutic agent is selected from glembatumumab vedotin-monomethyl auristatin E (MMAE) (Celldex), an anti-glycoprotein NMB (gpNMB) antibody (CR011) linked to the cytotoxic MMAE. gpNMB is a protein overexpressed by multiple tumor types associated with cancer cells’ ability to metastasize. [00198] In some embodiments, one or more other therapeutic agents is an antiproliferative compound. Such antiproliferative compounds include, but are not limited to aromatase inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase II inhibitors; microtubule active compounds; alkylating compounds; histone deacetylase inhibitors; compounds which induce cell differentiation processes; cyclooxygenase inhibitors; MMP inhibitors; mTOR inhibitors; antineoplastic antimetabolites; platin compounds; compounds targeting/decreasing a protein or lipid kinase activity and further anti-angiogenic compounds; compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase; gonadorelin agonists; anti-androgens; methionine aminopeptidase inhibitors; matrix metalloproteinase inhibitors; bisphosphonates; biological response modifiers; antiproliferative antibodies; heparanase inhibitors; inhibitors of Ras oncogenic isoforms; telomerase inhibitors; proteasome inhibitors; compounds used in the treatment of hematologic malignancies; compounds which target, decrease or inhibit the activity of Flt-3; Hsp90 inhibitors such as 17-AAG (17-allylaminogeldanamycin, NSC330507), 17- DMAG (17-dimethylaminoethylamino-17-demethoxy-geldanamycin, NSC707545), IPI-504, CNF1010, CNF2024, CNF1010 from Conforma Therapeutics; temozolomide (TEMODAL®); kinesin spindle protein inhibitors, such as SB715992 or SB743921 from GlaxoSmithKline, or pentamidine/chlorpromazine from CombinatoRx; MEK inhibitors such as ARRY142886 from Array BioPharma, AZd6244 from AstraZeneca, PD181461 from Pfizer and leucovorin. [00199] The term “aromatase inhibitor” as used herein relates to a compound which inhibits estrogen production, for instance, the conversion of the substrates androstenedione and testosterone to estrone and estradiol, respectively. The term includes, but is not limited to steroids, especially atamestane, exemestane and formestane and, in particular, non-steroids, especially aminoglutethimide, roglethimide, pyridoglutethimide, trilostane, testolactone, ketokonazole, vorozole, fadrozole, anastrozole and letrozole. Exemestane is marketed under the trade name AROMASIN™. Formestane is marketed under the trade name LENTARON™. Fadrozole is marketed under the trade name AFEMA™. Anastrozole is marketed under the trade name ARIMIDEX™. Letrozole is marketed under the trade names FEMARA™ or FEMAr™. Aminoglutethimide is marketed under the trade name ORIMETEN™. A combination of the disclosure comprising a chemotherapeutic agent which is an aromatase inhibitor is particularly useful for the treatment of hormone receptor positive tumors, such as breast tumors. [00200] The term "antiestrogen" as used herein relates to a compound which antagonizes the effect of estrogens at the estrogen receptor level. The term includes, but is not limited to tamoxifen, fulvestrant, raloxifene and raloxifene hydrochloride. Tamoxifen is marketed under the trade name NOLVADEX™. Raloxifene hydrochloride is marketed under the trade name EVISTA™. Fulvestrant can be administered under the trade name FASLODEX™Fulvestrant can be administered under the trade name Faslodex™. A combination of the disclosure comprising a chemotherapeutic agent which is an antiestrogen is particularly useful for the treatment of estrogen receptor positive tumors, such as breast tumors. [00201] The term "anti-androgen" as used herein relates to any substance which is capable of inhibiting the biological effects of androgenic hormones and includes, but is not limited to, bicalutamide (CASODEX™). The term "gonadorelin agonist" as used herein includes, but is not limited to abarelix, goserelin, and goserelin acetate. Goserelin can be administered under the trade name ZOLADEX™. [00202] The term "topoisomerase I inhibitor" as used herein includes, but is not limited to topotecan, gimatecan, irinotecan, camptothecian and its analogues, 9-nitrocamptothecin and the macromolecular camptothecin conjugate PNU-166148. Irinotecan can be administered, e.g., in the form as it is marketed, e.g., under the trademark CAMPTOSAR™. Topotecan is marketed under the trade name HYCAMPTIN™. [00203] The term "topoisomerase II inhibitor" as used herein includes, but is not limited to the anthracyclines such as doxorubicin (including liposomal formulation, such as CAELYX™), daunorubicin, epirubicin, idarubicin and nemorubicin, the anthraquinones mitoxantrone and losoxantrone, and the podophillotoxines etoposide and teniposide. Etoposide is marketed under the trade name ETOPOPHOS™. Teniposide is marketed under the trade name VM 26-Bristol Doxorubicin is marketed under the trade name ACRIBLASTIN™ or ADRIAMYCIN™. Epirubicin is marketed under the trade name FARMORUBICIN™. Idarubicin is marketed. under the trade name ZAVEDOS™. Mitoxantrone is marketed under the trade name NOVANTRON™. [00204] The term "microtubule active agent" relates to microtubule stabilizing, microtubule destabilizing compounds and microtublin polymerization inhibitors including, but not limited to taxanes, such as paclitaxel and docetaxel; vinca alkaloids, such as vinblastine or vinblastine sulfate, vincristine or vincristine sulfate, and vinorelbine; discodermolides; cochicine and epothilones and derivatives thereof. Paclitaxel is marketed under the trade name TAXOL™. Docetaxel is marketed under the trade name TAXOTERE™. Vinblastine sulfate is marketed under the trade name VINBLASTIN R.P™. Vincristine sulfate is marketed under the trade name FARMISTIN™. [00205] The term "alkylating agent" as used herein includes, but is not limited to, cyclophosphamide, ifosfamide, melphalan or nitrosourea (BCNU or Gliadel). Cyclophosphamide is marketed under the trade name CYCLOSTIN™. Ifosfamide is marketed under the trade name HOLOXAN™. [00206] The term "histone deacetylase inhibitors" or "HDAC inhibitors" relates to compounds which inhibit the histone deacetylase and which possess antiproliferative activity. This includes, but is not limited to, suberoylanilide hydroxamic acid (SAHA). [00207] The term "antineoplastic antimetabolite" includes, but is not limited to, 5-fluorouracil or 5-FU, capecitabine, gemcitabine, DNA demethylating compounds, such as 5-azacytidine and decitabine, methotrexate and edatrexate, and folic acid antagonists such as pemetrexed. Capecitabine is marketed under the trade name XELODA™. Gemcitabine is marketed under the trade name GEMZAR™. [00208] The term "platin compound" as used herein includes, but is not limited to, carboplatin, cis-platin, cisplatinum and oxaliplatin. Carboplatin can be administered, e.g., in the form as it is marketed, e.g., under the trademark CARBOPLAT™. Oxaliplatin can be administered, e.g., in the form as it is marketed, e.g. under the trademark ELOXATIN™. [00209] The term "compounds targeting/decreasing a protein or lipid kinase activity; or a protein or lipid phosphatase activity; or further anti-angiogenic compounds" as used herein includes, but is not limited to, protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, such as a) compounds targeting, decreasing or inhibiting the activity of the platelet-derived growth factor-receptors (PDGFR), such as compounds which target, decrease or inhibit the activity of PDGFR, especially compounds which inhibit the PDGF receptor, such as an N-phenyl-2-pyrimidine-amine derivative, such as imatinib, SU101, SU6668 and GFB- 111; b) compounds targeting, decreasing or inhibiting the activity of the fibroblast growth factor- receptors (FGFR); c) compounds targeting, decreasing or inhibiting the activity of the insulin-like growth factor receptor I (IGF-IR), such as compounds which target, decrease or inhibit the activity of IGF-IR, especially compounds which inhibit the kinase activity of IGF-I receptor, or antibodies that target the extracellular domain of IGF-I receptor or its growth factors; d) compounds targeting, decreasing or inhibiting the activity of the Trk receptor tyrosine kinase family, or ephrin B4 inhibitors; e) compounds targeting, decreasing or inhibiting the activity of the AxI receptor tyrosine kinase family; f) compounds targeting, decreasing or inhibiting the activity of the Ret receptor tyrosine kinase; g) compounds targeting, decreasing or inhibiting the activity of the Kit/SCFR receptor tyrosine kinase, such as imatinib; h) compounds targeting, decreasing or inhibiting the activity of the C-kit receptor tyrosine kinases, which are part of the PDGFR family, such as compounds which target, decrease or inhibit the activity of the c-Kit receptor tyrosine kinase family, especially compounds which inhibit the c-Kit receptor, such as imatinib; i) compounds targeting, decreasing or inhibiting the activity of members of the c-Abl family, their gene-fusion products (e.g., BCR-Abl kinase) and mutants, such as compounds which target decrease or inhibit the activity of c-Abl family members and their gene fusion products, such as an N-phenyl-2-pyrimidine-amine derivative, such as imatinib or nilotinib (AMN107); PD180970; AG957; NSC 680410; PD173955 from ParkeDavis; or dasatinib (BMS-354825); j) compounds targeting, decreasing or inhibiting the activity of members of the protein kinase C (PKC) and Raf family of serine/threonine kinases, members of the MEK, SRC, JAK/pan-JAK, FAK, PDK1, PKB/Akt, Ras/MAPK, PI3K, SYK, TYK2, BTK and TEC family, and/or members of the cyclin- dependent kinase family (CDK) including staurosporine derivatives, such as midostaurin; examples of further compounds include UCN-01, safingol, BAY 43-9006, Bryostatin 1, Perifosine; llmofosine; RO 318220 and RO 320432; GO 6976; lsis 3521; LY333531/LY379196; isochinoline compounds; FTIs; PD184352 or QAN697 (a P13K inhibitor) or AT7519 (CDK inhibitor); k) compounds targeting, decreasing or inhibiting the activity of protein-tyrosine kinase inhibitors, such as compounds which target, decrease or inhibit the activity of protein-tyrosine kinase inhibitors include imatinib mesylate (GLEEVEC™) or tyrphostin such as Tyrphostin A23/RG-50810; AG 99; Tyrphostin AG 213; Tyrphostin AG 1748; Tyrphostin AG 490; Tyrphostin B44; Tyrphostin B44 (+) enantiomer; Tyrphostin AG 555; AG 494; Tyrphostin AG 556, AG957 and adaphostin (4-{[(2,5- dihydroxyphenyl)methyl]amino}-benzoic acid adamantyl ester; NSC 680410, adaphostin); l) compounds targeting, decreasing or inhibiting the activity of the epidermal growth factor family of receptor tyrosine kinases (EGFR1 ErbB2, ErbB3, ErbB4 as homo- or heterodimers) and their mutants, such as compounds which target, decrease or inhibit the activity of the epidermal growth factor receptor family are especially compounds, proteins or antibodies which inhibit members of the EGF receptor tyrosine kinase family, such as EGF receptor, ErbB2, ErbB3 and ErbB4 or bind to EGF or EGF related ligands, CP 358774, ZD 1839, ZM 105180; trastuzumab (HERCEPTIN™), cetuximab (ERBITUX™), Iressa, Tarceva, OSI-774, Cl-1033, EKB-569, GW-2016, E1.1, E2.4, E2.5, E6.2, E6.4, E2.11, E6.3 or E7.6.3, and 7H- pyrrolo-[2,3-d]pyrimidine derivatives; m) compounds targeting, decreasing or inhibiting the activity of the c-Met receptor, such as compounds which target, decrease or inhibit the activity of c-Met, especially compounds which inhibit the kinase activity of c-Met receptor, or antibodies that target the extracellular domain of c-Met or bind to HGF, n) compounds targeting, decreasing or inhibiting the kinase activity of one or more JAK family members (JAK1/JAK2/JAK3/TYK2 and/or pan-JAK), including but not limited to PRT-062070, SB-1578, baricitinib, pacritinib, momelotinib, VX-509, AZD-1480, TG-101348, tofacitinib, and ruxolitinib; o) compounds targeting, decreasing or inhibiting the kinase activity of PI3 kinase (PI3K) including but not limited to ATU-027, SF-1126, DS-7423, PBI-05204, GSK-2126458, ZSTK-474, buparlisib, pictrelisib, PF-4691502, BYL-719, dactolisib, XL-147, XL-765, and idelalisib; and; and q) compounds targeting, decreasing or inhibiting the signaling effects of hedgehog protein (Hh) or smoothened receptor (SMO) pathways, including but not limited to cyclopamine, vismodegib, itraconazole, erismodegib, and IPI-926 (saridegib). [00210] The term “PI3K inhibitor” as used herein includes, but is not limited to compounds having inhibitory activity against one or more enzymes in the phosphatidylinositol-3-kinase family, including, but not limited to PI3Kα, PI3Kγ, PI3Kδ, PI3Kβ, PI3K-C2α, PI3K-C2β, PI3K- C2γ, Vps34, p110-α, p110-β, p110-γ, p110-δ, p85-α, p85-β, p55-γ, p150, p101, and p87. Examples of PI3K inhibitors useful in this disclosure include but are not limited to ATU-027, SF-1126, DS- 7423, PBI-05204, GSK-2126458, ZSTK-474, buparlisib, pictrelisib, PF-4691502, BYL-719, dactolisib, XL-147, XL-765, and idelalisib. [00211] The term “Bcl-2 inhibitor” as used herein includes, but is not limited to compounds having inhibitory activity against B-cell lymphoma 2 protein (Bcl-2), including but not limited to ABT-199, ABT-731, ABT-737, apogossypol, Ascenta’s pan-Bcl-2 inhibitors, curcumin (and analogs thereof), dual Bcl-2/Bcl-xL inhibitors (Infinity Pharmaceuticals/Novartis Pharmaceuticals), Genasense (G3139), HA14-1 (and analogs thereof; see WO2008118802), navitoclax (and analogs thereof, see US7390799), NH-1 (Shenayng Pharmaceutical University), obatoclax (and analogs thereof, see WO2004106328), S-001 (Gloria Pharmaceuticals), TW series compounds (Univ. of Michigan), and venetoclax. In some embodiments the Bcl-2 inhibitor is a small molecule therapeutic. In some embodiments the Bcl-2 inhibitor is a peptidomimetic. [00212] The term “BTK inhibitor” as used herein includes, but is not limited to compounds having inhibitory activity against Bruton’s Tyrosine Kinase (BTK), including, but not limited to AVL-292 and ibrutinib. [00213] The term “SYK inhibitor” as used herein includes, but is not limited to compounds having inhibitory activity against spleen tyrosine kinase (SYK), including but not limited to PRT- 062070, R-343, R-333, Excellair, PRT-062607, and fostamatinib. [00214] Further examples of BTK inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this disclosure can be found in WO2008039218 and WO2011090760, the entirety of which are incorporated herein by reference. [00215] Further examples of SYK inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this disclosure can be found in WO2003063794, WO2005007623, and WO2006078846, the entirety of which are incorporated herein by reference. [00216] Further examples of PI3K inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this disclosure can be found in WO2004019973, WO2004089925, WO2007016176, US8138347, WO2002088112, WO2007084786, WO2007129161, WO2006122806, WO2005113554, and WO2007044729 the entirety of which are incorporated herein by reference. [00217] Further examples of JAK inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this disclosure can be found in WO2009114512, WO2008109943, WO2007053452, WO2000142246, and WO2007070514, the entirety of which are incorporated herein by reference. [00218] Further anti-angiogenic compounds include compounds having another mechanism for their activity, e.g., unrelated to protein or lipid kinase inhibition e.g., thalidomide (THALOMID™) and TNP-470. [00219] Examples of proteasome inhibitors useful for use in combination with compounds of the disclosure include, but are not limited to bortezomib, disulfiram, epigallocatechin-3-gallate (EGCG), salinosporamide A, carfilzomib, ONX-0912, CEP-18770, and MLN9708. [00220] Compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase are e.g., inhibitors of phosphatase 1, phosphatase 2A, or CDC25, such as okadaic acid or a derivative thereof. [00221] Compounds which induce cell differentiation processes include, but are not limited to, retinoic acid, α- γ- or δ- tocopherol or α- γ- or δ-tocotrienol. [00222] The term cyclooxygenase inhibitor as used herein includes, but is not limited to, Cox- 2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acid and derivatives, such as celecoxib ( [00223] CELEBREX™), rofecoxib (VIOXX™), etoricoxib, valdecoxib or a 5-alkyl-2- arylaminophenylacetic acid, such as 5-methyl-2-(2'-chloro-6'-fluoroanilino)phenyl acetic acid, lumiracoxib. [00224] The term "bisphosphonates" as used herein includes, but is not limited to, etridonic, clodronic, tiludronic, pamidronic, alendronic, ibandronic, risedronic and zoledronic acid. Etridonic acid is marketed under the trade name DIDRONEL™. Clodronic acid is marketed under the trade name BONEFOS™. Tiludronic acid is marketed under the trade name Skelid™. Pamidronic acid is marketed under the trade name AREDIA™. Alendronic acid is marketed under the trade name FOSAMAX™. Ibandronic acid is marketed under the trade name BONDRANAT™. Risedronic acid is marketed under the trade name ACTONEL™. Zoledronic acid is marketed under the trade name ZOMETA™. The term "mTOR inhibitors" relates to compounds which inhibit the mammalian target of rapamycin (mTOR) and which possess antiproliferative activity such as sirolimus (RAPAMUNE®), everolimus (CERTICAN™), CCI-779 and ABT578. [00225] The term "heparanase inhibitor" as used herein refers to compounds which target, decrease or inhibit heparin sulfate degradation. The term includes, but is not limited to, PI-88. The term "biological response modifier" as used herein refers to a lymphokine or interferons. [00226] The term "inhibitor of Ras oncogenic isoforms", such as H-Ras, K-Ras, or N-Ras, as used herein refers to compounds which target, decrease or inhibit the oncogenic activity of Ras; for example, a "farnesyl transferase inhibitor" such as L-744832, DK8G557 or R115777 (ZARNESTRA ™). The term "telomerase inhibitor" as used herein refers to compounds which target, decrease or inhibit the activity of telomerase. Compounds which target, decrease or inhibit the activity of telomerase are especially compounds which inhibit the telomerase receptor, such as telomestatin. [00227] The term "methionine aminopeptidase inhibitor" as used herein refers to compounds which target, decrease or inhibit the activity of methionine aminopeptidase. Compounds which target, decrease or inhibit the activity of methionine aminopeptidase include, but are not limited to, bengamide or a derivative thereof. [00228] The term "proteasome inhibitor" as used herein refers to compounds which target, decrease or inhibit the activity of the proteasome. Compounds which target, decrease or inhibit the activity of the proteasome include, but are not limited to, Bortezomib (VELCADE™) and MLN 341. [00229] The term "matrix metalloproteinase inhibitor" or ("MMP" inhibitor) as used herein includes, but is not limited to, collagen peptidomimetic and nonpeptidomimetic inhibitors, tetracycline derivatives, e.g., hydroxamate peptidomimetic inhibitor batimastat and its orally bioavailable analogue marimastat (BB-2516), prinomastat (AG3340), metastat (NSC 683551) BMS-279251, BAY 12-9566, TAA211, MMI270B or AAJ996. [00230] The term "compounds used in the treatment of hematologic malignancies" as used herein includes, but is not limited to, FMS-like tyrosine kinase inhibitors, which are compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon, 1-β-D-arabinofuransylcytosine (ara-c) and bisulfan; and ALK inhibitors, which are compounds which target, decrease or inhibit anaplastic lymphoma kinase. [00231] Compounds which target, decrease or inhibit the activity of FMS-like tyrosine kinase receptors (Flt-3R) are especially compounds, proteins or antibodies which inhibit members of the Flt-3R receptor kinase family, such as PKC412, midostaurin, a staurosporine derivative, SU11248 and MLN518. [00232] The term "HSP90 inhibitors" as used herein includes, but is not limited to, compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90; degrading, targeting, decreasing or inhibiting the HSP90 client proteins via the ubiquitin proteosome pathway. Compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90 are especially compounds, proteins or antibodies which inhibit the ATPase activity of HSP90, such as 17-allylamino,17-demethoxygeldanamycin (17AAG), a geldanamycin derivative; other geldanamycin related compounds; radicicol and HDAC inhibitors. [00233] The term "antiproliferative antibodies" as used herein includes, but is not limited to, trastuzumab (HERCEPTIN™), Trastuzumab-DM1, erbitux, bevacizumab (AVASTIN™), rituximab (RITUXAN®), PRO64553 (anti-CD40) and 2C4 Antibody. By antibodies is meant intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least 2 intact antibodies, and antibodies fragments so long as they exhibit the desired biological activity. [00234] For the treatment of acute myeloid leukemia (AML), compounds of the current disclosure can be used in combination with standard leukemia therapies, especially in combination with therapies used for the treatment of AML. In particular, compounds of the current disclosure can be administered in combination with, for example, farnesyl transferase inhibitors and/or other drugs useful for the treatment of AML, such as Daunorubicin, Adriamycin, Ara-C, VP-16, Teniposide, Mitoxantrone, Idarubicin, Carboplatinum and PKC412. [00235] Other anti-leukemic compounds include, for example, Ara-C, a pyrimidine analog, which is the 2'-alpha-hydroxy ribose (arabinoside) derivative of deoxycytidine. Also included is the purine analog of hypoxanthine, 6-mercaptopurine (6-MP) and fludarabine phosphate. Compounds which target, decrease or inhibit activity of histone deacetylase (HDAC) inhibitors such as sodium butyrate and suberoylanilide hydroxamic acid (SAHA) inhibit the activity of the enzymes known as histone deacetylases. Specific HDAC inhibitors include MS275, SAHA, FK228 (formerly FR901228), Trichostatin A and compounds disclosed in US 6,552,065 including, but not limited to, N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)-ethyl]- amino]methyl]phenyl]- 2E-2-propenamide, or a pharmaceutically acceptable salt thereof and N-hydroxy-3-[4-[(2- hydroxyethyl){2-(1H-indol-3-yl)ethyl]-amino]methyl]phenyl]-2E-2- propenamide, or a pharmaceutically acceptable salt thereof, especially the lactate salt. Somatostatin receptor antagonists as used herein refer to compounds which target, treat or inhibit the somatostatin receptor such as octreotide, and SOM230. Tumor cell damaging approaches refer to approaches such as ionizing radiation. The term "ionizing radiation" referred to above and hereinafter means ionizing radiation that occurs as either electromagnetic rays (such as X-rays and gamma rays) or particles (such as alpha and beta particles). Ionizing radiation is provided in, but not limited to, radiation therapy and is known in the art. See Hellman, Principles of Radiation Therapy, Cancer, in Principles and Practice of Oncology, Devita et al., Eds., 4th Edition, Vol.1, pp.248-275 (1993). [00236] Also included are EDG binders and ribonucleotide reductase inhibitors. The term “EDG binders” as used herein refers to a class of immunosuppressants that modulates lymphocyte recirculation, such as FTY720. The term “ribonucleotide reductase inhibitors” refers to pyrimidine or purine nucleoside analogs including, but not limited to, fludarabine and/or cytosine arabinoside (ara-C), 6-thioguanine, 5-fluorouracil, cladribine, 6-mercaptopurine (especially in combination with ara-C against ALL) and/or pentostatin. Ribonucleotide reductase inhibitors are especially hydroxyurea or 2-hydroxy-1H-isoindole-1 ,3-dione derivatives. [00237] Also included are in particular those compounds, proteins or monoclonal antibodies of VEGF such as 1-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine or a pharmaceutically acceptable salt thereof, 1-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine succinate; ANGIOSTATIN™; ENDOSTATIN™; anthranilic acid amides; ZD4190; Zd6474; SU5416; SU6668; bevacizumab; or anti-VEGF antibodies or anti-VEGF receptor antibodies, such as rhuMAb and RHUFab, VEGF aptamer such as Macugon; FLT-4 inhibitors, FLT-3 inhibitors, VEGFR-2 IgGI antibody, Angiozyme (RPI 4610) and Bevacizumab (AVASTIN™). [00238] Photodynamic therapy as used herein refers to therapy which uses certain chemicals known as photosensitizing compounds to treat or prevent cancers. Examples of photodynamic therapy include treatment with compounds, such as VISUDYNE™ and porfimer sodium. [00239] Angiostatic steroids as used herein refers to compounds which block or inhibit angiogenesis, such as, e.g., anecortave, triamcinolone, hydrocortisone, 11-α-epihydrocotisol, cortexolone, 17α-hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone and dexamethasone. [00240] Implants containing corticosteroids refers to compounds, such as fluocinolone and dexamethasone. [00241] Other chemotherapeutic compounds include, but are not limited to, plant alkaloids, hormonal compounds and antagonists; biological response modifiers, preferably lymphokines or interferons; antisense oligonucleotides or oligonucleotide derivatives; shRNA or siRNA; or miscellaneous compounds or compounds with other or unknown mechanism of action. [00242] The structure of the active compounds identified by code numbers, generic or trade names may be taken from the actual edition of the standard compendium "The Merck Index" or from databases, e.g., Patents International (e.g., IMS World Publications). Exemplary Immuno-Oncology agents [00243] In some embodiments, one or more other therapeutic agent is an immuno-oncology agent. As used herein, the term “an immuno-oncology agent” refers to an agent which is effective to enhance, stimulate, and/or up-regulate immune responses in a subject. In some embodiments, the administration of an immuno-oncology agent with a compound of the disclosure has a synergic effect in treating a cancer. [00244] An immuno-oncology agent can be, for example, a small molecule drug, an antibody, or a biologic or small molecule. Examples of biologic immuno-oncology agents include, but are not limited to, cancer vaccines, antibodies, and cytokines. In some embodiments, an antibody is a monoclonal antibody. In some embodiments, a monoclonal antibody is humanized or human. [00245] In some embodiments, an immuno-oncology agent is (i) an agonist of a stimulatory (including a co-stimulatory) receptor or (ii) an antagonist of an inhibitory (including a co- inhibitory) signal on T cells, both of which result in amplifying antigen-specific T cell responses. [00246] Certain of the stimulatory and inhibitory molecules are members of the immunoglobulin super family (IgSF). One important family of membrane-bound ligands that bind to co-stimulatory or co-inhibitory receptors is the B7 family, which includes B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6. Another family of membrane bound ligands that bind to co-stimulatory or co-inhibitory receptors is the TNF family of molecules that bind to cognate TNF receptor family members, which includes CD40 and CD40L, OX-40, OX-40L, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, TACI, APRIL, BCMA, LTβR, LIGHT, DcR3, HVEM, VEGI/TL1A, TRAMP/DR3, EDAR, EDA1, XEDAR, EDA2, TNFR1, Lymphotoxin α/TNFβ, TNFR2, TNFα, LTβR, Lymphotoxin α1β2, FAS, FASL, RELT, DR6, TROY, NGFR. [00247] In some embodiments, an immuno-oncology agent is a cytokine that inhibits T cell activation (e.g., IL-6, IL-10, TGF-β, VEGF, and other immunosuppressive cytokines) or a cytokine that stimulates T cell activation, for stimulating an immune response. [00248] In some embodiments, a combination of a compound of the disclosure and an immuno- oncology agent can stimulate T cell responses. In some embodiments, an immuno-oncology agent is: (i) an antagonist of a protein that inhibits T cell activation (e.g., immune checkpoint inhibitors) such as CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, Galectin 9, CEACAM-1, BTLA, CD69, Galectin-1, TIGIT, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, and TIM- 4; or (ii) an agonist of a protein that stimulates T cell activation such as B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD70, CD27, CD40, DR3 and CD28H. [00249] In some embodiments, an immuno-oncology agent is an antagonist of inhibitory receptors on NK cells or an agonist of activating receptors on NK cells. In some embodiments, an immuno-oncology agent is an antagonist of KIR, such as lirilumab. [00250] In some embodiments, an immuno-oncology agent is an agent that inhibits or depletes macrophages or monocytes, including but not limited to CSF-1R antagonists such as CSF-1R antagonist antibodies including RG7155 (WO11/70024, WO11/107553, WO11/131407, WO13/87699, WO13/119716, WO13/132044) or FPA-008 (WO11/140249; WO13169264; WO14/036357). [00251] In some embodiments, an immuno-oncology agent is selected from agonistic agents that ligate positive costimulatory receptors, blocking agents that attenuate signaling through inhibitory receptors, antagonists, and one or more agents that increase systemically the frequency of anti-tumor T cells, agents that overcome distinct immune suppressive pathways within the tumor microenvironment (e.g., block inhibitory receptor engagement (e.g., PD-L1/PD-1 interactions), deplete or inhibit Tregs (e.g., using an anti-CD25 monoclonal antibody (e.g., daclizumab) or by ex vivo anti-CD25 bead depletion), inhibit metabolic enzymes such as IDO, or reverse/prevent T cell energy or exhaustion) and agents that trigger innate immune activation and/or inflammation at tumor sites. [00252] In some embodiments, an immuno-oncology agent is a CTLA-4 antagonist. In some embodiments, a CTLA-4 antagonist is an antagonistic CTLA-4 antibody. In some embodiments, an antagonistic CTLA-4 antibody is YERVOY (ipilimumab) or tremelimumab. [00253] In some embodiments, an immuno-oncology agent is a PD-1 antagonist. In some embodiments, a PD-1 antagonist is administered by infusion. In some embodiments, an immuno- oncology agent is an antibody or an antigen-binding portion thereof that binds specifically to a Programmed Death-1 (PD-1) receptor and inhibits PD-1 activity. In some embodiments, a PD-1 antagonist is an antagonistic PD-1 antibody. In some embodiments, an antagonistic PD-1 antibody is OPDIVO (nivolumab), KEYTRUDA (pembrolizumab), or MEDI-0680 (AMP-514; WO2012/145493). In some embodiments, an immuno-oncology agent may be pidilizumab (CT- 011). In some embodiments, an immuno-oncology agent is a recombinant protein composed of the extracellular domain of PD-L2 (B7-DC) fused to the Fc portion of IgG1, called AMP-224. [00254] In some embodiments, an immuno-oncology agent is a PD-L1 antagonist. In some embodiments, a PD-L1 antagonist is an antagonistic PD-L1 antibody. In some embodiments, a PD-L1 antibody is MPDL3280A (RG7446; WO2010/077634), durvalumab (MEDI4736), BMS- 936559 (WO2007/005874), and MSB0010718C (WO2013/79174). [00255] In some embodiments, an immuno-oncology agent is a LAG-3 antagonist. In some embodiments, a LAG-3 antagonist is an antagonistic LAG-3 antibody. In some embodiments, a LAG3 antibody is BMS-986016 (WO10/19570, WO14/08218), or IMP-731 or IMP-321 (WO08/132601, WO009/44273). [00256] In some embodiments, an immuno-oncology agent is a CD137 (4-1BB) agonist. In some embodiments, a CD137 (4-1BB) agonist is an agonistic CD137 antibody. In some embodiments, a CD137 antibody is urelumab or PF-05082566 (WO12/32433). [00257] In some embodiments, an immuno-oncology agent is a GITR agonist. In some embodiments, a GITR agonist is an agonistic GITR antibody. In some embodiments, a GITR antibody is BMS-986153, BMS-986156, TRX-518 (WO006/105021, WO009/009116), or MK- 4166 (WO11/028683). [00258] In some embodiments, an immuno-oncology agent is an indoleamine (2,3)- dioxygenase (IDO) antagonist. In some embodiments, an IDO antagonist is selected from epacadostat (INCB024360, Incyte); indoximod (NLG-8189, NewLink Genetics Corporation); capmanitib (INC280, Novartis); GDC-0919 (Genentech/Roche); PF-06840003 (Pfizer); BMS:F001287 (Bristol-Myers Squibb); Phy906/KD108 (Phytoceutica); an enzyme that breaks down kynurenine (Kynase, Ikena Oncology, formerly known as Kyn Therapeutics); and NLG-919 (WO09/73620, WO009/1156652, WO11/56652, WO12/142237). [00259] In some embodiments, an immuno-oncology agent is an OX40 agonist. In some embodiments, an OX40 agonist is an agonistic OX40 antibody. In some embodiments, an OX40 antibody is MEDI-6383 or MEDI-6469. [00260] In some embodiments, an immuno-oncology agent is an OX40L antagonist. In some embodiments, an OX40L antagonist is an antagonistic OX40 antibody. In some embodiments, an OX40L antagonist is RG-7888 (WO06/029879). [00261] In some embodiments, an immuno-oncology agent is a CD40 agonist. In some embodiments, a CD40 agonist is an agonistic CD40 antibody. In some embodiments, an immuno- oncology agent is a CD40 antagonist. In some embodiments, a CD40 antagonist is an antagonistic CD40 antibody. In some embodiments, a CD40 antibody is lucatumumab or dacetuzumab. [00262] In some embodiments, an immuno-oncology agent is a CD27 agonist. In some embodiments, a CD27 agonist is an agonistic CD27 antibody. In some embodiments, a CD27 antibody is varlilumab. [00263] In some embodiments, an immuno-oncology agent is MGA271 (to B7H3) (WO11/109400). [00264] In some embodiments, an immuno-oncology agent is abagovomab, adecatumumab, afutuzumab, alemtuzumab, anatumomab mafenatox, apolizumab, atezolimab, avelumab, blinatumomab, BMS-936559, catumaxomab, durvalumab, epacadostat, epratuzumab, indoximod, inotuzumab ozogamicin, intelumumab, ipilimumab, isatuximab, lambrolizumab, MED14736, MPDL3280A, nivolumab, obinutuzumab, ocaratuzumab, ofatumumab, olatatumab, pembrolizumab, pidilizumab, rituximab, ticilimumab, samalizumab, or tremelimumab. [00265] In some embodiments, an immuno-oncology agent is an immunostimulatory agent. For example, antibodies blocking the PD-1 and PD-L1 inhibitory axis can unleash activated tumor- reactive T cells and have been shown in clinical trials to induce durable anti-tumor responses in increasing numbers of tumor histologies, including some tumor types that conventionally have not been considered immunotherapy sensitive. See, e.g., Okazaki, T. et al. (2013) Nat. Immunol.14, 1212–1218; Zou et al. (2016) Sci. Transl. Med.8. The anti-PD-1 antibody nivolumab (OPDIVO®, Bristol-Myers Squibb, also known as ONO-4538, MDX1106 and BMS-936558), has shown potential to improve the overall survival in patients with RCC who had experienced disease progression during or after prior anti-angiogenic therapy. [00266] In some embodiments, the immunomodulatory therapeutic specifically induces apoptosis of tumor cells. Approved immunomodulatory therapeutics which may be used in the present disclosure include pomalidomide (POMALYST®, Celgene); lenalidomide (REVLIMID®, Celgene); ingenol mebutate (PICATO®, LEO Pharma). [00267] In some embodiments, an immuno-oncology agent is a cancer vaccine. In some embodiments, the cancer vaccine is selected from sipuleucel-T (PROVENGE®, Dendreon/Valeant Pharmaceuticals), which has been approved for treatment of asymptomatic, or minimally symptomatic metastatic castrate-resistant (hormone-refractory) prostate cancer; and talimogene laherparepvec (IMLYGIC®, BioVex/Amgen, previously known as T-VEC), a genetically modified oncolytic viral therapy approved for treatment of unresectable cutaneous, subcutaneous and nodal lesions in melanoma. In some embodiments, an immuno-oncology agent is selected from an oncolytic viral therapy such as pexastimogene devacirepvec (PexaVec/JX-594, SillaJen/formerly Jennerex Biotherapeutics), a thymidine kinase- (TK-) deficient vaccinia virus engineered to express GM-CSF, for hepatocellular carcinoma (NCT02562755) and melanoma (NCT00429312); pelareorep (REOLYSIN®, Oncolytics Biotech), a variant of respiratory enteric orphan virus (reovirus) which does not replicate in cells that are not RAS-activated, in numerous cancers, including colorectal cancer (NCT01622543); prostate cancer (NCT01619813); head and neck squamous cell cancer (NCT01166542); pancreatic adenocarcinoma (NCT00998322); and non-small cell lung cancer (NSCLC) (NCT 00861627); enadenotucirev (NG-348, PsiOxus, formerly known as ColoAd1), an adenovirus engineered to express a full length CD80 and an antibody fragment specific for the T-cell receptor CD3 protein, in ovarian cancer (NCT02028117); metastatic or advanced epithelial tumors such as in colorectal cancer, bladder cancer, head and neck squamous cell carcinoma and salivary gland cancer (NCT02636036); ONCOS-102 (Targovax/formerly Oncos), an adenovirus engineered to express GM-CSF, in melanoma (NCT03003676); and peritoneal disease, colorectal cancer or ovarian cancer (NCT02963831); GL- ONC1 (GLV-1h68/GLV-1h153, Genelux GmbH), vaccinia viruses engineered to express beta- galactosidase (beta-gal)/beta-glucoronidase or beta-gal/human sodium iodide symporter (hNIS), respectively, were studied in peritoneal carcinomatosis (NCT01443260); fallopian tube cancer, ovarian cancer (NCT 02759588); or CG0070 (Cold Genesys), an adenovirus engineered to express GM-CSF, in bladder cancer (NCT02365818). [00268] In some embodiments, an immuno-oncology agent is selected from JX-929 (SillaJen/formerly Jennerex Biotherapeutics), a TK- and vaccinia growth factor-deficient vaccinia virus engineered to express cytosine deaminase, which is able to convert the prodrug 5- fluorocytosine to the cytotoxic drug 5-fluorouracil; TG01 and TG02 (Targovax/formerly Oncos), peptide-based immunotherapy agents targeted for difficult-to-treat RAS mutations; and TILT-123 (TILT Biotherapeutics), an engineered adenovirus designated: Ad5/3-E2F-delta24-hTNFα-IRES- hIL20; and VSV-GP (ViraTherapeutics) a vesicular stomatitis virus (VSV) engineered to express the glycoprotein (GP) of lymphocytic choriomeningitis virus (LCMV), which can be further engineered to express antigens designed to raise an antigen-specific CD8+ T cell response. [00269] In some embodiments, an immuno-oncology agent is a T-cell engineered to express a chimeric antigen receptor, or CAR. The T-cells engineered to express such chimeric antigen receptor are referred to as a CAR-T cells. [00270] CARs have been constructed that consist of binding domains, which may be derived from natural ligands, single chain variable fragments (scFv) derived from monoclonal antibodies specific for cell-surface antigens, fused to endodomains that are the functional end of the T-cell receptor (TCR), such as the CD3-zeta signaling domain from TCRs, which is capable of generating an activation signal in T lymphocytes. Upon antigen binding, such CARs link to endogenous signaling pathways in the effector cell and generate activating signals similar to those initiated by the TCR complex. [00271] For example, in some embodiments the CAR-T cell is one of those described in U.S. Patent 8,906,682 (June et al.; hereby incorporated by reference in its entirety), which discloses CAR-T cells engineered to comprise an extracellular domain having an antigen binding domain (such as a domain that binds to CD19), fused to an intracellular signaling domain of the T cell antigen receptor complex zeta chain (such as CD3 zeta). When expressed in the T cell, the CAR is able to redirect antigen recognition based on the antigen binding specificity. In the case of CD19, the antigen is expressed on malignant B cells. Over 200 clinical trials are currently in progress employing CAR-T in a wide range of indications. [https://clinicaltrials.gov/ct2/results?term=chimeric+antigen+receptors&pg=1]. [00272] In some embodiments, an immunostimulatory agent is an activator of retinoic acid receptor-related orphan receptor ^ (ROR ^t). ROR ^t is a transcription factor with key roles in the differentiation and maintenance of Type 17 effector subsets of CD4+ (Th17) and CD8+ (Tc17) T cells, as well as the differentiation of IL-17 expressing innate immune cell subpopulations such as NK cells. In some embodiments, an activator of ROR ^t is LYC-55716 (Lycera), which is currently being evaluated in clinical trials for the treatment of solid tumors (NCT02929862). [00273] In some embodiments, an immunostimulatory agent is an agonist or activator of a toll- like receptor (TLR). Suitable activators of TLRs include an agonist or activator of TLR9 such as SD-101 (Dynavax). SD-101 is an immunostimulatory CpG which is being studied for B-cell, follicular and other lymphomas (NCT02254772). Agonists or activators of TLR8 which may be used in the present disclosure include motolimod (VTX-2337, VentiRx Pharmaceuticals) which is being studied for squamous cell cancer of the head and neck (NCT02124850) and ovarian cancer (NCT02431559). [00274] Other immuno-oncology agents that can be used in the present disclosure include urelumab (BMS-663513, Bristol-Myers Squibb), an anti-CD137 monoclonal antibody; varlilumab (CDX-1127, Celldex Therapeutics), an anti-CD27 monoclonal antibody; BMS-986178 (Bristol- Myers Squibb), an anti-OX40 monoclonal antibody; lirilumab (IPH2102/BMS-986015, Innate Pharma, Bristol-Myers Squibb), an anti-KIR monoclonal antibody; monalizumab (IPH2201, Innate Pharma, AstraZeneca) an anti-NKG2A monoclonal antibody; andecaliximab (GS-5745, Gilead Sciences), an anti-MMP9 antibody; MK-4166 (Merck & Co.), an anti-GITR monoclonal antibody. [00275] In some embodiments, an immunostimulatory agent is selected from elotuzumab, mifamurtide, an agonist or activator of a toll-like receptor, and an activator of ROR ^t. [00276] In some embodiments, an immunostimulatory therapeutic is recombinant human interleukin 15 (rhIL-15). rhIL-15 has been tested in the clinic as a therapy for melanoma and renal cell carcinoma (NCT01021059 and NCT01369888) and leukemias (NCT02689453). In some embodiments, an immunostimulatory agent is recombinant human interleukin 12 (rhIL-12). In some embodiments, an IL-15 based immunotherapeutic is heterodimeric IL-15 (hetIL-15, Novartis/Admune), a fusion complex composed of a synthetic form of endogenous IL-15 complexed to the soluble IL-15 binding protein IL-15 receptor alpha chain (IL15:sIL-15RA), which has been tested in Phase 1 clinical trials for melanoma, renal cell carcinoma, non-small cell lung cancer and head and neck squamous cell carcinoma (NCT02452268). In some embodiments, a recombinant human interleukin 12 (rhIL-12) is NM-IL-12 (Neumedicines, Inc.), NCT02544724, or NCT02542124. [00277] In some embodiments, an immuno-oncology agent is selected from those descripted in Jerry L. Adams et al., “Big opportunities for small molecules in immuno-oncology,” Cancer Therapy 2015, Vol.14, pages 603-622, the content of which is incorporated herein by reference in its entirety. In some embodiments, an immuno-oncology agent is selected from the examples described in Table 1 of Jerry L. Adams et al. In some embodiments, an immuno-oncology agent is a small molecule targeting an immuno-oncology target selected from those listed in Table 2 of Jerry L. Adams et al. In some embodiments, an immuno-oncology agent is a small molecule agent selected from those listed in Table 2 of Jerry L. Adams et al. [00278] In some embodiments, an immuno-oncology agent is selected from the small molecule immuno-oncology agents described in Peter L. Toogood, “Small molecule immuno-oncology therapeutic agents,” Bioorganic & Medicinal Chemistry Letters 2018, Vol.28, pages 319-329, the content of which is incorporated herein by reference in its entirety. In some embodiments, an immuno-oncology agent is an agent targeting the pathways as described in Peter L. Toogood. [00279] In some embodiments, an immuno-oncology agent is selected from those described in Sandra L. Ross et al., “Bispecific T cell engager (BITE®) antibody constructs can mediate bystander tumor cell killing”, PLoS ONE 12(8): e0183390, the content of which is incorporated herein by reference in its entirety. In some embodiments, an immuno-oncology agent is a bispecific T cell engager (BITE®) antibody construct. In some embodiments, a bispecific T cell engager (BITE®) antibody construct is a CD19/CD3 bispecific antibody construct. In some embodiments, a bispecific T cell engager (BITE®) antibody construct is an EGFR/CD3 bispecific antibody construct. In some embodiments, a bispecific T cell engager (BITE®) antibody construct activates T cells. In some embodiments, a bispecific T cell engager (BITE®) antibody construct activates T cells, which release cytokines inducing upregulation of intercellular adhesion molecule 1 (ICAM-1) and FAS on bystander cells. In some embodiments, a bispecific T cell engager (BITE®) antibody construct activates T cells which result in induced bystander cell lysis. In some embodiments, the bystander cells are in solid tumors. In some embodiments, the bystander cells being lysed are in proximity to the BITE®-activated T cells. In some embodiment, the bystander cells comprises tumor-associated antigen (TAA) negative cancer cells. In some embodiment, the bystander cells comprise EGFR-negative cancer cells. In some embodiments, an immuno- oncology agent is an antibody which blocks the PD-L1/PD1 axis and/or CTLA4. In some embodiments, an immuno-oncology agent is an ex vivo expanded tumor-infiltrating T cell. In some embodiments, an immuno-oncology agent is a bispecific antibody construct or chimeric antigen receptors (CARs) that directly connect T cells with tumor-associated surface antigens (TAAs). Exemplary Immune Checkpoint Inhibitors [00280] In some embodiments, an immuno-oncology agent is an immune checkpoint inhibitor as described herein. [00281] The term “checkpoint inhibitor” as used herein relates to agents useful in preventing cancer cells from avoiding the immune system of the patient. One of the major mechanisms of anti-tumor immunity subversion is known as “T-cell exhaustion,” which results from chronic exposure to antigens that has led to up-regulation of inhibitory receptors. These inhibitory receptors serve as immune checkpoints in order to prevent uncontrolled immune reactions. [00282] PD-1 and co-inhibitory receptors such as cytotoxic T-lymphocyte antigen 4 (CTLA-4, B and T Lymphocyte Attenuator (BTLA; CD272), T cell Immunoglobulin and Mucin domain-3 (Tim-3), Lymphocyte Activation Gene-3 (Lag-3; CD223), and others are often referred to as a checkpoint regulators. They act as molecular “gatekeepers” that allow extracellular information to dictate whether cell cycle progression and other intracellular signaling processes should proceed. [00283] In some embodiments, an immune checkpoint inhibitor is an antibody to PD-1. PD-1 binds to the programmed cell death 1 receptor (PD-1) to prevent the receptor from binding to the inhibitory ligand PDL-1, thus overriding the ability of tumors to suppress the host anti-tumor immune response. [00284] In one aspect, the checkpoint inhibitor is a biologic therapeutic or a small molecule. In another aspect, the checkpoint inhibitor is a monoclonal antibody, a humanized antibody, a fully human antibody, a fusion protein or a combination thereof. In a further aspect, the checkpoint inhibitor inhibits a checkpoint protein selected from CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands or a combination thereof. In an additional aspect, the checkpoint inhibitor interacts with a ligand of a checkpoint protein selected from CTLA-4, PDL1, PDL2, PDl, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands or a combination thereof. In an aspect, the checkpoint inhibitor is an immunostimulatory agent, a T cell growth factor, an interleukin, an antibody, a vaccine or a combination thereof. In a further aspect, the interleukin is IL-7 or IL-15. In a specific aspect, the interleukin is glycosylated IL-7. In an additional aspect, the vaccine is a dendritic cell (DC) vaccine. [00285] Checkpoint inhibitors include any agent that blocks or inhibits in a statistically significant manner, the inhibitory pathways of the immune system. Such inhibitors may include small molecule inhibitors or may include antibodies, or antigen binding fragments thereof, that bind to and block or inhibit immune checkpoint receptors or antibodies that bind to and block or inhibit immune checkpoint receptor ligands. Illustrative checkpoint molecules that can be targeted for blocking or inhibition include, but are not limited to, CTLA-4, PDL1, PDL2, PD1, B7-H3, B7- H4, BTLA, HVEM, GAL9, LAG3, TIM3, VISTA, KIR, 2B4 (belongs to the CD2 family of molecules and is expressed on all NK, γδ, and memory CD8+ (αβ) T cells), CD160 (also referred to as BY55), CGEN-15049, CHK 1 and CHK2 kinases, A2aR, and various B-7 family ligands. B7 family ligands include, but are not limited to, B7- 1, B7-2, B7-DC, B7-H1, B7-H2, B7-H3, B7- H4, B7-H5, B7-H6 and B7-H7. Checkpoint inhibitors include antibodies, or antigen binding fragments thereof, other binding proteins, biologic therapeutics, or small molecules, that bind to and block or inhibit the activity of one or more of CTLA-4, PDL1, PDL2, PD1, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD 160 and CGEN-15049. Illustrative immune checkpoint inhibitors include, but are not limited to, Tremelimumab (CTLA-4 blocking antibody), anti-OX40, PD-L1 monoclonal Antibody (Anti-B7-Hl; MEDI4736), MK-3475 (PD-1 blocker), Nivolumab (anti-PD1 antibody), CT-011 (anti-PD1 antibody), BY55 monoclonal antibody, AMP224 (anti-PDL1 antibody), BMS- 936559 (anti-PDL1 antibody), MPLDL3280A (anti-PDL1 antibody), MSB0010718C (anti-PDL1 antibody), and ipilimumab (anti-CTLA-4 checkpoint inhibitor). Checkpoint protein ligands include, but are not limited to PD-L1, PD-L2, B7-H3, B7- H4, CD28, CD86 and TIM-3. [00286] In certain embodiments, the immune checkpoint inhibitor is selected from a PD-1 antagonist, a PD-L1 antagonist, and a CTLA-4 antagonist. In some embodiments, the checkpoint inhibitor is selected from the group consisting of nivolumab (OPDIVO®), ipilimumab (YERVOY®), and pembrolizumab (KEYTRUDA®). In some embodiments, the checkpoint inhibitor is selected from nivolumab (anti-PD-1 antibody, OPDIVO®, Bristol-Myers Squibb); pembrolizumab (anti-PD-1 antibody, KEYTRUDA®, Merck); ipilimumab (anti-CTLA-4 antibody, YERVOY®, Bristol-Myers Squibb); durvalumab (anti-PD-L1 antibody, IMFINZI®, AstraZeneca); and atezolizumab (anti-PD-L1 antibody, TECENTRIQ®, Genentech). [00287] In some embodiments, the checkpoint inhibitor is selected from the group consisting of lambrolizumab (MK-3475), nivolumab (BMS-936558), pidilizumab (CT-011), AMP-224, MDX-1105, MEDI4736, MPDL3280A, BMS-936559, ipilimumab, lirlumab, IPH2101, pembrolizumab (KEYTRUDA®), and tremelimumab. [00288] In some embodiments, an immune checkpoint inhibitor is REGN2810 (Regeneron), an anti-PD-1 antibody tested in patients with basal cell carcinoma (NCT03132636); NSCLC (NCT03088540); cutaneous squamous cell carcinoma (NCT02760498); lymphoma (NCT02651662); and melanoma (NCT03002376); pidilizumab (CureTech), also known as CT- 011, an antibody that binds to PD-1, in clinical trials for diffuse large B-cell lymphoma and multiple myeloma; avelumab (BAVENCIO®, Pfizer/Merck KGaA), also known as MSB0010718C), a fully human IgG1 anti-PD-L1 antibody, in clinical trials for non-small cell lung cancer, Merkel cell carcinoma, mesothelioma, solid tumors, renal cancer, ovarian cancer, bladder cancer, head and neck cancer, and gastric cancer; or PDR001 (Novartis), an inhibitory antibody that binds to PD-1, in clinical trials for non-small cell lung cancer, melanoma, triple negative breast cancer and advanced or metastatic solid tumors. Tremelimumab (CP-675,206; Astrazeneca) is a fully human monoclonal antibody against CTLA-4 that has been in studied in clinical trials for a number of indications, including: mesothelioma, colorectal cancer, kidney cancer, breast cancer, lung cancer and non-small cell lung cancer, pancreatic ductal adenocarcinoma, pancreatic cancer, germ cell cancer, squamous cell cancer of the head and neck, hepatocellular carcinoma, prostate cancer, endometrial cancer, metastatic cancer in the liver, liver cancer, large B-cell lymphoma, ovarian cancer, cervical cancer, metastatic anaplastic thyroid cancer, urothelial cancer, fallopian tube cancer, multiple myeloma, bladder cancer, soft tissue sarcoma, and melanoma. AGEN-1884 (Agenus) is an anti-CTLA4 antibody that is being studied in Phase 1 clinical trials for advanced solid tumors (NCT02694822). [00289] In some embodiments, a checkpoint inhibitor is an inhibitor of T-cell immunoglobulin mucin containing protein-3 (TIM-3). TIM-3 inhibitors that may be used in the present disclosure include TSR-022, LY3321367 and MBG453. TSR-022 (Tesaro) is an anti-TIM-3 antibody which is being studied in solid tumors (NCT02817633). LY3321367 (Eli Lilly) is an anti-TIM-3 antibody which is being studied in solid tumors (NCT03099109). MBG453 (Novartis) is an anti- TIM-3 antibody which is being studied in advanced malignancies (NCT02608268). [00290] In some embodiments, a checkpoint inhibitor is an inhibitor of T cell immunoreceptor with Ig and ITIM domains, or TIGIT, an immune receptor on certain T cells and NK cells. TIGIT inhibitors that may be used in the present disclosure include BMS-986207 (Bristol-Myers Squibb), an anti-TIGIT monoclonal antibody (NCT02913313); OMP-313M32 (Oncomed); and anti-TIGIT monoclonal antibody (NCT03119428). [00291] In some embodiments, a checkpoint inhibitor is an inhibitor of Lymphocyte Activation Gene-3 (LAG-3). LAG-3 inhibitors that may be used in the present disclosure include BMS- 986016 and REGN3767 and IMP321. BMS-986016 (Bristol-Myers Squibb), an anti-LAG-3 antibody, is being studied in glioblastoma and gliosarcoma (NCT02658981). REGN3767 (Regeneron), is also an anti-LAG-3 antibody, and is being studied in malignancies (NCT03005782). IMP321 (Immutep S.A.) is an LAG-3-Ig fusion protein, being studied in melanoma (NCT02676869); adenocarcinoma (NCT02614833); and metastatic breast cancer (NCT00349934). [00292] Checkpoint inhibitors that may be used in the present disclosure include OX40 agonists. OX40 agonists that are being studied in clinical trials include PF-04518600/PF-8600 (Pfizer), an agonistic anti-OX40 antibody, in metastatic kidney cancer (NCT03092856) and advanced cancers and neoplasms (NCT02554812; NCT05082566); GSK3174998 (Merck), an agonistic anti-OX40 antibody, in Phase 1 cancer trials (NCT02528357); MEDI0562 (Medimmune/AstraZeneca), an agonistic anti-OX40 antibody, in advanced solid tumors (NCT02318394 and NCT02705482); MEDI6469, an agonistic anti-OX40 antibody (Medimmune/AstraZeneca), in patients with colorectal cancer (NCT02559024), breast cancer (NCT01862900), head and neck cancer (NCT02274155) and metastatic prostate cancer (NCT01303705); and BMS-986178 (Bristol-Myers Squibb) an agonistic anti-OX40 antibody, in advanced cancers (NCT02737475). [00293] Checkpoint inhibitors that may be used in the present disclosure include CD137 (also called 4-1BB) agonists. CD137 agonists that are being studied in clinical trials include utomilumab (PF-05082566, Pfizer) an agonistic anti-CD137 antibody, in diffuse large B-cell lymphoma (NCT02951156) and in advanced cancers and neoplasms (NCT02554812 and NCT05082566); urelumab (BMS-663513, Bristol-Myers Squibb), an agonistic anti-CD137 antibody, in melanoma and skin cancer (NCT02652455) and glioblastoma and gliosarcoma (NCT02658981); and CTX-471 (Compass Therapeutics), an agonistic anti-CD137 antibody in metastatic or locally advanced malignancies (NCT03881488). [00294] Checkpoint inhibitors that may be used in the present disclosure include CD27 agonists. CD27 agonists that are being studied in clinical trials include varlilumab (CDX-1127, Celldex Therapeutics) an agonistic anti-CD27 antibody, in squamous cell head and neck cancer, ovarian carcinoma, colorectal cancer, renal cell cancer, and glioblastoma (NCT02335918); lymphomas (NCT01460134); and glioma and astrocytoma (NCT02924038). [00295] Checkpoint inhibitors that may be used in the present disclosure include glucocorticoid- induced tumor necrosis factor receptor (GITR) agonists. GITR agonists that are being studied in clinical trials include TRX518 (Leap Therapeutics), an agonistic anti-GITR antibody, in malignant melanoma and other malignant solid tumors (NCT01239134 and NCT02628574); GWN323 (Novartis), an agonistic anti-GITR antibody, in solid tumors and lymphoma (NCT 02740270); INCAGN01876 (Incyte/Agenus), an agonistic anti-GITR antibody, in advanced cancers (NCT02697591 and NCT03126110); MK-4166 (Merck), an agonistic anti-GITR antibody, in solid tumors (NCT02132754) and MEDI1873 (Medimmune/AstraZeneca), an agonistic hexameric GITR-ligand molecule with a human IgG1 Fc domain, in advanced solid tumors (NCT02583165). [00296] Checkpoint inhibitors that may be used in the present disclosure include inducible T- cell co-stimulator (ICOS, also known as CD278) agonists. ICOS agonists that are being studied in clinical trials include MEDI-570 (Medimmune), an agonistic anti-ICOS antibody, in lymphomas (NCT02520791); GSK3359609 (Merck), an agonistic anti-ICOS antibody, in Phase 1 (NCT02723955); JTX-2011 (Jounce Therapeutics), an agonistic anti-ICOS antibody, in Phase 1 (NCT02904226). [00297] Checkpoint inhibitors that may be used in the present disclosure include killer IgG-like receptor (KIR) inhibitors. KIR inhibitors that are being studied in clinical trials include lirilumab (IPH2102/BMS-986015, Innate Pharma/Bristol-Myers Squibb), an anti-KIR antibody, in leukemias (NCT01687387, NCT02399917, NCT02481297, NCT02599649), multiple myeloma (NCT02252263), and lymphoma (NCT01592370); IPH2101 (1-7F9, Innate Pharma) in myeloma (NCT01222286 and NCT01217203); and IPH4102 (Innate Pharma), an anti-KIR antibody that binds to three domains of the long cytoplasmic tail (KIR3DL2), in lymphoma (NCT02593045). [00298] Checkpoint inhibitors that may be used in the present disclosure include CD47 inhibitors of interaction between CD47 and signal regulatory protein alpha (SIRPa). CD47/SIRPa inhibitors that are being studied in clinical trials include ALX-148 (Alexo Therapeutics), an antagonistic variant of (SIRPa) that binds to CD47 and prevents CD47/SIRPa-mediated signaling, in phase 1 (NCT03013218); TTI-621 (SIRPa-Fc, Trillium Therapeutics), a soluble recombinant fusion protein created by linking the N-terminal CD47-binding domain of SIRPa with the Fc domain of human IgG1, acts by binding human CD47, and preventing it from delivering its “do not eat” signal to macrophages, is in clinical trials in Phase 1 (NCT02890368 and NCT02663518); CC-90002 (Celgene), an anti-CD47 antibody, in leukemias (NCT02641002); and Hu5F9-G4 (Forty Seven, Inc.), in colorectal neoplasms and solid tumors (NCT02953782), acute myeloid leukemia (NCT02678338) and lymphoma (NCT02953509). [00299] Checkpoint inhibitors that may be used in the present disclosure include CD73 inhibitors. CD73 inhibitors that are being studied in clinical trials include MEDI9447 (Medimmune), an anti-CD73 antibody, in solid tumors (NCT02503774); and BMS-986179 (Bristol-Myers Squibb), an anti-CD73 antibody, in solid tumors (NCT02754141). [00300] Checkpoint inhibitors that may be used in the present disclosure include agonists of stimulator of interferon genes protein (STING, also known as transmembrane protein 173, or TMEM173). Agonists of STING that are being studied in clinical trials include MK-1454 (Merck), an agonistic synthetic cyclic dinucleotide, in lymphoma (NCT03010176); and ADU- S100 (MIW815, Aduro Biotech/Novartis), an agonistic synthetic cyclic dinucleotide, in Phase 1 (NCT02675439 and NCT03172936). [00301] Checkpoint inhibitors that may be used in the present disclosure include CSF1R inhibitors. CSF1R inhibitors that are being studied in clinical trials include pexidartinib (PLX3397, Plexxikon), a CSF1R small molecule inhibitor, in colorectal cancer, pancreatic cancer, metastatic and advanced cancers (NCT02777710) and melanoma, non-small cell lung cancer, squamous cell head and neck cancer, gastrointestinal stromal tumor (GIST) and ovarian cancer (NCT02452424); and IMC-CS4 (LY3022855, Lilly), an anti-CSF-1R antibody, in pancreatic cancer (NCT03153410), melanoma (NCT03101254), and solid tumors (NCT02718911); and BLZ945 (4-[2((1R,2R)-2-hydroxycyclohexylamino)-benzothiazol-6-yloxyl]-pyridine-2- carboxylic acid methylamide, Novartis), an orally available inhibitor of CSF1R, in advanced solid tumors (NCT02829723). [00302] Checkpoint inhibitors that can be used in the present disclosure include NKG2A receptor inhibitors. NKG2A receptor inhibitors that are being studied in clinical trials include monalizumab (IPH2201, Innate Pharma), an anti-NKG2A antibody, in head and neck neoplasms (NCT02643550) and chronic lymphocytic leukemia (NCT02557516). [00303] In some embodiments, the immune checkpoint inhibitor is selected from nivolumab, pembrolizumab, ipilimumab, avelumab, durvalumab, atezolizumab, or pidilizumab. EXAMPLES [00304] The following examples are included to demonstrate various aspects of the present disclosure. It should be appreciated by those of skill in the art that the techniques disclosed in the examples that follow represent techniques discovered by the inventors to function well in the practice of the disclosure, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific examples which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the disclosure. EXAMPLE 1: Synthesis of Certain Exemplary Compounds Synthesis of 3-[[2-Fluoro-3-(methylsulfamoylamino)phenyl]methyl]-7-pyrimidin-2-yloxy- 4H-1,3-benzoxazin-2-one (I-3)
Figure imgf000102_0001
Step 1: 3-Amino-2-fluorobenzonitrile
Figure imgf000102_0002
[00305] To a stirred solution of 2-fluoro-3-nitrobenzonitrile (2 g, 12.04 mmol, 1 eq) in EtOH (20 mL) and H2O (4 mL) was added NH4Cl (6.44 g, 120.40 mmol, 10 eq) and Fe (3.36 g, 60.20 mmol, 5 eq). The reaction mixture was stirred at 90 °C for 12 h under N2. The reaction mixture was filtered through celite and the filtrate was concentrated to yield a residue which was diluted with H2O (100 mL) and extracted with EtOAc (100 mL × 3). The combined organic layers were washed with brine (30 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to yield 3-amino-2-fluorobenzonitrile (1.4 g, crude) as a black brown solid.1H NMR (400 MHz, DMSO-d6) δ ppm 7.11-7.02 (m, 2H), 6.95-6.89 (m, 1H), 5.75-5.63 (m, 2H); ES-LCMS m/z 137.3 [M+H]+. Step 2: tert-Butyl N-tert-butoxycarbonyl-N-(3-cyano-2-fluoro-phenyl)carbamate
Figure imgf000103_0001
[00306] To a stirred solution of 3-amino-2-fluorobenzonitrile (1.4 g, 10.28 mmol, 1 eq) in THF (40 mL) was added Boc2O (6.73 g, 30.85 mmol, 7.09 mL, 3 eq), DMAP (251.29 mg, 2.06 mmol, 0.2 eq) and TEA (2.08 g, 20.57 mmol, 2.86 mL, 2 eq).The reaction mixture was stirred at 80 °C for 12 h under N2. The reaction mixture was concentrated under reduced pressure to yield a residue which was purified by flash silica gel chromatography (from PE/EtOAc = 30/1 to 10/1, TLC: PE/EtOAc = 3/1, Rf = 0.60) to yield tert-butyl N-tert-butoxycarbonyl-N-(3-cyano-2-fluoro- phenyl)carbamate (1.5 g, 3.48 mmol, 33.8% yield, 78.0% purity) as a white solid.1H NMR (400 MHz, DMSO-d6) δ ppm 7.99-7.93 (m, 1H), 7.89-7.81 (m, 1H), 7.49-7.43 (m, 1H), 1.38 (s, 18H); ES-LCMS m/z 695.1 [2M+Na]+. Step 3: tert-Butyl N-[3-(aminomethyl)-2-fluoro-phenyl]-N-tert-butoxycarbonyl-carbamate
Figure imgf000103_0002
[00307] To a solution of tert-butyl N-tert-butoxycarbonyl-N-(3-cyano-2-fluoro- phenyl)carbamate (1.5 g, 3.48 mmol, 78.0% purity, 1 eq) in MeOH (60 mL) was added Raney-Ni (298.02 mg, 3.48 mmol, 1 eq) and NH3.H2O (1.31 g, 10.44 mmol, 1.44 mL, 28%, 3 eq) under N2. The suspension was degassed under vacuum and purged with H2 several times. The mixture was stirred under H2 (50 psi) at 20 °C for 12 h. The reaction mixture was filtered through a pad of celite and the filtrate was concentrated to yield tert-butyl N-[3-(aminomethyl)-2-fluoro-phenyl]-N-tert- butoxycarbonyl-carbamate (1 g, crude) as brown oil.1H NMR (400 MHz, DMSO-d6) δ ppm 7.51- 7.42 (m, 1H), 7.22-7.11 (m, 2H), 3.80-3.71 (m, 2H), 1.42-1.36 (m, 18H); ES-LCMS m/z 681.3 [2M+H]+. Step 4: 2-Hydroxy-4-pyrimidin-2-yloxy-benzaldehyde
Figure imgf000103_0003
[00308] To a stirred solution of 2,4-dihydroxybenzaldehyde (5 g, 36.20 mmol, 1 eq) in DMF (80 mL) was added Cs2CO3 (35.38 g, 108.60 mmol, 3 eq) and 2-bromopyrimidine (5.76 g, 36.20 mmol, 1 eq). The reaction mixture was stirred at 80 °C for 12 h under N2. The reaction mixture was diluted with H2O (300 mL) and extracted with EtOAc (500mL × 3). The combined organic layers were washed with brine (300 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to yield a residue which was purified by flash silica gel chromatography (from PE/EtOAc = 100/1 to 1/1, TLC: PE/EtOAc = 0/1, Rf = 0.70) to yield 2-hydroxy-4-pyrimidin-2- yloxy-benzaldehyde (1.4 g, 6.02 mmol, 16.6% yield, 93.0% purity) as a yellow solid.1H NMR (400 MHz, CDCl3) δ ppm 6.85-6.90 (m, 2H), 7.14 (t, J = 4.75 Hz, 1H), 7.63 (d, J = 8.25 Hz, 1H), 8.62 (d, J = 4.75 Hz, 2H), 9.88 (s, 1H), 11.30 (s, 1H); ES-LCMS m/z 217.0 [M+H]+. Step 5: tert-Butyl N-tert-butoxycarbonyl-N-[2-fluoro-3-[[(2-hydroxy-4-pyrimidin-2-yloxy- phenyl)methylamino]methyl]phenyl]carbamate
Figure imgf000104_0001
[00309] To a solution of 2-hydroxy-4-pyrimidin-2-yloxy-benzaldehyde (400 mg, 1.72 mmol, 93.0% purity, 1 eq) and tert-butyl N-[3-(aminomethyl)-2-fluoro-phenyl]-N-tert-butoxycarbonyl- carbamate (585.70 mg, 1.72 mmol, 1 eq) in MeOH (10 mL) was added AcOH (103.33 mg, 1.72 mmol, 98.41 μL, 1 eq) and NaBH3CN (324.39 mg, 5.16 mmol, 3 eq) at 0 °C. The mixture was stirred at 50 °C for 12 h. The reaction mixture was concentrated under reduced pressure to yield a residue which was purified by flash silica gel chromatography (from DCM/MeOH = 100/1 to 30/1, TLC: DCM/MeOH = 10/1, Rf = 0.40) to yield tert-butyl N-tert-butoxycarbonyl-N-[2-fluoro-3- [[(2-hydroxy-4-pyrimidin-2-yloxy-phenyl)methylamino]methyl]phenyl]carbamate (770 mg, 1.28 mmol, 74.5% yield, 90.0% purity) as a red solid.1H NMR (400 MHz, DMSO-d6) δ ppm 8.63 (d, J = 4.8 Hz, 2H), 7.50-7.42 (m, 1H), 7.28-7.20 (m, 3H), 7.14 (d, J = 8.0 Hz, 1H), 6.57-6.52 (m, 2H), 3.80 (br d, J = 16.9 Hz, 4H), 1.37 (s, 18H); ES-LCMS m/z 541.2 [M+H]+. Step 6: tert-Butyl N-tert-butoxycarbonyl-N-[2-fluoro-3-[(2-oxo-7-pyrimidin-2-yloxy-4H-1,3- benzoxazin-3-yl)methyl]phenyl]carbamate
Figure imgf000104_0002
[00310] To a stirred solution of tert-butyl N-tert-butoxycarbonyl-N-[2-fluoro-3-[[(2-hydroxy- 4-pyrimidin-2-yloxy-phenyl)methylamino]methyl]phenyl]carbamate (80 mg, 136.15 µmol, 92% purity, 1 eq) in THF (5 mL) was added CDI (66.23 mg, 408.45 µmol, 3 eq).The reaction mixture was stirred at 45 °C for 12 h under N2. The reaction mixture was concentrated under reduced pressure to yield a residue which was purified by preparative TLC (DCM/MeOH = 10/1, TLC: DCM/MeOH = 10/1, Rf = 0.55) to yield tert-butyl N-tert-butoxycarbonyl-N-[2-fluoro-3-[(2-oxo- 7-pyrimidin-2-yloxy-4H-1,3-benzoxazin-3-yl)methyl]phenyl]carbamate (60 mg, 100.6 μmol, 73.9% yield, 95.0% purity) as a white solid.1H NMR (400 MHz, CD3OD) δ ppm 8.59 (d, J = 4.9 Hz, 2H), 7.56-7.43 (m, 1H), 7.29-7.20 (m, 4H), 6.98 (dd, J = 2.3, 8.4 Hz, 1H), 6.92 (d, J = 2.3 Hz, 1H), 4.80 (s, 2H), 4.56 (s, 2H), 1.35 (s, 18H); ES-LCMS m/z 411.1 [M-Boc-t-Butyl+H]+. Step 7: 3-[(3-Amino-2-fluoro-phenyl)methyl]-7-pyrimidin-2-yloxy-4H-1,3-benzoxazin-2-one
Figure imgf000105_0001
[00311] To a stirred solution of tert-butyl N-tert-butoxycarbonyl-N-[2-fluoro-3-[(2-oxo-7- pyrimidin-2-yloxy-4H-1,3-benzoxazin-3-yl)methyl]phenyl]carbamate (50.00 mg, 83.84 μmol, 95.0% purity, 1 eq) in DCM (2.5 mL) was added TFA (0.5 mL).The reaction mixture was stirred at 25 °C for 1 h under N2. The reaction mixture was concentrated under reduced pressure to yield 3-[(3-amino-2-fluoro-phenyl)methyl]-7-pyrimidin-2-yloxy-4H-1,3-benzoxazin-2-one (35 mg, crude, TFA) as a white solid.1H NMR (400 MHz, CD3OD) δ ppm 8.60 (d, J = 4.9 Hz, 2H), 7.23 (s, 2H), 7.18-7.18 (m, 1H), 7.21-7.13 (m, 2H), 6.98 (dd, J = 2.3, 8.3 Hz, 1H), 6.93-6.93 (m, 1H), 6.92 (d, J = 2.3 Hz, 1H), 4.80 (s, 2H), 4.55 (s, 2H); ES-LCMS m/z 367.0 [M+H]+. Step 8: 3-[[2-Fluoro-3-(methylsulfamoylamino)phenyl]methyl]-7-pyrimidin-2-yloxy-4H-1,3- benzoxazin-2-one
Figure imgf000105_0002
[00312] To a stirred solution of 3-[(3-amino-2-fluoro-phenyl)methyl]-7-pyrimidin-2-yloxy-4H- 1,3-benzoxazin-2-one (35 mg, 72.86 μmol, 1 eq, TFA) in DCM (3 mL) was added DIEA (28.25 mg, 218.58 μmol, 38.07 μL, 3 eq) and METHYLSULFAMOYL CHLORIDE (8.50 mg, 65.57 μmol, 0.9 eq) under N2 atmosphere at 0 °C. The reaction mixture was stirred at 25 °C for 12 h under N2. The reaction mixture was concentrated under reduced pressure to yield a residue which was purified by preparative HPLC (column: Phenomenex Luna C18150*25mm*10 μm; mobile phase: [water( NH4HCO3)-ACN]; B%: 22%-52%, 8min) followed by lyophilization to yield 3-[[2- fluoro-3-(methylsulfamoylamino)phenyl]methyl]-7-pyrimidin-2-yloxy-4H-1,3-benzoxazin-2-one (5.6 mg, 11.85 μmol, 16.3% yield, 97.2% purity) as a white solid.1H NMR (400 MHz, CD3OD) δ ppm 8.62 (d, J = 4.9 Hz, 2H), 7.54 (dt, J = 2.0, 7.6 Hz, 1H), 7.28-7.24 (m, 2H), 7.22-7.14 (m, 2H), 7.00 (dd, J = 2.3, 8.3 Hz, 1H), 6.94 (d, J = 2.3 Hz, 1H), 4.81 (s, 2H), 4.55 (s, 2H), 2.64 (s, 3H); ES-LCMS m/z 460.1 [M+H]+. Synthesis of I-10
Figure imgf000106_0001
Step 1: 1-(2-chloro-3-fluoropyridin-4-yl)-N-(2,4-dimethoxybenzyl)methanamine
Figure imgf000106_0002
[00313] A solution of 1-(2,4-dimethoxyphenyl)methanamine (20.0 g, 119 mmol, 1.0 eq) in MeOH (300 mL) was treated with 2-chloro-3-fluoropyridine-4-carbaldehyde (22.9 g, 143 mmol, 1.2 eq) for 14 h at room temperature under nitrogen atmosphere followed by the addition of NaBH4 (9.0 g, 239 mmol, 2.0 eq) in portions at 0°C. The resulting mixture was stirred for 1 h at room temperature, then quenched with water/Ice at 0°C. The resulting mixture was extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with brine (2 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. to afford [(2-chloro-3-fluoropyridin-4-yl)methyl][(2,4-dimethoxyphenyl)methyl]amine (31.0 g, 83%) as an off-white solid. The crude product was used in the next step directly without further purification. [00314] ES-LCMS m/z 311 [M+H]+. Step 2: 4-((((2-chloro-3-fluoropyridin-4-yl)methyl)amino)methyl)benzene-1,3-diol
Figure imgf000107_0001
[00315] Into a 500 mL 3-necked round-bottom flask were added [(2-chloro-3-fluoropyridin-4- yl)methyl][(2,4-dimethoxyphenyl)methyl]amine (31.0 g, 99 mmol, 1.0 eq) and DCE (400 mL) at room temperature. To the above mixture was added BBr3 (249.9 g, 997 mmol, 10.0 eq) dropwise over 30 min at 0°C. The resulting mixture was stirred for additional 30 h at room temperature then was quenched with water/ice at 0°C. The mixture residue was basified to pH 8 with saturated Na2CO3 (aq.). The resulting mixture was extracted with CH2Cl2 (3 x 200 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1:1) to afford 4-({[(2-chloro-3-fluoropyridin-4- yl)methyl]amino}methyl)benzene-1,3-diol (18.0 g, 63%) as a yellow oil. [00316] ES-LCMS m/z 283 [M+H]+. Step 3: 3-((2-chloro-3-fluoropyridin-4-yl)methyl)-7-hydroxy-3,4-dihydro-2H- benzo[e][1,3]oxazin-2-one
Figure imgf000107_0002
[00317] Into a 500 mL 3-necked round-bottom flask were added 4-({[(2-chloro-3- fluoropyridin-4-yl)methyl]amino}methyl)benzene-1,3-diol (18.0 g, 63 mmol, 1.0 eq), DCM (300 mL) and Pyridine (50.4 g, 636 mmol, 10.0 eq) at room temperature. To the above mixture was added CDI (12.4 g, 76 mmol, 1.2 eq) in portions over 30 min at 0°C. The resulting mixture was stirred for additional 2 h at room temperature. The reaction was quenched with water/ice at room temperature. The mixture residue was acidified to pH 5 with HCl (aq.). The resulting mixture was extracted with CH2Cl2 (3 x 100 mL). The combined organic layers were washed with brine (1 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1:1) to afford 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-hydroxy-4H-1,3-benzoxazin-2- one (15.0 g, 76%) as a yellow solid. [00318] ES-LCMS m/z 309 [M+H]+. Step 4: 3-((2-chloro-3-fluoropyridin-4-yl)methyl)-7-((3-fluoropyridin-2-yl)oxy)-3,4-dihydro- 2H-benzo[e][1,3]oxazin-2-one
Figure imgf000108_0001
[00319] Into a 500 mL 3-necked round-bottom flask were added 3-[(2-chloro-3-fluoropyridin- 4-yl)methyl]-7-hydroxy-4H-1,3-benzoxazin-2-one (15.0 g, 48 mmol, 1.0 eq), 2,3-difluoropyridine (8.4 g, 72 mmol, 1.5 eq), Cs2CO3 (47.5 g, 145 mmol, 3.0 eq) and DMF (200 mL) at room temperature. The resulting mixture was stirred for 16 h at room temperature. The resulting mixture was diluted with EtOAc (500 mL) and washed with brine (5 x 100 mL) and concentrated under reduced pressure then purified by silica gel column chromatography, eluted with PE / EA (1:1) to afford 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-6-[(1E)-2-[(3-fluoropyridin-2-yl)oxy]prop-1- en-1-yl]-5-methyl-4H-1,3-oxazin-2-one (11.5 g, 58%) as a yellow solid. [00320] ES-LCMS m/z 404 [M+H]+. Step 5: 3-((2-((diphenylmethylene)amino)-3-fluoropyridin-4-yl)methyl)-7-((3-fluoropyridin- 2-yl)oxy)-3,4-dihydro-2H-benzo[e][1,3]oxazin-2-one
Figure imgf000108_0002
[00321] To a solution of 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-[(3-fluoropyridin-2- yl)oxy]-4H-1,3-benzoxazin-2-one (11.5 g, 28 mmol, 1.0 eq) and diphenylmethanimine (7.7 g, 42 mmol, 1.5 eq) in dioxane (200 mL) were added Pd2(dba)3 (5.2 g, 5.6 mmol, 0.2 eq), XantPhos (6.6 g, 11.4 mmol, 0.4 eq) and Cs2CO3 (27.8 g, 85 mmol, 3.0 eq). After stirring for 2 h at 100°C under a nitrogen atmosphere, the resulting mixture was concentrated under reduced pressure. Then dissolved in EtOAc (200 mL) and washed with H2O (2 x 100 mL) and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1:1) to afford 3-({2-[(diphenylmethylidene)amino]-3-fluoropyridin-4-yl}methyl)-7-[(3-fluoropyridin- 2-yl)oxy]-4H-1,3-benzoxazin-2-one (7.6 g, 48%) as a yellow solid. [00322] ES-LCMS m/z 549 [M+H]+. Step 6: 3-((2-amino-3-fluoropyridin-4-yl)methyl)-7-((3-fluoropyridin-2-yl)oxy)-3,4-dihydro- 2H-benzo[e][1,3]oxazin-2-one
Figure imgf000109_0001
[00323] Into a 250 mL 3-necked round-bottom flask were added 3-({2- [(diphenylmethylidene)amino]-3-fluoropyridin-4-yl}methyl)-7-[(3-fluoropyridin-2-yl)oxy]-4H- 1,3-benzoxazin-2-one (7.6 g, 13.8 mmol, 1.0 eq), dioxane (40 mL) and HCl(gas)in 1,4-dioxane (60 mL) at room temperature. The resulting mixture was stirred for 15 h at room temperature. Then was concentrated under reduced pressure and was purified by trituration with Et2O (100mL). The precipitated solids were collected by filtration to afford 3-[(2-amino-3-fluoropyridin-4-yl)methyl]- 7-[(3-fluoropyridin-2-yl)oxy]-4H-1,3-benzoxazin-2-one (4.9 g, 92%) as a yellow solid. [00324] ES-LCMS m/z 385 [M+H]+. Step 7: 3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-[(3-fluoropyridin-2- yl)oxy]-4H-1,3-benzoxazin-2-one
Figure imgf000109_0002
[00325] Into a 100 mL 3-necked round-bottom flask were added 3-[(2-amino-3-fluoropyridin- 4-yl)methyl]-7-[(3-fluoropyridin-2-yl)oxy]-4H-1,3-benzoxazin-2-one (4.9 g, 12.7 mmol, 1.0 eq), DMF (50 mL) and Pyridine (10.0 g, 127 mmol, 10.0 eq) at room temperature. To the above mixture was added N-methylsulfamoyl chloride (2.5 g, 19 mmol, 1.5 eq) in ACN (5 mL) dropwise over 10 min at 0°C. The resulting mixture was stirred for additional 1 h at room temperature. After the reaction was completed, the reaction mixture was diluted with water. The solid was collected by filtration and then triturated with isopropyl ether (100mL). The solid was collected by filtration to afford 3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-[(3-fluoropyridin-2- yl)oxy]-4H-1,3-benzoxazin-2-one (3.4 g, 55%) as a white solid. [00326] ES-LCMS m/z 478.1 [M+H]+. [00327] 1H NMR (300 MHz, DMSO-d6) δ 10.43 (s, 1H), 8.10 - 8.03 (m, 1H), 7.99 - 7.93 (m, 1H), 7.92 - 7.83 (m, 1H), 7.33 - 7.27 (m, 1H), 7.26 - 7.19 (m, 1H), 7.12 - 6.93 (m, 4H), 4.71 (s, 2H), 4.57 (s, 2H), 2.56 - 2.50 (m, 3H). Synthesis of I-50 and I-54
Figure imgf000110_0001
[00328] A solution of 4-fluorobenzene-1,3-diol (55 g, 429.34 mmol, 1.00 equiv) in chlorobenzene (550 mL) was treated with acetic acid (77.35 g, 1288.03 mmol, 3.00 equiv) at room temperature under nitrogen atmosphere followed by the addition of BF3•Et2O (304.68 g, 2146.72 mmol, 5.00 equiv) dropwise at 0°C. The resulting mixture was stirred for additional 5 h at 100°C. The resulting mixture was concentrated, then layered with water (500 mL) and EtOAc (1000 mL), the water layer was extracted with EtOAc (2 x 1000 mL). The combined organic layers were washed with brine (2x400 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by trituration with diethyl ether (600 mL). This resulted in 1-(5-fluoro-2,4-dihydroxyphenyl)ethanone (54 g, 73.92%) as a dark yellow solid. Step 2:
Figure imgf000111_0001
[00329] A mixture of 1-(5-fluoro-2,4-dihydroxyphenyl)ethanone (120 g, 705.30 mmol, 1.00 equiv) and 1-(2-chloro-3-fluoropyridin-4-yl)methanamine (203.86 g, 1269.55 mmol, 1.80 equiv) in MeOH (1.20 L) was stirred for overnight at 80°C under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The precipitated solids were collected by filtration and washed with MeOH (3x100 mL). This resulted in 4-[(1E)-1-{[(2-chloro-3-fluoropyridin-4- yl)methyl]imino}ethyl]-6-fluorobenzene-1,3-diol (210 g, 95.22%) as a light yellow solid. Step 3.
Figure imgf000111_0002
[00330] To a stirred mixture of 4-[(1E)-1-{[(2-chloro-3-fluoropyridin-4- yl)methyl]imino}ethyl]-6-fluorobenzene-1,3-diol (210 g, 671.57 mmol, 1.00 equiv) in THF (4.50 L) was added NaBH(OAc)3 (1423.33 g, 6715.70 mmol, 10.00 equiv) in portions at 0°C under nitrogen atmosphere. The mixture was stirred at room temperature overnight. The reaction was quenched by the addition of Water/Ice (1 L) at 0°C. The mixture was basified to pH=8 with Na2CO3. The aqueous layer was extracted with EtOAc (4x1 L). The resulting mixture was concentrated under vacuum to afford 4-(1-{[(2-chloro-3-fluoropyridin-4-yl)methyl]amino}ethyl)- 6-fluorobenzene-1,3-diol (296.40 g, crude) as a yellow oil. Step 4.
Figure imgf000111_0003
[00331] To a stirred mixture of 4-(1-{[(2-chloro-3-fluoropyridin-4-yl)methyl]amino}ethyl)-6- fluorobenzene-1,3-diol (324 g, 1029.48 mmol, 1.00 equiv) and Et3N (312.53 g, 3088.46 mmol, 3.00 equiv) in DCM (3.20 L) were added CDI (250.40 g, 1544.23 mmol, 1.50 equiv) in portions at 0°C under nitrogen atmosphere. The resulting mixture was stirred overnight at room temperature under nitrogen atmosphere, then 1N aqueous HCl solution (1 L) was added and layered, the aqueous layer was extracted with DCM (3x500 mL). The combined organic layers were dried and concentrated under reduced pressure. The residue was purified by trituration with diethyl ether (500 mL). This resulted in 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-6-fluoro-7-hydroxy-4- methyl-4H-1,3-benzoxazin-2-one (120 g, 34.15%) as a yellow white solid. Step 5.
Figure imgf000112_0001
[00332] To a stirred mixture of 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-6-fluoro-7-hydroxy- 4-methyl-4H-1,3-benzoxazin-2-one (71 g, 208.39 mmol, 1.00 equiv) and K2CO3 (86.40 g, 625.16 mmol, 3.00 equiv) in DMSO (710 mL) was added 2,3-difluoropyridine (47.96 g, 416.78 mmol, 2.00 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred overnight at 80°C under nitrogen atmosphere. The mixture was cooled to room temperature and diluted with water/ice (1.50 L) at 0°C, then extracted with EtOAc (3x800 mL). The combined organic layers were washed with brine (2x300 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by trituration with diethyl ether (500 mL). This resulted in 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-6-fluoro- 7-[(3-fluoropyridin-2-yl)oxy]-4-methyl-4H-1,3-benzoxazin-2-one (57.21 g, 63.01%) as an off- white solid. Step 6.
Figure imgf000113_0001
[00333] To a stirred mixture of 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-6-fluoro-7-[(3- fluoropyridin-2-yl)oxy]-4-methyl-4H-1,3-benzoxazin-2-one (55 g, 126.20 mmol, 1.00 equiv), Xantphos (29.21 g, 50.48 mmol, 0.40 equiv) and Cs2CO3 (123.36 g, 378.62 mmol, 3.00 equiv) in 1,4-dioxane (1650 mL) were added Pd2(dba)3 (23.11 g, 25.24 mmol, 0.30 equiv) and diphenylmethanimine (34.31 g, 189.31 mmol, 1.50 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 3 h at 100°C under nitrogen atmosphere. The mixture was cooled to room temperature and diluted with EtOAc (1 L) and water (500 mL). The precipitated solids were filtered off and the filtrate was layered. The aqueous layer was extracted with EtOAc (3x500 mL). The combined organic layers were dried and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1:1) to afford 3-({2-[(diphenylmethylidene)amino]-3-fluoropyridin-4-yl}methyl)-6-fluoro-7-[(3- fluoropyridin-2-yl)oxy]-4-methyl-4H-1,3-benzoxazin-2-one (33.81 g, 46.10%) as a brown solid. Step 7.
Figure imgf000113_0002
[00334] To a stirred mixture of 3-({2-[(diphenylmethylidene)amino]-3-fluoropyridin-4- yl}methyl)-6-fluoro-7-[(3-fluoropyridin-2-yl)oxy]-4-methyl-4H-1,3-benzoxazin-2-one (35 g, 60.29 mmol, 1.00 equiv) in DCM (70 mL) were added HCl (gas) in 1,4-dioxane (4 M) (350 mL) dropwise at 0°C under nitrogen atmosphere. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The mixture was concentrated under reduced pressure. The residue was purified by trituration with diethyl ether (350 mL). This resulted in 3- [(2-amino-3-fluoropyridin-4-yl)methyl]-6-fluoro-7-[(3-fluoropyridin-2-yl)oxy]-4-methyl-4H- 1,3-benzoxazin-2-one (24.12 g, 96.10%) as an off-white solid. Step 8 and 9.
Figure imgf000114_0001
[00335] A solution of 3-[(2-amino-3-fluoropyridin-4-yl)methyl]-6-fluoro-7-[(3-fluoropyridin- 2-yl)oxy]-4-methyl-4H-1,3-benzoxazin-2-one (24.00 g, 57.642 mmol, 1.00 equiv) in DMF (240 mL) was treated with pyridine (45.60 g, 576.420 mmol, 10.00 equiv) at 0°C under nitrogen atmosphere followed by the addition of N-methylsulfamoyl chloride (14.94 g, 115.284 mmol, 2.00 equiv) dropwise at 0°C. The resulting mixture was stirred for 1 h at 0°C. The solution was poured slowly to water/ice (580 mL) with stirring. The precipitated solids were collected by filtration and washed with water (3x100 mL). The filter cake was dried in vacuum and purified by trituration with EtOAc (200 mL). The racemic mixture (18.7 g) was finally separated by SFC with the following conditions (Column: CHIRALPAK IH, 3*25 cm, 5 μm; Mobile Phase A: CO2, Mobile Phase B: MeOH (0.1% 2M NH3-MeOH); Flow rate: 80 mL/min; Gradient: isocratic 30% B; Wave Length: 220 nm; Retention Time 1 (min): 5.1; Retention Time 2 (min): 6; Sample Solvent: MeOH (0.1% 2M NH3-MeOH); Injection Volume: 3 mL; Number of Runs: 140). After remove of solvents, this resulted in (4R)-6-fluoro-3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4- yl}methyl)-7-[(3-fluoropyridin-2-yl)oxy]-4-methyl-4H-1,3-benzoxazin-2-one (enantiomer 1, the first peak, 5.4163 g, 18.44%) as a white solid and (4S)-6-fluoro-3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-[(3-fluoropyridin-2-yl)oxy]-4-methyl-4H-1,3- benzoxazin-2-one (enantiomer 2, the second peak, 5.1992 g, 17.70%) as a white solid. Step 10.
Figure imgf000114_0002
[00336] To a stirred solution of 2-chloro-3-fluoropyridine-4-carbaldehyde (210 g, 1316.28 mmol, 1.00 equiv) in methanol (2100 mL) was added NaBH4 (49.80 g, 1316.28 mmol, 1.00 equiv) in portions at 0°C under nitrogen atmosphere. The resulting mixture was stirred for additional 1 h at room temperature. The solution was diluted with water/ice (2000 mL) and extracted with EtOAc (5x800 mL). The combined organic layers were washed with brine (1x1000 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in (2-chloro-3-fluoropyridin-4-yl)methanol (231 g, crude) as a white solid. Step 11.
Figure imgf000115_0001
[00337] To a stirred solution of (2-chloro-3-fluoropyridin-4-yl)methanol (231 g, 1429.80 mmol, 1.00 equiv) in ACN (4.6 L) was added PBr3 (387.03 g, 1429.80 mmol, 1.00 equiv) dropwise at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 16 h at room temperature. The reaction was quenched by the addition of water/ice (6 L) at 0°C. The resulting mixture was extracted with EtOAc (3x3 L). The combined organic layers were washed with brine (1x3 L), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 4-(bromomethyl)-2-chloro-3-fluoropyridine (289.5 g, 90.21%) as a dark red oil. Step 12.
Figure imgf000115_0002
[00338] To a stirred solution of 4-(bromomethyl)-2-chloro-3-fluoropyridine (289 g, 1287.53 mmol, 1.00 equiv) in methanol (4.00 L) was added NH3 (g) in MeOH (7 N) (8.50 L) dropwise at room temperature under nitrogen atmosphere. The reaction mixture was stirred overnight at room temperature. `The resulting solution was concentrated and diluted with water/ice (4 L), then basified to pH=8 with saturated Na2CO3 (aq.). The resulting mixture was extracted with iPrOH:CH2Cl2=1:5 (5x3 L). The combined organic layers were washed with brine (1x2 L), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 1-(2-chloro-3-fluoropyridin-4-yl)methanamine (175 g, 84.64%) as a yellow oil. Synthesis of I-147
Figure imgf000116_0001
Step 1: 3-((2-chloro-3-fluoropyridin-4-yl)methyl)-4-methyl-7-((tetrahydro-2H-pyran-2-yl)oxy)- 3,4-dihydro-2H-benzo[e][1,3]oxazin-2-one
Figure imgf000116_0002
[00339] To a stirred mixture of 4-methyl-7-(oxan-2-yloxy)-3,4-dihydro-1,3-benzoxazin-2-one (10.0 g, 37 mmol, 1.0 equiv) and cesium fluoride (17.3 g, 113 mmol, 3.0 equiv) in DMF (100.0 mL) was added 4-(bromomethyl)-2-chloro-3-fluoropyridine (12.8 g, 56 mmol, 1.5 equiv) at 0°C. The mixture was stirred for 0.5 h at room temperature. The resulting mixture was diluted with EA (150 mL) and washed with brine (3 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE and THF (1:1) to afford 3-[(2-chloro-3-fluoropyridin-4- yl)methyl]-4-methyl-7-(oxan-2-yloxy)-4H-1,3-benzoxazin-2-one (8.3 g, 53%) as a light yellow solid. ES-LCMS m/z 407 [M+H] + Step 2: 3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-7-(oxan-2- yloxy)-4H-1,3-benzoxazin-2-one
Figure imgf000117_0001
[00340] A mixture of 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-4-methyl-7-(oxan-2-yloxy)- 4H-1,3-benzoxazin-2-one (8.3 g, 20 mmol, 1.0 equiv), methyl(sulfamoyl)amine (2.7 g, 24 mmol, 1.2 equiv), Pd2(dba)3 (3.7 g, 4 mmol, 0.2 equiv), tBuXantphos (5.6 g, 8 mmol, 0.4 equiv) and K3PO4 (12.9 g, 61 mmol, 3.0 equiv) in t-BuOH (100 mL) was stirred for 1 h at 100°C under nitrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with EA (3 x 50 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE and THF (1:1) to afford 3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-7-(oxan-2-yloxy)-4H-1,3-benzoxazin- 2-one (4.2 g, 42%) as a light brown solid. ES-LCMS m/z 481 [M+H] + Step 3: 3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-hydroxy-4-methyl-4H- 1,3-benzoxazin-2-one
Figure imgf000117_0002
[00341] To a stirred mixture of 3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)- 4-methyl-7-(oxan-2-yloxy)-4H-1,3-benzoxazin-2-one (4.2 g, 8.7 mmol, 1.0 equiv) in DCM (30.0 mL) was added HCl (4M in dioxane, 30.0 mL) at room temperature. The resulting mixture was stirred for additional 2 h at room temperature. Then concentrated under reduced pressure. The residue was purified by trituration with diethyl ether (100 mL). This resulted in 3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-hydroxy-4-methyl-4H-1,3-benzoxazin-2-one (1.8 g, 51%) as a light yellow solid. ES-LCMS m/z 397 [M+H]+. Step 4: 7-[(5-bromo-1-{[2-(trimethylsilyl)ethoxy]methyl}-1,2,4-triazol-3-yl)oxy]-3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-5-methyl-4H-1,3-benzoxazin-2-one
Figure imgf000118_0001
[00342] A mixture of 3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7- hydroxy-4-methyl-4H-1,3-benzoxazin-2-one (1.6 g, 3.9 mmol, 1.0 equiv) and potassium carbonate (1.6 g, 11.7 mmol, 3.0 equiv) in DMF (16.0 mL) was stirred for 2 h at 80°C. The resulting mixture was diluted with EA (100 mL) and washed with brine (3 x 50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE and THF (1:1) to afford 7-[(5-bromo-1-{[2- (trimethylsilyl)ethoxy]methyl}-1,2,4-triazol-3-yl)oxy]-3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-5-methyl-4H-1,3-benzoxazin-2-one (950 mg, 36%) as an off-white solid. ES-LCMS m/z 673 [M+H] +. Step 5: 3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-{[5-(methoxymethyl)-2- {[2-(trimethylsilyl)ethoxy]methyl}-1,2,4-triazol-3-yl]oxy}-4-methyl-4H-1,3-benzoxazin-2-one
Figure imgf000118_0002
[00343] Into a 8 mL vial were added 7-[(5-bromo-2-{[2-(trimethylsilyl)ethoxy]methyl}-1,2,4- triazol-3-yl)oxy]-3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-4H- 1,3-benzoxazin-2-one (150 mg, 0.2 mmol, 1.0 equiv), potassium trifluoro(methoxymethyl)boranuide (101 mg, 0.7 mmol, 3.0 equiv), Ir[dF(CF3)ppy]2(dtbpy)PF6 (50 mg, 0.05 mmol, 0.2 equiv), Ni(DME)Cl2 & dtbpy (48 mg, 0.09 mmol, 0.4 equiv), 2,6-lutidine (71 mg, 0.7 mmol, 3.0 equiv), dioxane (0.8 mL) and DMAC (0.2 mL) at room temperature. The mixture was stirred for 2 h at room temperature in radiation of 20 W Blue LEDs under nitrogen atmosphere. The resulting mixture was diluted with EtOAc (20 mL) and washed with water (10 mL). After dried over Na2SO4, the organic layer was concentrated under reduced pressure and purified by silica gel column chromatography, eluted with PE and EA (1:1) to afford 3-({3-fluoro- 2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-{[5-(methoxymethyl)-2-{[2- (trimethylsilyl)ethoxy]methyl}-1,2,4-triazol-3-yl]oxy}-4-methyl-4H-1,3-benzoxazin-2-one potassium (37 mg, 24%) as a yellow solid. ES-LCMS m/z 638 [M+H] +. Step 6: 3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-{[5-(methoxymethyl)- 2H-1,2,4-triazol-3-yl]oxy}-4-methyl-4H-1,3-benzoxazin-2-one
Figure imgf000119_0001
[00344] A mixture of 3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-{[5- (methoxymethyl)-2-{[2-(trimethylsilyl)ethoxy]methyl}-1,2,4-triazol-3-yl]oxy}-4-methyl-4H- 1,3-benzoxazin-2-one (150 mg, 0.23 mmol, 1.0 equiv) and HCl (4M in i-PrOH, 2.0 mL) was stirred for 1 h at room temperature. The resulting mixture was concentrated under vacuum. The crude product was purified by reverse phase flash with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% NH3.H2O), 10% to 60% gradient in 15 min; detector, UV 254 nm. This resulted in 3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-{[5- (methoxymethyl)-2H-1,2,4-triazol-3-yl]oxy}-4-methyl-4H-1,3-benzoxazin-2-one (15.8 mg, 13%) as an off-white solid. ES-LCMS m/z 508 [M+H] + Step 7: (4S)-3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-{[5- (methoxymethyl)-2H-1,2,4-triazol-3-yl]oxy}-4-methyl-4H-1,3-benzoxazin-2-one
Figure imgf000119_0002
[00345] The racemate product (15.8 mg, 0.03 mmol) was separated by chiral SFC with the following conditions (Column: CHIRALPAK IG, 3*25 cm, 5 μm; Mobile Phase A: CO2, Mobile Phase B: MEOH: DCM=2: 1(0.1% 2M NH3-MeOH); Flow rate: 80 mL/min; Gradient: isocratic 50% B; Wave Length: 230 nm; RT1(min): 5.27; Sample Solvent: MeOH-HPLC; Injection Volume: 1.5 mL; Number Of Runs: 5) to afford (4S)-3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4- yl}methyl)-7-{[5-(methoxymethyl)-2H-1,2,4-triazol-3-yl]oxy}-4-methyl-4H-1,3-benzoxazin-2- one (4.9 mg, 28%) as a yellow solid. ES-LCMS m/z 508 [M+H]+.1H NMR (300 MHz, DMSO-d6) δ 13.80 (s, 1H), 7.98 - 7.92 (m, 1H), 7.32 (d, J = 8.2 Hz, 1H), 7.02 - 6.93 (m, 2H), 6.82 - 6.76 (m, 1H), 4.88 (d, J = 16.7 Hz, 1H), 4.69 - 4.63 (m, 1H), 4.53 (d, J = 16.5 Hz, 1H), 4.46 (s, 2H), 3.34 (s, 3H), 2.52 - 2.50 (m, 3H), 1.42 (d, J = 6.4 Hz, 3H). Synthesis of I-146
Figure imgf000120_0001
Step 1: (4R)-3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-{[5- (methoxymethyl)-2H-1,2,4-triazol-3-yl]oxy}-4-methyl-4H-1,3-benzoxazin-2-one
Figure imgf000120_0002
[00346] The racemate product (15.8 mg, 0.03 mmol, 1.0 equiv) was separated by chiral SFC with the following conditions (Column: CHIRALPAK IG, 3*25 cm, 5 μm; Mobile Phase A: CO2, Mobile Phase B: MEOH: DCM=2: 1(0.1% 2M NH3-MeOH); Flow rate: 80 mL/min; Gradient: isocratic 50% B; Wave Length: 230 nm; RT2(min): 7.17; Sample Solvent: MeOH--HPLC; Injection Volume: 1.5 mL; Number Of Runs: 5) to afford (4R)-3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-{[5-(methoxymethyl)-2H-1,2,4-triazol-3- yl]oxy}-4-methyl-4H-1,3-benzoxazin-2-one (6.9 mg, 42%) as a yellow solid. ES-LCMS m/z 508 [M+H]+.1H NMR (300 MHz, DMSO-d6) δ 13.80 (s, 1H), 10.44 (br, s, 1H), 8.00 (d, J = 5.2 Hz, 1H), 7.33 (d, J = 8.3 Hz, 1H), 7.05 - 6.79 (m, 4H), 4.90 (d, J = 16.3 Hz, 1H), 4.73 - 4.63 (m, 1H), 4.55 (d, J = 16.4 Hz, 1H), 4.46 (s, 2H), 3.34 (s, 3H), 2.53 - 2.52 (m, 3H), 1.43 (d, J = 6.5 Hz, 3H). Synthesis of I-149
Figure imgf000121_0001
Step 1:3-((2-chloro-3-fluoropyridin-4-yl)methyl)-4-methyl-7-((tetrahydro-2H-pyran-2-yl)oxy)- 3,4-dihydro-2H-benzo[e][1,3]oxazin-2-one
Figure imgf000122_0001
[00347] In a 250-mL round bottom flask was placed a mixture of 4-methyl-7-(oxan-2-yloxy)- 3,4-dihydro-1,3-benzoxazin-2-one (6.00 g, 22.78 mmol, 1.0 eq) and CsF (10.38 g, 68.36 mmol, 3.0 eq) in DMF (60.0 mL). Then 4-(bromomethyl)-2-chloro-3-fluoropyridine (7.67 g, 34.18 mmol, 1.5 eq) was added in portions at 0°C. The reaction mixture was stirred for 1.5 h at room temperature. The resulting mixture was diluted with EtOAc (300 mL), washed with water (3 x 100 mL) and brine (100mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / THF (70:30) to afford 3-[(2-chloro-3-fluoropyridin-4- yl)methyl]-4-methyl-7-(oxan-2-yloxy)-4H-1,3-benzoxazin-2-one (10.00 g, 95%) as a yellow oil. ES-LCMS m/z 407 [M+H] + Step 2: 3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-7-(oxan-2- yloxy)-4H-1,3-benzoxazin-2-one
Figure imgf000122_0002
[00348] In a 250-mL round bottom flask was placed a mixture of 3-[(2-chloro-3-fluoropyridin- 4-yl)methyl]-4-methyl-7-(oxan-2-yloxy)-4H-1,3-benzoxazin-2-one (9.00 g, 22.12 mmol, 1.0 eq), methyl(sulfamoyl)amine (2.92 g, 26.54 mmol, 1.2 eq), K3PO4 (9.39 g, 44.24 mmol, 2 eq) and t- BuOH (90 mL). Then t-BuXPhos (1.88 g, 4.42 mmol, 0.2 eq) and Pd2(dba)3 (4.05 g, 4.42 mmol, 0.2 eq) were added under nitrogen atmosphere. The reaction mixture was stirred for 30 min at 100 °C under nitrogen atmosphere. The resulting mixture was cooled to room temperature, diluted with EtOAc (100 mL) and water (100 mL) with stirring. The layers were separated and the aqueous layer extracted with EtOAc (2 x 100 mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / THF (50:50) to afford 3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-7- (oxan-2-yloxy)-4H-1,3-benzoxazin-2-one (2.20 g, 20%) as a yellow oil. ES-LCMS m/z 481 [M+H] + Step 3: 3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-hydroxy-4-methyl-4H- 1,3-benzoxazin-2-one
Figure imgf000123_0001
[00349] In a 50-mL round bottom flask was placed a solution of 3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-7-(oxan-2-yloxy)-4H-1,3-benzoxazin- 2-one (2.00 g, 4.16 mmol, 1.0 eq) in DCM (20.0 mL). The solution was cooled to 0 °C and HCl (4M in 1,4-dioxane, 20.0 mL) were added. The reaction mixture was stirred for 2h at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by trituration with diethyl ether (100 mL). This resulted in 3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-hydroxy-4-methyl-4H-1,3-benzoxazin-2-one (1.30 g, 78%) as a yellow solid. ES-LCMS m/z 397 [M+H]+. Step 4: 7-[(5-chloro-2-{[2-(trimethylsilyl)ethoxy]methyl}-1,2,4-triazol-3-yl)oxy]-3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-4H-1,3-benzoxazin-2-one
Figure imgf000123_0002
[00350] Into a 40 mL vial was placed a mixture of 3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-hydroxy-4-methyl-4H-1,3-benzoxazin-2-one (800 mg, 2.02 mmol, 1.0 eq), 3,5-dichloro-1-{[2-(trimethylsilyl)ethoxy]methyl}-1,2,4-triazole (811 mg, 3.02 mmol, 1.5 eq), K2CO3 (836 mg, 6.05 mmol, 3 eq) and DMF (8.0 mL). The mixture was stirred for 1.5 h at 100°C. The resulting mixture was cooled to room temperature, diluted with EtOAc (100 mL), washed with water (3 x 30 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / THF (20:80) to afford 7-[(5-chloro-2-{[2- (trimethylsilyl)ethoxy]methyl}-1,2,4-triazol-3-yl)oxy]-3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-4H-1,3-benzoxazin-2-one (400 mg, 31%) as a yellow oil. ES-LCMS m/z 628 [M+H] +. Step 5: 7-[(5-chloro-2H-1,2,4-triazol-3-yl)oxy]-3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-4H-1,3-benzoxazin-2-one
Figure imgf000124_0001
[00351] Into a 40 mL vial was placed a solution of 7-[(5-chloro-2-{[2- (trimethylsilyl)ethoxy]methyl}-1,2,4-triazol-3-yl)oxy]-3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-4H-1,3-benzoxazin-2-one (400 mg, 0.63 mmol, 1.0 eq) in HCl (4M in iPrOH, 4.0 mL). The solution was stirred for 1 h at room temperature. The resulting solution was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Welch Utimate AQ-C18, 50*250mm*10μm; Mobile Phase A: Water(10 mmol/L NH4HCO3+0.1%NH3·H2O), Mobile Phase B: ACN; Flow rate: 80 mL/min; Gradient: 5% B to 65% B in 15 min, 65% B; Wave Length: 220 nm) to afford 7-[(5-chloro-2H-1,2,4-triazol-3-yl)oxy]-3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-4H-1,3-benzoxazin-2-one (75.00 mg, 23%) as a white solid. ES-LCMS m/z 498 [M+H] +. Step 6: (4S)-7-[(5-chloro-2H-1,2,4-triazol-3-yl)oxy]-3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-4H-1,3-benzoxazin-2-one
Figure imgf000124_0002
[00352] The racemate product 7-[(5-chloro-2H-1,2,4-triazol-3-yl)oxy]-3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-4H-1,3-benzoxazin-2-one (75.00 mg) was separated by chiral SFC with the following conditions (Column: CHIRAL ART Cellulose- SC, 3*25 cm, 5 μm; Mobile Phase A: CO2, Mobile Phase B: MeOH: DCM=1: 1(0.1% 2M NH3- MEOH); Flow rate: 80 mL/min; Gradient: isocratic 50% B; Column Temperature(℃): 35; Back Pressure(bar): 100; Wave Length: 220 nm; RT1(min): 4.2; Sample Solvent: MeOH: DCM=2: 1; Injection Volume: 5 mL; Number Of Runs: 5.) afford (4S)-7-[(5-chloro-2H-1,2,4-triazol-3- yl)oxy]-3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-4H-1,3- benzoxazin-2-one (14.90 mg, 19%) as a white solid. ES-LCMS m/z 498.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 14.14 (br, s, 1H), 10.44 (br, s, 1H), 8.02 (d, J = 5.1 Hz, 1H), 7.40 - 7.34 (m, 1H), 7.16 - 7.07 (m, 2H), 7.03 - 6.98 (m, 1H), 6.96 - 6.89 (m, 1H), 4.90 (d, J = 16.4 Hz, 1H), 4.76 - 4.67 (m, 1H), 4.56 (d, J = 16.4 Hz, 1H), 2.52 - 2.47 (m, 3H), 1.44 (d, J = 6.5 Hz, 3H). Step 7: 3,5-dichloro-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-1,2,4-triazole
Figure imgf000125_0001
[00353] Into a 40mL vial, was added a solution of 3,5-dichloro-1H-1,2,4-triazole (1.20 g, 8.67 mmol, 1.0 eq) in ACN (12.0 mL), then SEM-Cl (2.18 g, 13.05 mmol, 1.5 eq) and K2CO3 (3.61 g, 26.10 mmol, 3 eq) were added. The mixture was stirred for 1h at room temperature. The resulting mixture was diluted with EtOAc (50 mL) and water (50 mL) with stirring. The layers were separated and the aqueous layer extracted with EtOAc (2 x 50 mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / THF (90:10) to afford 3,5-dichloro-1-{[2- (trimethylsilyl)ethoxy]methyl}-1,2,4-triazole (1.00 g, 42%) as a colorless oil. ES-LCMS m/z 268 [M+H] +. Synthesis of I-148
Figure imgf000125_0002
Step 1: (4R)-7-[(5-chloro-2H-1,2,4-triazol-3-yl)oxy]-3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-4H-1,3-benzoxazin-2-one
Figure imgf000125_0003
[00354] The racemate product 7-[(5-chloro-2H-1,2,4-triazol-3-yl)oxy]-3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-4H-1,3-benzoxazin-2-one (75.00 mg) was separated by chiral SFC with the following conditions (Column: CHIRAL ART Cellulose- SC, 3*25 cm, 5 μm; Mobile Phase A: CO2, Mobile Phase B: MeOH: DCM=1: 1(0.1% 2M NH3- MEOH); Flow rate: 80 mL/min; Gradient: isocratic 50% B; Column Temperature(℃): 35; Back Pressure(bar): 100; Wave Length: 220 nm; RT2(min): 5; Sample Solvent: MeOH: DCM=2: 1; Injection Volume: 5 mL; Number Of Runs: 5.) afford (4R)-7-[(5-chloro-2H-1,2,4-triazol-3- yl)oxy]-3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-4-methyl-4H-1,3- benzoxazin-2-one (16.30 mg, 23%) as a white solid. ES-LCMS m/z 498.1 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 14.14 (br, s, 1H), 10.45 (br, s, 1H), 8.02 (d, J = 5.1 Hz, 1H), 7.40 - 7.34 (m, 1H), 7.15 - 7.07 (m, 2H), 7.02 - 6.97 (m, 1H), 6.96 - 6.89 (m, 1H), 4.90 (d, J = 16.4 Hz, 1H), 4.76 - 4.67 (m, 1H), 4.56 (d, J = 16.5 Hz, 1H), 2.53 - 2.50 (m, 3H), 1.44 (d, J = 6.4 Hz, 3H). Synthesis of I-142
Figure imgf000127_0002
Step 1: 3,5-dimethoxypyridine-2-carbonitrile O O
Figure imgf000127_0001
[00355] To a stirred solution of 3,5-difluoropyridine-2-carbonitrile (99.0 g, 706 mmol, 1.0 equiv) in MeOH (4.0 L) was added MeONa (152.0 g, 2826 mmol, 4.0 equiv) in portions at 0℃ under nitrogen atmosphere. The resulting mixture was stirred for 1 h at 80℃. The mixture was allowed to cool down to 5℃. The precipitated solids were collected by filtration and washed with MeOH (1.0 L). The filter cake was purified by trituration with 1M aqueous HCl (600 mL). The precipitated solids were collected by filtration and washed with H2O (100 mL), after dried, 3,5- dimethoxypyridine-2-carbonitrile (112 g, 96%) was obtained as a white solid. ES-LCMS m/z 165 [M+H] + Step 2: 1-(3,5-dimethoxypyridin-2-yl)methanamine
Figure imgf000128_0001
[00356] To a stirred solution of LAH (293 mL, 292 mmol, 1.2 equiv) in THF (50.0 mL) were added 3,5-dimethoxypyridine-2-carbonitrile (40.0 g, 243 mmol, 1.0 equiv) in THF (450.0 mL) dropwise at 0℃ under nitrogen atmosphere. The resulting mixture was stirred for 1 h at room temperature. The reaction was quenched by the addition of Na2SO4.10H2O at 0℃. After stirred at room temperature overnight, the resulting mixture was filtered, the filter cake was washed with THF (100 mL). The filtrate was concentrated under reduced pressure. This resulted in 1-(3,5- dimethoxypyridin-2-yl)methanamine (32 g, 78%) as a brown oil. ES-LCMS m/z 169 [M+H] + Step 3: [(2-chloro-3-fluoropyridin-4-yl)methyl][(3,5-dimethoxypyridin-2-yl)methyl]amine
Figure imgf000128_0002
[00357] A solution of 1-(3,5-dimethoxypyridin-2-yl)methanamine (20.1 g, 119 mmol, 1.0 equiv) and 2-chloro-3-fluoropyridine-4-carbaldehyde (17.2 g, 107 mmol, 0.9 equiv) in MeOH (200.0 mL) was stirred for 1 h at 60℃. The resulting mixture was concentrated under reduced pressure. The residue was dissolved in DCM (400.0 mL), then NaBH(OAc)3 (75.9 g, 358 mmol, 3.0 equiv) was added in portions at 0℃. The mixture was stirred for additional 30 min at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was dissolved in 1M aqueous HCl (200 mL) and washed with ethyl ether (200 mL). The aqueous layer was basified to pH 8 with NaOH (aq.) and extracted with EtOAc (3 x 200 mL). The combined organic layers were washed with brine (200 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product (29.8 g) was used in the next step directly without further purification. ES-LCMS m/z 312 [M+H]+. Step 4: 2-({[(2-chloro-3-fluoropyridin-4-yl)methyl]amino}methyl)pyridine-3,5-diol
Figure imgf000129_0001
[00358] To a stirred solution of [(2-chloro-3-fluoropyridin-4-yl)methyl][(3,5- dimethoxypyridin-2-yl)methyl]amine (29.8 g, 95 mmol, 1.0 equiv) in DCE (400 mL) was added BBr3 (239.5 g, 956 mmol, 10.0 equiv) dropwise at room temperature. The mixture was stirred for 16 h at 80℃ under an atmosphere of N2. The resulting mixture was concentrated under reduced pressure and treatment with water/ice at 0℃, then washed with ethyl ether (2 x 250 mL). The aqueous layer was basified to pH 8 with NaOH (aq.) and extracted with EtOAc (3 x 250 mL). The combined organic layers were washed with brine (250 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product (25.3 g) was used in the next step directly without further purification. ES-LCMS m/z 284 [M+H] +. Step 5: 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-hydroxy-4H-pyrido[2,3-e][1,3]oxazin-2- one
Figure imgf000129_0002
[00359] To a stirred solution of 2-({[(2-chloro-3-fluoropyridin-4- yl)methyl]amino}methyl)pyridine-3,5-diol (25.3 g, 89 mmol, 1.0 equiv) and TEA (45.1 g, 446 mmol, 5.0 equiv) in THF (520.0 mL) was added CDI (17.3 g, 107 mmol, 1.2 equiv) in portions at 0℃. The solution was stirred for additional 0.5 h at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was dissolved in 1 M aqueous HCl (500 mL) and extracted with EtOAc (3 x 500 mL). The combined organic layers were washed with brine (500 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product (30.0 g) was purified by Prep-HPLC with the following conditions (Column: Welch Xtimate 150-DAC; Mobile Phase A: Water (0.1%TFA), Mobile Phase B: ACN; Flow rate: 400 ml/min; Gradient: 5% B to 40% B in 38min, 40% to 40% B in 10min;) to afford 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-hydroxy-4H-pyrido[2,3-e][1,3]oxazin-2-one (13 g, 47%) as a brown solid. ES-LCMS m/z 310 [M+H] +. Step 6: 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-[(3-fluoropyridin-2-yl)oxy]-4H-pyrido[2,3- e][1,3]oxazin-2-one
Figure imgf000130_0001
[00360] To a stirred solution of 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-hydroxy-4H- pyrido[2,3-e][1,3]oxazin-2-one (7.2 g, 23 mmol, 1.0 equiv) and 3-fluoro-2-iodopyridine (10.4 g, 46 mmol, 2.0 equiv) in DMSO (200.0 mL) were added CuI (4.4 g, 23 mmol, 1.0 equiv), 2,2,6,6- tetramethylheptane-3,5-dione (6.4 g, 34 mmol, 1.5 equiv) and K3PO4 (14.8 g, 69 mmol, 3.0 equiv) at room temperature under nitrogen atmosphere. The mixture was stirred for 4 h at 100℃. The resulting mixture was cooled to room temperature and diluted with EtOAc (400 mL) and H2O (500 ml). After filtered, the filter cake was washed with EtOAc (80 mL). The filtrate was layered, the aqueous layer was extracted with EtOAc (3 x 500 mL). The combined organic layers were washed with brine (500 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (3:5) to afford 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-[(3-fluoropyridin-2- yl)oxy]-4H-pyrido[2,3-e][1,3]oxazin-2-one (3.0 g, 31%) as a white solid. ES-LCMS m/z 405 [M+H] +. Step 7: 3-({2-[(diphenylmethylidene)amino]-3-fluoropyridin-4-yl}methyl)-7-[(3-fluoropyridin- 2-yl)oxy]-4H-pyrido[2,3-e][1,3]oxazin-2-one
Figure imgf000130_0002
[00361] To a stirred solution of 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-[(3-fluoropyridin- 2-yl)oxy]-4H-pyrido[2,3-e][1,3]oxazin-2-one (2.7 g, 6.8 mmol, 1.0 equiv), Xantphos (0.79 g, 1.4 mmol, 0.2 equiv), Pd2(dba)3 (0.93 g, 1.0 mmol, 0.15 equiv) and Cs2CO3 (4.4 g, 13.6 mmol, 2.0 equiv) in dioxane (30.0 mL) was added diphenylmethanimine (1.8 g, 10.2 mmol, 1.5 equiv) at room temperature under nitrogen atmosphere. The mixture was stirred for 2 h at 100℃. The resulting mixture was cooled to room temperature, diluted with EtOAc (60 mL) and H2O (50 ml). After layered, the aqueous layer was extracted with EtOAc (3 x 60 mL). The combined organic layers were washed with brine (60 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / THF (5:2) to afford 3-({2-[(diphenylmethylidene)amino]-3- fluoropyridin-4-yl}methyl)-7-[(3-fluoropyridin-2-yl)oxy]-4H-pyrido[2,3-e][1,3]oxazin-2-one (1.6 g, 43%) as a brown yellow solid. ES-LCMS m/z 550 [M+H] +. Step 8: 3-[(2-amino-3-fluoropyridin-4-yl)methyl]-7-[(3-fluoropyridin-2-yl)oxy]-4H-pyrido[2,3- e][1,3]oxazin-2-one
Figure imgf000131_0001
[00362] To a stirred solution of 3-({2-[(diphenylmethylidene)amino]-3-fluoropyridin-4- yl}methyl)-7-[(3-fluoropyridin-2-yl)oxy]-4H-pyrido[2,3-e][1,3]oxazin-2-one (7.2 g, 13 mmol, 1.0 equiv) in DCM (50.0 mL) was added HCl( 4M in 1,4-dioxane, 72.0 mL, 288 mmol, 22.0 equiv) dropwise at 0℃ under nitrogen atmosphere. The solution was stirred for 1 h at room temperature. The resulting mixture was concentrated under vacuum. The residue was diluted with H2O (80 mL) and extracted with EtOAc (2 x 30 mL). The aqueous layer was collected and basified to pH 7 with NaHCO3, extracted with CH2Cl2 (3 x 80 mL). The combined organic layers were washed with brine (80 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 3-[(2-amino-3-fluoropyridin-4-yl)methyl]-7-[(3-fluoropyridin- 2-yl)oxy]-4H-pyrido[2,3-e][1,3]oxazin-2-one (3.7 g, 74%) as a white solid. ES-LCMS m/z 386 [M+H] +. Step 9: 3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-[(3-fluoropyridin-2- yl)oxy]-4H-pyrido[2,3-e][1,3]oxazin-2-one
Figure imgf000131_0002
[00363] To a stirred solution of 3-[(2-amino-3-fluoropyridin-4-yl)methyl]-7-[(3-fluoropyridin- 2-yl)oxy]-4H-pyrido[2,3-e][1,3]oxazin-2-one (3.5 g, 9.1 mmol, 1.0 equiv) and pyridine (7.2 g, 90 mmol, 10.0 equiv) in DMF (35.0 mL) was added N-methylsulfamoyl chloride (3.3 g, 25 mmol, 28 equiv) dropwise at 0℃ under nitrogen atmosphere The resulting mixture was stirred for 30 min at 0℃. The reaction was quenched by poured into water (70 mL) with stirring. The resulting mixture was extracted with EtOAc (3 x 80 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by trituration with EtOAc (25 mL). This resulted in 3- ({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-[(3-fluoropyridin-2-yl)oxy]-4H- pyrido[2,3-e][1,3]oxazin-2-one (3.67 g, 83%) as an off-white solid. ES-LCMS m/z 479.2 [M+H] +.1H NMR (300 MHz, DMSO-d6) δ 10.40 (s, 1H), 8.32 (d, J = 2.3 Hz, 1H), 8.08 - 8.02 (m, 1H), 7.98 - 7.86 (m, 2H), 7.63 (d, J = 2.3 Hz, 1H), 7.30 - 7.22 (m, 1H), 7.19 - 7.12 (m, 1H), 7.04 - 7.00 (m, 1H), 4.76 - 4.64 (m, 4H), 2.54 - 2.50 (m, 3H). Synthesis of I-150
Figure imgf000132_0001
Step 1: 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-[(2-methyl-1,2,4-triazol-3-yl)oxy]-4H-1,3- benzoxazin-2-one
Figure imgf000132_0002
[00364] Into a 8 mL vial were added 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-hydroxy-4H- 1,3-benzoxazin-2-one (200 mg, 0.64 mmol, 1.0 equiv), 5-bromo-1-methyl-1,2,4-triazole (125 mg, 0.77 mmol, 1.2 equiv), RuPhos G3 (108 mg, 0.13 mmol, 0.2 equiv), Cs2CO3 (633 mg, 1.94 mmol, 3.0 equiv) and dioxane (2.0 mL) at room temperature. The mixture was stirred for 7 h at 100°C under nitrogen atmosphere. The resulting mixture was cooled to room temperature and diluted with water (20 mL), extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1:1) to afford 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-[(2-methyl-1,2,4-triazol- 3-yl)oxy]-4H-1,3-benzoxazin-2-one (100 mg, 39%) as a yellow solid. ES-LCMS m/z 390 [M+H]+ Step 2: 3-({3-fluoro-2-[(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-[(2-methyl-1,2,4- triazol-3-yl)oxy]-4H-1,3-benzoxazin-2-one
Figure imgf000133_0001
[00365] Into a 8 mL vial were added 3-[(2-chloro-3-fluoropyridin-4-yl)methyl]-7-[(2-methyl- 1,2,4-triazol-3-yl)oxy]-4H-1,3-benzoxazin-2-one (100 mg, 0.26 mmol, 1.0 equiv), (methylsulfamoyl)amine (34 mg, 0.31 mmol, 1.2 equiv), Pd2(dba)3 (47 mg, 0.05 mmol, 0.2 equiv), t-BuXPhos (43 mg, 0.10 mmol, 0.4 equiv), K3PO4 (163 mg, 0.77 mmol, 3.0 equiv) and t-BuOH (1.0 mL) at room temperature. The mixture was stirred for 2 h at 100°C under nitrogen atmosphere. The resulting mixture was cooled to room temperature and diluted with water (20 mL), extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Welch Ultimate XB- C18, 50*250 mm, 10 μm; Mobile Phase A: 0.1%FA, Mobile Phase B: ACN; Flow rate: 90 mL/min; Gradient: 5% B-3min, 13%B-43%B-12 min.) to afford 3-({3-fluoro-2- [(methylsulfamoyl)amino]pyridin-4-yl}methyl)-7-[(2-methyl-1,2,4-triazol-3-yl)oxy]-4H-1,3- benzoxazin-2-one (12.5 mg, 10%) as a white solid. ES-LCMS m/z 464.2 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 1H), 8.06 (d, J = 5.1 Hz, 1H), 7.70 (s, 1H), 7.32 (d, J = 8.3 Hz, 1H), 7.18 - 7.10 (m, 2H), 7.10 - 7.00 (m, 2H), 4.70 (s, 2H), 4.57 (s, 2H), 3.73 (s, 3H), 2.53 - 2.51 (m, 3H). Synthesis of I-144
Figure imgf000134_0001
Step 1: 3-(benzyloxy)-5-bromopyridine-2-carbonitrile
Figure imgf000134_0002
[00366] To a stirred mixture of NaH (18.95 g, 789.45 mmol, 1.20 equiv) in THF (1.10 L), benzyl alcohol (81.10 g, 749.98 mmol, 1.14 equiv) was added dropwise at 0℃. The mixture was stirred for 1 h at room temperature. To the above mixture, a solution of 5-bromo-3-nitropyridine- 2-carbonitrile (150 g, 657.88 mmol, 1.0 equiv) in THF (1.00 L) was added dropwise over 30 min at 10~25°C. The resulting mixture was stirred for 1 h at room temperature. The reaction was quenched by the addition of ice H2O (2.00 L) at ice bath. The resulting mixture was extracted with EA (3 x 1 L). The combined organic layers were washed with brine (1 L), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by trituration with MTBE (500 mL). This resulted in 3-(benzyloxy)-5-bromopyridine-2- carbonitrile (155.00 g, 81%) as a black-red solid. ES-LCMS m/z 289 [M+H] + Step 2: 3-(benzyloxy)-5-hydroxypyridine-2-carbonitrile
Figure imgf000135_0001
[00367] A mixture of 3-(benzyloxy)-5-bromopyridine-2-carbonitrile (152.00 g, 525.71 mmol, 1.0 equiv), acetohydroxamic acid (118.39 g, 1577.13 mmol, 3.0 equiv) and K2CO3 (365.93 g, 2628.56 mmol, 5.0 equiv) in DMSO (3.00 L) was stirred for 1 h at 80℃. The resulting mixture was cooled to room temperature, diluted with H2O (2.00 L) and acidified to pH=4 with HCl (aq), then extracted with EA(3 x 500 mL). The combined organic layers were washed with brine (500 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by trituration with MTBE (500 mL). This resulted in 3- (benzyloxy)-5-hydroxypyridine-2-carbonitrile (67.90 g, 56%) as a yellow solid. ES-LCMS m/z 227 [M+H] + Step 3: 3-(benzyloxy)-5-[(3-fluoropyridin-2-yl)oxy]pyridine-2-carbonitrile
Figure imgf000135_0002
[00368] To a stirred mixture of 3-(benzyloxy)-5-hydroxypyridine-2-carbonitrile (60.90 g, 269.19 mmol, 1.0 equiv) and CsF (122.67 g, 807.57 mmol, 3.0 equiv) in Pyridine (1.20 L) was added 2,3-difluoropyridine (154.90 g, 1345.94 mmol, 5.0 equiv) dropwise at 100 ℃ over 5 h. The resulting mixture was stirred for additional 5 h at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature, EtOAc (1000 mL) and H2O (1000 L) was added with stirring, after layered, the aqueous layer was extracted with EtOAc (2 x 1000 mL). The combined organic layers were washed with brine (1000 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by trituration with MTBE (500 mL). This resulted in 3-(benzyloxy)-5-[(3-fluoropyridin-2-yl)oxy]pyridine-2- carbonitrile (45.80 g, 52%) as a brown solid. ES-LCMS m/z 322 [M+H]+. Step 4: 1-[3-(benzyloxy)-5-[(3-fluoropyridin-2-yl)oxy]pyridin-2-yl]ethanamine
Figure imgf000136_0001
[00369] To a stirred mixture of 3-(benzyloxy)-5-[(3-fluoropyridin-2-yl)oxy]pyridine-2- carbonitrile (10.00 g, 31.12 mmol, 1.0 equiv) in THF (100 mL) was added bromo(methyl)magnesium (7.42 g, 62.24 mmol, 2.0 equiv) dropwise at -10 ℃ under nitrogen atmosphere. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. The reaction was treatment by the addition of MeOH (100 mL) at 0℃. To the above mixture was added NaBH4 (0.94 g, 24.89 mmol, 0.8 equiv) in portions over 5 min at 0℃. The resulting mixture was stirred for additional 10 min at room temperature. The reaction was quenched by the addition of HCl in MeOH (4M, 50 mL) at 0℃. The resulting mixture was filtered, the filter cake was washed with MeOH (3 x 20 mL). The filtrate was concentrated under reduced pressure. This resulted in 1-[3-(benzyloxy)-5-[(3-fluoropyridin-2-yl)oxy]pyridin-2-yl]ethanamine (7.00 g, 66%) as a white solid.ES-LCMS m/z 340 [M+H] +. Step 5: 2-(1-aminoethyl)-5-((3-fluoropyridin-2-yl)oxy)pyridin-3-ol
Figure imgf000136_0002
[00370] To a solution of 1-[3-(benzyloxy)-5-[(3-fluoropyridin-2-yl)oxy]pyridin-2- yl]ethanamine (7.00 g, 20.63 mmol, 1.0 equiv) in DCE (100 mL) was added BBr3 (20.67 g, 82.504 mmol, 4 equiv) dropwise at -10 ℃. Then stirred the solution at 80°C for 8 h. The resulting mixture was concentrated and treatment with H2O (30 mL) at 0°C. The mixture was basified to pH=7 with Na2CO3(aq), extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product 2-(1-aminoethyl)-5-[(3-fluoropyridin-2-yl)oxy]pyridin-3-ol (9.00 g crude) was used in the next step directly without further purification. ES-LCMS m/z 250 [M+H]+. Step 6: 2-(1-((2-fluoro-3-nitrobenzyl)amino)ethyl)-5-((3-fluoropyridin-2-yl)oxy)pyridin-3-ol
Figure imgf000136_0003
[00371] Into a 40 mL vial were added 2-(1-aminoethyl)-5-[(3-fluoropyridin-2-yl)oxy]pyridin- 3-ol (300 mg, 1.20 mmol, 1.0 equiv), 2-fluoro-3-nitrobenzaldehyde (200 mg, 1.20 mmol, 1.0 equiv), DIEA (466 mg, 3.61 mmol, 3.0 equiv) and MeOH (4.0 mL) at room temperature. Then stirred the solution at the same temperature for 2 h. To this stirred solution was added NaBH4 (45 mg, 1.20 mmol, 1.0 equiv) in portions at 0 °C. After 10 min the reaction was quenched by the addition of H2O (2 mL) at 0°C, the resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with MeOH in DCM (10%) to afford 2-(1-{[(2-fluoro-3-nitrophenyl)methyl]amino}ethyl)-5-[(3-fluoropyridin-2- yl)oxy]pyridin-3-ol (180 mg, 37%) as yellow solid. ES-LCMS m/z 403 [M+H] +. Step 7: 3-(2-fluoro-3-nitrobenzyl)-7-((3-fluoropyridin-2-yl)oxy)-4-methyl-3,4-dihydro-2H- pyrido[2,3-e][1,3]oxazin-2-one
Figure imgf000137_0001
[00372] To a stirred solution of 2-(1-{[(2-fluoro-3-nitrophenyl)methyl]amino}ethyl)-5-[(3- fluoropyridin-2-yl)oxy]pyridin-3-ol (180 mg, 0.44 mmol, 1.0 equiv) and Pyridine (353 mg, 4.47 mmol, 10.0 equiv) in DMF (3.0 mL) was added CDI (87 mg, 0.53 mmol, 1.2 equiv) in portions under N2 atmosphere. Then stirred the reaction mixture at 0°C for 1 h. The reaction was quenched by the addition of H2O (20 mL) at 0°C. The resulting mixture was extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE: EtOAc (1:1) to afford 3-[(2-fluoro-3- nitrophenyl)methyl]-7-[(3-fluoropyridin-2-yl)oxy]-4-methyl-4H-pyrido[2,3-e][1,3]oxazin-2-one (100 mg, 52%) as yellow solid. ES-LCMS m/z 429 [M+H] +. Step 8: 3-(3-amino-2-fluorobenzyl)-7-((3-fluoropyridin-2-yl)oxy)-4-methyl-3,4-dihydro-2H- pyrido[2,3-e][1,3]oxazin-2-one
Figure imgf000137_0002
[00373] Into a 8 mL vial were added 3-[(2-fluoro-3-nitrophenyl)methyl]-7-[(3-fluoropyridin-2- yl)oxy]-4-methyl-4H-pyrido[2,3-e][1,3]oxazin-2-one (100 mg, 0.23 mmol, 1.0 equiv), Fe (130 mg, 2.33 mmol, 10.0 equiv) and CH3COOH (2.0 mL) at room temperature. Then stirred the reaction mixture at 50°C for 2 h. The mixture was allowed to cool down to room temperature and diluted H2O (20 mL), then basified to pH>8 with Na2CO3. The resulting mixture was extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE: EtOAc (1:2) to afford 3-[(3-amino-2-fluorophenyl)methyl]-7-[(3-fluoropyridin-2-yl)oxy]-4-methyl-4H-pyrido[2,3- e][1,3]oxazin-2-one (65 mg, 69%) as a white solid. ES-LCMS m/z 399 [M+H] +. Step 9: 3-({3-[(ethylsulfamoyl)amino]-2-fluorophenyl}methyl)-7-[(3-fluoropyridin-2-yl)oxy]-4- methyl-4H-pyrido[2,3-e][1,3]oxazin-2-one
Figure imgf000138_0001
[00374] To a stirred solution of 3-[(3-amino-2-fluorophenyl)methyl]-7-[(3-fluoropyridin-2- yl)oxy]-4-methyl-4H-pyrido[2,3-e][1,3]oxazin-2-one (60 mg, 0.15 mmol, 1.0 equiv) and pyridine (71 mg, 0.91 mmol, 6.0 equiv) in DMF (2.0 mL) was added N-ethylsulfamoyl chloride (64 mg, 0.45 mmol, 3.0 equiv) dropwise at 0°C under N2 atmosphere. Then stirred the reaction mixture at room temperature for 2 h. The resulting solution was applied to reverse phase flash with the following conditions (column, C18 silica gel; mobile phase, MeCN in Water (0.1% NH3.H2O), 20% to 55% gradient in 15 min; detector, UV 254 nm.) to afford 3-({3-[(ethylsulfamoyl)amino]- 2-fluorophenyl}methyl)-7-[(3-fluoropyridin-2-yl)oxy]-4-methyl-4H-pyrido[2,3-e][1,3]oxazin-2- one (60 mg, 78%) as a white solid. ES-LCMS m/z 506 [M+H] +. Step 10: (4R)-3-({3-[(ethylsulfamoyl)amino]-2-fluorophenyl}methyl)-7-[(3-fluoropyridin-2- yl)oxy]-4-methyl-4H-pyrido[2,3-e][1,3]oxazin-2-one
Figure imgf000138_0002
[00375] The racemate product 3-({3-[(ethylsulfamoyl)amino]-2-fluorophenyl}methyl)-7-[(3- fluoropyridin-2-yl)oxy]-4-methyl-4H-pyrido[2,3-e][1,3]oxazin-2-one (50 mg, 0.09 mmol) was separated by Prep-SFC with the following conditions (Column: CHIRAL ART Cellulose-SC, 3*25 cm, 5 μm; Mobile Phase A: CO2, Mobile Phase B: MeOH: DCM=2: 1(0.1% 2M NH3-MeOH); Flow rate: 80 mL/min; Gradient: isocratic 45% B; Wave Length: 220 nm; RT1(min): 3.7; Sample Solvent: MeOH-HPLC; Injection Volume: 2 mL; Number Of Runs: 5) to afford (4R)-3-({3- [(ethylsulfamoyl)amino]-2-fluorophenyl}methyl)-7-[(3-fluoropyridin-2-yl)oxy]-4-methyl-4H- pyrido[2,3-e][1,3]oxazin-2-one (19.0 mg, 38%) as a white solid. ES-LCMS m/z 506 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.41 (s, 1H), 8.32 (d, J = 2.3 Hz, 1H), 7.98 - 7.86 (m, 2H), 7.66 (d, J = 2.3 Hz, 1H), 7.45 - 7.36 (m, 1H), 7.33 (t, J = 5.6 Hz, 1H), 7.30 - 7.22 (m, 1H), 7.21 - 7.09 (m, 2H), 4.94 (d, J = 15.4 Hz, 1H), 4.67 - 4.49 (m, 2H), 2.95 - 2.84 (m, 2H), 1.50 (d, J = 6.6 Hz, 3H), 0.94 (t, J = 7.2 Hz, 3H). Synthesis of I-143
Figure imgf000139_0001
Step 1: (4S)-3-({3-[(ethylsulfamoyl)amino]-2-fluorophenyl}methyl)-7-[(3-fluoropyridin-2- yl)oxy]-4-methyl-4H-pyrido[2,3-e][1,3]oxazin-2-one
Figure imgf000139_0002
[00376] The racemate product 3-({3-[(ethylsulfamoyl)amino]-2-fluorophenyl}methyl)-7-[(3- fluoropyridin-2-yl)oxy]-4-methyl-4H-pyrido[2,3-e][1,3]oxazin-2-one (50 mg, 0.099 mmol) was separated by Prep-SFC with the following conditions (Column: CHIRAL ART Cellulose-SC, 3*25 cm, 5 μm; Mobile Phase A: CO2, Mobile Phase B: MeOH: DCM=2: 1(0.1% 2M NH3-MeOH); Flow rate: 80 mL/min; Gradient: isocratic 45% B; Wave Length: 220 nm; RT2(min): 4.8; Sample Solvent: MeOH-HPLC; Injection Volume: 2 mL; Number Of Runs: 5) to afford (4S)-3-({3- [(ethylsulfamoyl)amino]-2-fluorophenyl}methyl)-7-[(3-fluoropyridin-2-yl)oxy]-4-methyl-4H- pyrido[2,3-e][1,3]oxazin-2-one (18.8 mg, 38%) as a white solid. ES-LCMS m/z 506.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ 9.41 (s, 1H), 8.32 (d, J = 2.3 Hz, 1H), 7.98 - 7.87 (m, 2H), 7.66 (d, J = 2.3 Hz, 1H), 7.44 - 7.38 (m, 1H), 7.35 - 7.30 (m, 1H), 7.29 - 7.23 (m, 1H), 7.19 - 7.11 (m, 2H), 4.94 (d, J = 15.4 Hz, 1H), 4.66 - 4.50 (m, 2H), 2.95 - 2.84 (m, 2H), 1.50 (d, J = 6.6 Hz, 3H), 0.94 (t, J = 7.2 Hz, 3H). EXAMPLE 2: SPR Assays MEK-RAF binding assay [00377] Compound effects on binding affinity of MEK to BRAF or CRAF were followed by surface plasmon resonance (SPR) with single-cycle kinetic analysis. GST-BRAF or GST-CRAF was immobilized onto a chip and increasing concentrations of His-tagged MEK1 were flowed over the sensor chip. 500 mM ATP was added to the running buffer. The dissociation constant for MEK1 binding to either BRAF or CRAF was calculated in the absence and presence of compound. Instruments were carried out on a Biacore 8K instrument. Sensorgrams were double-reference and DMSO corrected. Data was fit to a single-cycle kinetic model. [00378] Experimental results are provided in Table 2, below. The compound numbers correspond to the compound numbers in Table 1. The activity is classified as A-D, wherein A represents IC50 < 100 nM; B represents 100 nM ≤ IC50 < 1 uM; C represents 1 ≤ IC50 < 10 uM; and D represents IC50 ≥10 uM. TABLE 2.
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
MEK-KSR binding assay [00379] Compound effects on binding affinity of MEK to KSR1 or KSR2 are followed by surface plasmon resonance (SPR) with single-cycle kinetic analysis. GST-KSR1 or GST-KSR2 is immobilized onto a chip and increasing concentrations of His-tagged MEK1 are flowed over the sensor chip. 500 mM ATP is added to the running buffer. The dissociation constant for MEK1 binding to either BRAF or CRAF is calculated in the absence and presence of compound. Instruments are carried out on a Biacore 8K instrument. Sensorgrams are double-reference and DMSO corrected. Data is fit to a single-cycle kinetic model. EXAMPLE 3: Immunoprecipitation Assay [00380] Immunoprecipitation experiments were performed by plating about 450,000 HCT116 cells per well in 6-well plates. Cells were plated for 48 h so as to reach approximately 70% confluency before transfection. Then, 24 h after transfection, cells were treated with vehicle (0.1% DMSO) or a compound (about 200 nM) for 1 h. Cells were washed twice in cold PBS and then transferred to a pre-chilled tube in 0.6 ml of PBS solution. Cells were spun at 1,800g in a cold centrifuge for 10 min, and supernatant was aspirated. To lyse cells, pellets were resuspended in NP-40 buffer (50 mM Tris pH 7.8, 100 mM NaCl, 0.5% (v/v) NP-40, 10% (v/v) glycerol, 1 mM EDTA) supplemented with protease and phosphatase inhibitor cocktail (Thermo Fisher, 78440) and incubated on ice for 30 min. Lysates were centrifuged for 20 min at 2,100g, and supernatants are collected. Cleared lysates were quantified using BCA reagent (Pierce, 23225), with BSA as a standard. Five micrograms of rabbit anti-MEK1 antibody (Millipore-Sigma, 07-641), or rabbit IgG (Millipore Sigma, 12-370), was immobilized on 50 µl of Sepharose Protein A Resin (Thermo Fisher, 53139) and washed three times in 300 μl NP-40 buffer before initiating immunoprecipitations. [00381] Next, for MEK1 immunoprecipitation, 250 μg of total cell lysate in a total volume of 0.6 ml was mixed with the pre-immobilized anti-MEK1 antibody pre-conjugated to Protein A resin. Samples were incubated at 4 °C on an end-over-end rotator for 4 h, followed by three washes in 0.6 ml volume of NP-40 buffer. Next, proteins were denatured and released from resin by the addition of 80 µl volume of 1× SDS sample buffer. Samples were boiled at 90 °C for 2 min, spun, and then applied to a 4–12% bis-tris glycine gel (Bio-Rad, 3450125) ran in MOPS-SDS buffer (Thermo Fisher, NP0001) for 60 min at 150 V. Gels were then transferred onto nitrocellulose in 20% methanol in Tris-glycine buffer (95 V, 250 A). Transfers were confirmed using Ponceau red and then analyzed by western blot. Signals for MEK, Flag-tagged proteins, BRAF, and GAPDH were detected by enhanced chemiluminescence on a ChemDoc XRS+ imaging system (Biorad). [00382] Experimental results are provided in Table 3, below. The compound numbers correspond to the compound numbers in Table 1. The activity is classified as A-D, wherein A represents IC50 < 100 nM; B represents 100 nM ≤ IC50 < 1 uM; C represents 1 ≤ IC50 < 10 uM; and D represents IC50 ≥10 uM. TABLE 3.
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
4 hour – pERK1/2 HTRF Assay [00383] AsPC1 cells were seeded at 20,000 cells/well overnight in 96-well plates, then treated with a 12-point dose response of compounds (Starting dose of 3000 nM and 3-fold dilution) for 4 hours. Phosphorylation of ERK1/2 on residues Thr202 and Tyr204 were detected using a Homogenous Time-Resolved Fluorescence (HTRF) assay (Cisbio Cat# 64ERKPEG). Briefly, ERK phosphorylation was detected using a sandwich assay format using two phospho-ERK specific antibodies, one labelled with Eu3+ - Cryptate (donor) and the second with d2 (acceptor). The Fluorescence Resonance Energy Transfer (FRET) towards the d2 acceptor was detected at a wavelength of 665 nm. ERK1/2 phosphorylation levels were normalized to total ERK1/2 protein levels in each well. IC50 values were determined by fitting a variable slope, four parameters curve to the compound concentration to normalized Phospho-ERK1/2 relationship. The activity is classified as A-D, wherein A represents IC50 < 100 nM; B represents 100 nM ≤ IC50 < 1 uM; C represents 1 ≤ IC50 < 10 uM; and D represents IC50 ≥10 uM. Table 4.
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
4 hour – pMEK1/2 MSD Assay [00384] AsPC1 cells were seeded at 10,000 cells/ well overnight in 96-well plates, then treated with a 12-point dose response of compounds (Starting dose of 1000 nM and 3-fold dilution) for 4 hours. Phosphorylation of MEK1/2 on residues Ser217 and Ser221 were detected using a MesoScale Discovery (MSD) assay (MSD Cat# K15129D-2). MEK1/2 phosphorylation levels were normalized to total MEK1/2 protein detected in each well on the same MSD plate. IC50 values were determined by fitting a variable slope, four parameters curve to the compound concentration to normalized Phospho-MEK1/2 relationship. The activity is classified as A-D, wherein A represents IC50 < 100 nM; B represents 100 nM ≤ IC50 < 1 uM; C represents 1 ≤ IC50 < 10 uM; and D represents IC50 ≥10 uM. Table 5.
Figure imgf000153_0001
EXAMPLE 4: Biological Assay for Inhibiting Mouse Xenograft Tumor Growth [00385] Exemplary compounds having MEK inhibitory activity can be tested, alone or in combination with a second anti-cancer agent, such as a TEAD inhibitor or an ERK5 inhibitor, for ability to inhibit tumor growth in patient-derived mouse xenograft models of pancreatic cancer and lung cancer. Assay procedures are described below. Procedures for Patient-Derived Mouse Xenograft (PDX) Tumor Growth Assay [00386] Balb/c mice (6-8 weeks old) are inoculated subcutaneously in the right flank with a primary human tumor xenograft model tumor fragment (2-3 mm3 in diameter) for tumor development. In one study, mice are inoculated with a human lung adenocarcinoma tumor model (MSCLC, ADC model LU6424) that harbors a BRAF mutation (LU6424); in a second study, mice are inoculated with a human pancreatic tumor model (adenosquamous carcinoma model PA6258) with a K-Ras G12D mutation (PA6258). When mean tumor volume reaches approximately 150- 200 mm3, animals are randomly allocated to appropriate treatment groups. Mice are treated with one of the following: (1) Vehicle control, (2) Exemplary MEK inhibitor alone, (3) Second anti- cancer agent alone, or (4) Combination of MEK inhibitor and second anti-cancer agent. Tumors are measured twice per week using calipers. EXAMPLE 5: Mouse Pharmacokinetic Studies [00387] Exemplary compounds were tested to determine their pharmacokinetic properties in mice using cassette dosing protocols. Assay procedures and results are described below. Part I – Procedures for Mouse Casette Pharmacokinetics Studies [00388] Pharmacokinetics studies are conducted using cassette dosing protocols, with test compounds administered per mouse. Treatment groups are 3 mice (~6-8 week old, ~20-30g balb/c male mice) per route of administration. Compounds are formulated in 5% DMSO and 95% PEG400 with final concentrations of 0.5 mg/mL for IV and PO dosing. Compounds are dosed at 2 mg/kg for IV dosing and 5 mg/kg for PO dosing. Blood samples are withdrawn at 0.083, 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, and 24 hours following IV dosing, and 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, and 24 hours following PO dosing. Blood samples are centrifuged at 4,000 g for 5 minutes at 4 °C to obtain plasma samples. Plasma samples are diluted with acetonitrile containing internal standard, vortexed for 30 seconds, then centrifuged at 4,000 rpm for 15 minutes at 4 °C. The supernatant is diluted with water, and compound concentration is determined by HPLC/MS/MS compared to calibration standards of 0.5-1,000 ng/mL. EXAMPLE 6: In vivo efficacy studies using human MIA PaCa-2 xenografts in mice [00389] The antitumor activity of MEKi was evaluated in vivo using a human cell line derived xenografts grown from MIA PaCa-2 cell line in immunodeficient mice. The tumor cell lines are maintained in vitro as monolayer culture in medium at 37°C in an atmosphere of 5% CO2 in air. The tumor cells are routinely sub-cultured before confluence by trypsin-EDTA treatment, not to exceed 4-5 passages. The cells growing in an exponential growth phase are harvested for tumor inoculation. MIA PaCa-2 tumor cells (1 x 106 cells) were implanted into Nu/Nu nude mice subcutaneously on the right flank with tumor cells in a 1:1 mixture with matrigel. Tumors are allowed to grow to approximately 250 mm3. At this time, mice are assigned to groups such that the mean tumor volume is the same for each treatment group (n = 8 mice/group). Compounds were administrated to the tumor-bearing mice via QD oral gavage at a dose of 1 mg/kg. After 10 days of dosing, when tumors in the vehicle control mice reached the ethical endpoint (mean tumor volume > 1500 mm3). [00390] While a number of embodiments of this disclosure are described, it is apparent that the basic examples may be altered to provide other embodiments that utilize the compounds and methods of this disclosure. Therefore, it will be appreciated that the scope of this disclosure is to be defined by the application and appended claims rather than by the specific embodiments that have been represented by way of example. INCORPORATION BY REFERENCE [00391] The entire disclosure of each of the patent documents and scientific articles referred to herein is incorporated by reference for all purposes. EQUIVALENTS [00392] The disclosure may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting the disclosure described herein. Scope of the disclosure is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.

Claims

CLAIMS WHAT IS CLAIMED IS: 1. A compound of Formula (I):
Figure imgf000156_0001
or a pharmaceutically acceptable salt thereof, wherein: Ring A and Ring B are independently an optionally substituted ring selected from a phenyl ring, a 5-6 membered heteroaromatic ring having 1-4 heteroatoms independently selected from N, O, or S, or a 8-11 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from N, O, or S; X1 is C(R21) or N; X2 is C(R22) or N; X3 is C(R23) or N; R3 and R4 are independently halogen, R, or -C(O)-N(R)2; or R3 and R4, taken together with the atom to which they attach, form C=O, or an optionally substituted 3-8 membered carbocyclic ring, or an optionally substituted 3-8 membered heterocyclic ring having 1-3 heteroatoms independently selected from N, O, or S; each of R21, R22, and R23 is independently halogen, R, -OR, -N(R)2, -N(R)-C(O)-R, or - C(O)-N(R)2; L1 is -C(R5)(R6)- or -NR-; each of R5 and R6 is independently halogen, R, or R5 and R6, together with the atom to which they attach, form an optionally substituted 3-8 membered carbocyclic ring or an optionally substituted 3-8 membered heterocyclic ring having 1-3 heteroatoms independently selected from N, O, or S; and each R is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S.
2. A compound of Formula (II):
Figure imgf000157_0001
or a pharmaceutically acceptable salt thereof, wherein: W is S or S(O)2; Ring A and Ring B are independently an optionally substituted ring selected from a phenyl ring, a 5-6 membered heteroaromatic ring having 1-4 heteroatoms independently selected from N, O, or S, or a 8-11 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from N, O, or S; X1 is C(R21) or N; X2 is C(R22) or N; X3 is C(R23) or N; R3 and R4 are independently halogen, R, or -C(O)-N(R)2; or R3 and R4, taken together with the atom to which they attach, form C=O, or an optionally substituted 3-8 membered carbocyclic ring, or an optionally substituted 3-8 membered heterocyclic ring having 1-3 heteroatoms independently selected from N, O, or S; each of R21, R22, and R23 is independently halogen, R, -OR, -N(R)2, -N(R)-C(O)-R, or - C(O)-N(R)2; L1 is -C(R5)(R6)- or -NR-; each of R5 and R6 is independently halogen, R, or R5 and R6, together with the atom to which they attach, form an optionally substituted 3-8 membered carbocyclic ring or an optionally substituted 3-8 membered heterocyclic ring having 1-3 heteroatoms independently selected from N, O, or S; and each R is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S.
3. The compound of claim 1 or 2, wherein Ring B is an optionally substituted ring selected from a phenyl ring or a 6 membered heteroaromatic ring having 1-4 N atoms.
4. The compound of any one of claims 1-3, wherein Ring B is or
Figure imgf000158_0001
Figure imgf000158_0002
, and R1 is halogen, R, -OR, or -N(R)2.
5. The compound of claim 4, wherein R1 is hydrogen or halogen.
6. The compound of claim 5, wherein R1 is fluoro.
7. The compound of any one of claims 1 to 6, wherein R21 is hydrogen.
8. The compound of any one of claims 1 to 7, wherein R22 is hydrogen or halogen.
9. The compound of any one of claims 1 to 8, wherein R23 is hydrogen or halogen.
10. The compound of any one of claims 1 to 9, wherein R3 and R4 are independently hydrogen or optionally substituted C1-C6 aliphatic, or R3 and R4, taken together with the atom to which they attach, form C(=O).
11. The compound of claim 10, wherein R3 and R4 are independently C1-C6 aliphatic.
12. The compound of claim 10, wherein R3 is hydrogen, and R4 is -CH3.
13. The compound of any one of claims 1 to 12, wherein each of R5 and R6 is independently halogen or R.
14. The compound of claim 13, wherein each of R5 and R6 is independently halogen, hydrogen, or C1-C6 aliphatic.
15. The compound of any one of claims 1 to 14, wherein Ring A is an optionally substituted ring selected from a phenyl ring or a 6 membered heteroaromatic ring having 1-4 N atoms.
16. The compound of any one of claims 1 to 15, wherein Ring A is
Figure imgf000159_0001
or R7 is independently halogen, R, -OR, or -N(R)2 8
Figure imgf000159_0002
; and R is halogen, R, -OR, -N(R)2,
Figure imgf000159_0003
, or
Figure imgf000159_0004
; each of R10 and R11 is independently H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S, optionally substituted phenyl, or optionally substituted 5-6 membered heteroaryl having 1-4 heteroatoms independently selected from N, O, or S, or R10 and R11 taken together with the N atom to which they attach, form optionally substituted 3-8 membered heterocyclyl having 0, 1, or 2 heteroatoms independently selected from N, O, or S in addition to the N atom to which R10 and R11 attach; and R12 is H, optionally substituted C1-C6 aliphatic, optionally substituted 3-8 membered carbocyclyl, or optionally substituted 3-8 membered heterocyclyl having 1, 2, or 3 heteroatoms independently selected from N, O, or S.
17. The compound of claim 16, wherein each R is independently H or optionally substituted C1-C6 aliphatic.
18. The compound of claim 16, wherein R7 is independently H, halogen, or C1-C6 aliphatic.
19. The compound of claim 18, wherein R7 is independently H or fluoro.
20. The compound of any one of claims 16 to 19, wherein R8 is ,
Figure imgf000160_0001
21. The compound of any one of claims 16 to 20, wherein R8 is
Figure imgf000160_0003
each 10 11
Figure imgf000160_0002
of R and R is independently H, C1-C6 aliphatic, or 3-8 membered heterocyclyl having 1, 2, or 3 N atoms; and R12 is H, optionally substituted C1-C6 aliphatic, or optionally substituted 3-8 membered carbocyclyl.
22. The compound of claim 20 or 21, wherein R10 and R11 are independently hydrogen or C1- C6 alkyl.
23. The compound of any one of claims 18 to 22, wherein R12 is hydrogen or C1-C6 alkyl.
24. The compound of any one of claims 1 to 23, wherein the compound is a compound selected from those listed in Table 1, or a pharmaceutically acceptable salt thereof.
25. A pharmaceutical composition comprising the compound of any one of claims 1-24, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
26. A method of treating a disorder mediated by MEK, comprising administering a therapeutically effective amount of a compound of any one of claims 1-24, or the pharmaceutical composition of claim 25, to a subject in need thereof to treat the disorder.
27. The method of claim 26, wherein the disorder is cancer.
28. A method for treating a cancer in a patient, comprising administering the compound of any one of claims 1-24, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of claim 25, to the patient in need thereof to treat the cancer.
29. A method of inhibiting MEK activity, comprising contacting MEK or a KSR-MEK complex or a RAF-MEK complex with an effective amount of a compound of any one of claims 1-24, or the pharmaceutical composition of claim 25, to inhibit MEK activity.
PCT/US2023/064091 2022-03-10 2023-03-10 Mek inhibitors and uses thereof WO2023173057A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US202263269140P 2022-03-10 2022-03-10
US63/269,140 2022-03-10
US202263375864P 2022-09-16 2022-09-16
US63/375,864 2022-09-16
US202363479118P 2023-01-09 2023-01-09
US63/479,118 2023-01-09

Publications (1)

Publication Number Publication Date
WO2023173057A1 true WO2023173057A1 (en) 2023-09-14

Family

ID=85873845

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/064091 WO2023173057A1 (en) 2022-03-10 2023-03-10 Mek inhibitors and uses thereof

Country Status (1)

Country Link
WO (1) WO2023173057A1 (en)

Citations (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR901228A (en) 1943-01-16 1945-07-20 Deutsche Edelstahlwerke Ag Ring gap magnet system
JPS60214358A (en) * 1984-04-11 1985-10-26 Fuji Photo Film Co Ltd Silver halide color photosensitive material
WO2001042246A2 (en) 1999-12-10 2001-06-14 Pfizer Products Inc. PYRROLO[2,3-d]PYRIMIDINE COMPOUNDS
WO2002088112A1 (en) 2001-04-27 2002-11-07 Zenyaku Kogyo Kabushiki Kaisha Heterocyclic compound and antitumor agent containing the same as active ingredient
US6552065B2 (en) 2000-09-01 2003-04-22 Novartis Ag Deacetylase inhibitors
WO2003063794A2 (en) 2002-02-01 2003-08-07 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds and their uses
WO2004019973A1 (en) 2002-08-14 2004-03-11 Atugen Ag Use of protein kinase n beta
WO2004089925A1 (en) 2003-04-03 2004-10-21 Semafore Pharmaceuticals, Inc. Pi-3 kinase inhibitor prodrugs
WO2004106328A1 (en) 2003-05-30 2004-12-09 Gemin X Biotechnologies Inc. Triheterocyclic compounds, compositions, and methods for treating cancer or viral diseases
WO2005007623A2 (en) 2003-07-03 2005-01-27 The Trustees Of The University Of Pennsylvania Inhibition of syk kinase expression
WO2005113554A2 (en) 2004-05-13 2005-12-01 Icos Corporation Method of preparing 3-phenyl-2-[9h-purin-6-ylamino)-methyl]-3h-quinazolin-4-one and substituted and related compounds
WO2006029879A2 (en) 2004-09-17 2006-03-23 F.Hoffmann-La Roche Ag Anti-ox40l antibodies
WO2006078846A1 (en) 2005-01-19 2006-07-27 Rigel Pharmaceuticals, Inc. Prodrugs of 2,4-pyrimidinediamine compounds and their uses
WO2006105021A2 (en) 2005-03-25 2006-10-05 Tolerrx, Inc. Gitr binding molecules and uses therefor
WO2006122806A2 (en) 2005-05-20 2006-11-23 Novartis Ag 1,3-dihydro-imidazo [4,5-c] quinolin-2-ones as lipid kinase inhibitors
WO2007005874A2 (en) 2005-07-01 2007-01-11 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
WO2007016176A2 (en) 2005-07-28 2007-02-08 Phoenix Biotechnology Inc. Scf extract containing cardiac glycoside
WO2007044729A2 (en) 2005-10-07 2007-04-19 Exelixis, Inc. N- (3-amino-quinoxalin-2-yl) -sulfonamide derivatives and their use as phosphatidylinositol 3-kinase inhibitors
WO2007053452A1 (en) 2005-11-01 2007-05-10 Targegen, Inc. Bi-aryl meta-pyrimidine inhibitors of kinases
WO2007070514A1 (en) 2005-12-13 2007-06-21 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as janus kinase inhibitors
WO2007084786A1 (en) 2006-01-20 2007-07-26 Novartis Ag Pyrimidine derivatives used as pi-3 kinase inhibitors
WO2007129161A2 (en) 2006-04-26 2007-11-15 F. Hoffmann-La Roche Ag Thieno [3, 2-d] pyrimidine derivative useful as pi3k inhibitor
WO2008039218A2 (en) 2006-09-22 2008-04-03 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US7390799B2 (en) 2005-05-12 2008-06-24 Abbott Laboratories Apoptosis promoters
WO2008109943A1 (en) 2007-03-12 2008-09-18 Cytopia Research Pty Ltd Phenyl amino pyrimidine compounds and uses thereof
WO2008118802A1 (en) 2007-03-23 2008-10-02 Regents Of The University Of Minnesota Therapeutic compounds
EP1982982A1 (en) * 2006-02-09 2008-10-22 Chugai Seiyaku Kabushiki Kaisha Novel coumarin derivative having antitumor activity
WO2008132601A1 (en) 2007-04-30 2008-11-06 Immutep Cytotoxic anti-lag-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
WO2009009116A2 (en) 2007-07-12 2009-01-15 Tolerx, Inc. Combination therapies employing gitr binding molecules
WO2009044273A2 (en) 2007-10-05 2009-04-09 Immutep Use of recombinant lag-3 or the derivatives thereof for eliciting monocyte immune response
WO2009073620A2 (en) 2007-11-30 2009-06-11 Newlink Genetics Ido inhibitors
WO2009114512A1 (en) 2008-03-11 2009-09-17 Incyte Corporation Azetidine and cyclobutane derivatives as jak inhibitors
WO2009156652A1 (en) 2008-05-29 2009-12-30 Saint-Gobain Centre De Recherches Et D'etudes Europeen Cellular structure containing aluminium titanate
WO2010019570A2 (en) 2008-08-11 2010-02-18 Medarex, Inc. Human antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
WO2010077634A1 (en) 2008-12-09 2010-07-08 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
WO2011028683A1 (en) 2009-09-03 2011-03-10 Schering Corporation Anti-gitr antibodies
WO2011056652A1 (en) 2009-10-28 2011-05-12 Newlink Genetics Imidazole derivatives as ido inhibitors
WO2011070024A1 (en) 2009-12-10 2011-06-16 F. Hoffmann-La Roche Ag Antibodies binding preferentially human csf1r extracellular domain 4 and their use
WO2011090760A1 (en) 2009-12-29 2011-07-28 Avila Therapeutics, Inc. Heteroaryl compounds and uses thereof
WO2011107553A1 (en) 2010-03-05 2011-09-09 F. Hoffmann-La Roche Ag Antibodies against human csf-1r and uses thereof
WO2011131407A1 (en) 2010-03-05 2011-10-27 F. Hoffmann-La Roche Ag Antibodies against human csf-1r and uses thereof
WO2011140249A2 (en) 2010-05-04 2011-11-10 Five Prime Therapeutics, Inc. Antibodies that bind csf1r
WO2012032433A1 (en) 2010-09-09 2012-03-15 Pfizer Inc. 4-1bb binding molecules
US8138347B2 (en) 2007-05-18 2012-03-20 Glaxosmithkline Llc Quinoline derivatives as PI3 kinase inhibitors
WO2012142237A1 (en) 2011-04-15 2012-10-18 Newlink Geneticks Corporation Fused imidazole derivatives useful as ido inhibitors
WO2012145493A1 (en) 2011-04-20 2012-10-26 Amplimmune, Inc. Antibodies and other molecules that bind b7-h1 and pd-1
WO2013079174A1 (en) 2011-11-28 2013-06-06 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
WO2013087699A1 (en) 2011-12-15 2013-06-20 F. Hoffmann-La Roche Ag Antibodies against human csf-1r and uses thereof
WO2013119716A1 (en) 2012-02-06 2013-08-15 Genentech, Inc. Compositions and methods for using csf1r inhibitors
WO2013132044A1 (en) 2012-03-08 2013-09-12 F. Hoffmann-La Roche Ag Combination therapy of antibodies against human csf-1r and uses thereof
WO2013169264A1 (en) 2012-05-11 2013-11-14 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
WO2014008218A1 (en) 2012-07-02 2014-01-09 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
WO2014036357A1 (en) 2012-08-31 2014-03-06 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
US8906682B2 (en) 2010-12-09 2014-12-09 The Trustees Of The University Of Pennsylvania Methods for treatment of cancer
WO2017053706A1 (en) 2015-09-23 2017-03-30 The General Hospital Corporation Tead transcription factor autopalmitoylation inhibitors
WO2017111076A1 (en) 2015-12-24 2017-06-29 協和発酵キリン株式会社 α, β UNSATURATED AMIDE COMPOUND
WO2018204532A1 (en) 2017-05-03 2018-11-08 Vivace Therapeutics, Inc. Non-fused tricyclic compounds
WO2018235926A1 (en) 2017-06-23 2018-12-27 協和発酵キリン株式会社 α, β-UNSATURATED AMIDE COMPOUND
WO2019040380A1 (en) 2017-08-21 2019-02-28 Vivace Therapeutics, Inc. Benzosulfonyl compounds
WO2019113236A1 (en) 2017-12-06 2019-06-13 Vivace Therapeutics, Inc. Benzocarbonyl compounds
WO2019222431A1 (en) 2018-05-16 2019-11-21 Vivace Therapeutics, Inc. Oxadiazole compounds
WO2019232216A1 (en) 2018-05-31 2019-12-05 Genentech, Inc. Therapeutic compounds
US20200035098A1 (en) 2018-07-24 2020-01-30 May Mobility, Inc. Systems and methods for implementing multimodal safety operations with an autonomous agent
WO2020051099A1 (en) 2018-09-03 2020-03-12 Genentech, Inc. Carboxamide and sulfonamide derivatives useful as tead modulators
WO2020081572A1 (en) 2018-10-15 2020-04-23 Dana-Farber Cancer Institute, Inc. Transcriptional enhanced associate domain (tead) transcription factor inhibitors and uses thereof
WO2020097389A1 (en) 2018-11-09 2020-05-14 Vivace Therapeutics, Inc. Bicyclic compounds
WO2020190774A1 (en) 2019-03-15 2020-09-24 The General Hospital Corporation Novel small molecule inhibitors of tead transcription factors
WO2020214734A1 (en) 2019-04-16 2020-10-22 Vivace Therapeutics, Inc. Bicyclic compounds
WO2020243423A1 (en) 2019-05-31 2020-12-03 Ikena Oncology, Inc. Tead inhibitors and uses thereof
WO2020243415A2 (en) 2019-05-31 2020-12-03 Ikena Oncology, Inc. Tead inhibitors and uses thereof
WO2022051565A1 (en) 2020-09-04 2022-03-10 Ikena Oncology, Inc. Substituted 4-piperidinyl-quinazolines, 4-piperidinyl-pyrimidine-2-amines, and related compounds and their use in treating medical conditions
WO2022051569A1 (en) 2020-09-04 2022-03-10 Ikena Oncology, Inc. Substituted 3-piperidinyl-pyrrolo[2,3-b]pyridines and related compounds and their use in treating medical conditions
WO2022051567A1 (en) 2020-09-04 2022-03-10 Ikena Oncology, Inc. Substituted pyrido[2,3-b]pyrazinones and reuated compounds and their use in treating medicau conditions
WO2022051568A1 (en) 2020-09-04 2022-03-10 Ikena Oncology, Inc. Substituted 4-piperidinyl-pyrrolo[2,3-b]pyridines and related compounds and their use in treating medical conditions
WO2022120354A1 (en) 2020-12-02 2022-06-09 Ikena Oncology, Inc. Tead inhibitors and uses thereof
WO2022120353A1 (en) 2020-12-02 2022-06-09 Ikena Oncology, Inc. Tead inhibitors and uses thereof
WO2022120355A1 (en) 2020-12-02 2022-06-09 Ikena Oncology, Inc. Tead degraders and uses thereof

Patent Citations (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR901228A (en) 1943-01-16 1945-07-20 Deutsche Edelstahlwerke Ag Ring gap magnet system
JPS60214358A (en) * 1984-04-11 1985-10-26 Fuji Photo Film Co Ltd Silver halide color photosensitive material
WO2001042246A2 (en) 1999-12-10 2001-06-14 Pfizer Products Inc. PYRROLO[2,3-d]PYRIMIDINE COMPOUNDS
US6552065B2 (en) 2000-09-01 2003-04-22 Novartis Ag Deacetylase inhibitors
WO2002088112A1 (en) 2001-04-27 2002-11-07 Zenyaku Kogyo Kabushiki Kaisha Heterocyclic compound and antitumor agent containing the same as active ingredient
WO2003063794A2 (en) 2002-02-01 2003-08-07 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds and their uses
WO2004019973A1 (en) 2002-08-14 2004-03-11 Atugen Ag Use of protein kinase n beta
WO2004089925A1 (en) 2003-04-03 2004-10-21 Semafore Pharmaceuticals, Inc. Pi-3 kinase inhibitor prodrugs
WO2004106328A1 (en) 2003-05-30 2004-12-09 Gemin X Biotechnologies Inc. Triheterocyclic compounds, compositions, and methods for treating cancer or viral diseases
WO2005007623A2 (en) 2003-07-03 2005-01-27 The Trustees Of The University Of Pennsylvania Inhibition of syk kinase expression
WO2005113554A2 (en) 2004-05-13 2005-12-01 Icos Corporation Method of preparing 3-phenyl-2-[9h-purin-6-ylamino)-methyl]-3h-quinazolin-4-one and substituted and related compounds
WO2006029879A2 (en) 2004-09-17 2006-03-23 F.Hoffmann-La Roche Ag Anti-ox40l antibodies
WO2006078846A1 (en) 2005-01-19 2006-07-27 Rigel Pharmaceuticals, Inc. Prodrugs of 2,4-pyrimidinediamine compounds and their uses
WO2006105021A2 (en) 2005-03-25 2006-10-05 Tolerrx, Inc. Gitr binding molecules and uses therefor
US7390799B2 (en) 2005-05-12 2008-06-24 Abbott Laboratories Apoptosis promoters
WO2006122806A2 (en) 2005-05-20 2006-11-23 Novartis Ag 1,3-dihydro-imidazo [4,5-c] quinolin-2-ones as lipid kinase inhibitors
WO2007005874A2 (en) 2005-07-01 2007-01-11 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
WO2007016176A2 (en) 2005-07-28 2007-02-08 Phoenix Biotechnology Inc. Scf extract containing cardiac glycoside
WO2007044729A2 (en) 2005-10-07 2007-04-19 Exelixis, Inc. N- (3-amino-quinoxalin-2-yl) -sulfonamide derivatives and their use as phosphatidylinositol 3-kinase inhibitors
WO2007053452A1 (en) 2005-11-01 2007-05-10 Targegen, Inc. Bi-aryl meta-pyrimidine inhibitors of kinases
WO2007070514A1 (en) 2005-12-13 2007-06-21 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as janus kinase inhibitors
WO2007084786A1 (en) 2006-01-20 2007-07-26 Novartis Ag Pyrimidine derivatives used as pi-3 kinase inhibitors
EP1982982A1 (en) * 2006-02-09 2008-10-22 Chugai Seiyaku Kabushiki Kaisha Novel coumarin derivative having antitumor activity
WO2007129161A2 (en) 2006-04-26 2007-11-15 F. Hoffmann-La Roche Ag Thieno [3, 2-d] pyrimidine derivative useful as pi3k inhibitor
WO2008039218A2 (en) 2006-09-22 2008-04-03 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
WO2008109943A1 (en) 2007-03-12 2008-09-18 Cytopia Research Pty Ltd Phenyl amino pyrimidine compounds and uses thereof
WO2008118802A1 (en) 2007-03-23 2008-10-02 Regents Of The University Of Minnesota Therapeutic compounds
WO2008132601A1 (en) 2007-04-30 2008-11-06 Immutep Cytotoxic anti-lag-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
US8138347B2 (en) 2007-05-18 2012-03-20 Glaxosmithkline Llc Quinoline derivatives as PI3 kinase inhibitors
WO2009009116A2 (en) 2007-07-12 2009-01-15 Tolerx, Inc. Combination therapies employing gitr binding molecules
WO2009044273A2 (en) 2007-10-05 2009-04-09 Immutep Use of recombinant lag-3 or the derivatives thereof for eliciting monocyte immune response
WO2009073620A2 (en) 2007-11-30 2009-06-11 Newlink Genetics Ido inhibitors
WO2009114512A1 (en) 2008-03-11 2009-09-17 Incyte Corporation Azetidine and cyclobutane derivatives as jak inhibitors
WO2009156652A1 (en) 2008-05-29 2009-12-30 Saint-Gobain Centre De Recherches Et D'etudes Europeen Cellular structure containing aluminium titanate
WO2010019570A2 (en) 2008-08-11 2010-02-18 Medarex, Inc. Human antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
WO2010077634A1 (en) 2008-12-09 2010-07-08 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
WO2011028683A1 (en) 2009-09-03 2011-03-10 Schering Corporation Anti-gitr antibodies
WO2011056652A1 (en) 2009-10-28 2011-05-12 Newlink Genetics Imidazole derivatives as ido inhibitors
WO2011070024A1 (en) 2009-12-10 2011-06-16 F. Hoffmann-La Roche Ag Antibodies binding preferentially human csf1r extracellular domain 4 and their use
WO2011090760A1 (en) 2009-12-29 2011-07-28 Avila Therapeutics, Inc. Heteroaryl compounds and uses thereof
WO2011107553A1 (en) 2010-03-05 2011-09-09 F. Hoffmann-La Roche Ag Antibodies against human csf-1r and uses thereof
WO2011131407A1 (en) 2010-03-05 2011-10-27 F. Hoffmann-La Roche Ag Antibodies against human csf-1r and uses thereof
WO2011140249A2 (en) 2010-05-04 2011-11-10 Five Prime Therapeutics, Inc. Antibodies that bind csf1r
WO2012032433A1 (en) 2010-09-09 2012-03-15 Pfizer Inc. 4-1bb binding molecules
US8906682B2 (en) 2010-12-09 2014-12-09 The Trustees Of The University Of Pennsylvania Methods for treatment of cancer
WO2012142237A1 (en) 2011-04-15 2012-10-18 Newlink Geneticks Corporation Fused imidazole derivatives useful as ido inhibitors
WO2012145493A1 (en) 2011-04-20 2012-10-26 Amplimmune, Inc. Antibodies and other molecules that bind b7-h1 and pd-1
WO2013079174A1 (en) 2011-11-28 2013-06-06 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
WO2013087699A1 (en) 2011-12-15 2013-06-20 F. Hoffmann-La Roche Ag Antibodies against human csf-1r and uses thereof
WO2013119716A1 (en) 2012-02-06 2013-08-15 Genentech, Inc. Compositions and methods for using csf1r inhibitors
WO2013132044A1 (en) 2012-03-08 2013-09-12 F. Hoffmann-La Roche Ag Combination therapy of antibodies against human csf-1r and uses thereof
WO2013169264A1 (en) 2012-05-11 2013-11-14 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
WO2014008218A1 (en) 2012-07-02 2014-01-09 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
WO2014036357A1 (en) 2012-08-31 2014-03-06 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
WO2017053706A1 (en) 2015-09-23 2017-03-30 The General Hospital Corporation Tead transcription factor autopalmitoylation inhibitors
US20190010136A1 (en) 2015-12-24 2019-01-10 Kyowa Hakko Kirin Co., Ltd. Alpha,beta-unsaturated amide compound
WO2017111076A1 (en) 2015-12-24 2017-06-29 協和発酵キリン株式会社 α, β UNSATURATED AMIDE COMPOUND
WO2018204532A1 (en) 2017-05-03 2018-11-08 Vivace Therapeutics, Inc. Non-fused tricyclic compounds
WO2018235926A1 (en) 2017-06-23 2018-12-27 協和発酵キリン株式会社 α, β-UNSATURATED AMIDE COMPOUND
WO2019040380A1 (en) 2017-08-21 2019-02-28 Vivace Therapeutics, Inc. Benzosulfonyl compounds
WO2019113236A1 (en) 2017-12-06 2019-06-13 Vivace Therapeutics, Inc. Benzocarbonyl compounds
WO2019222431A1 (en) 2018-05-16 2019-11-21 Vivace Therapeutics, Inc. Oxadiazole compounds
WO2019232216A1 (en) 2018-05-31 2019-12-05 Genentech, Inc. Therapeutic compounds
US20200035098A1 (en) 2018-07-24 2020-01-30 May Mobility, Inc. Systems and methods for implementing multimodal safety operations with an autonomous agent
WO2020051099A1 (en) 2018-09-03 2020-03-12 Genentech, Inc. Carboxamide and sulfonamide derivatives useful as tead modulators
WO2020081572A1 (en) 2018-10-15 2020-04-23 Dana-Farber Cancer Institute, Inc. Transcriptional enhanced associate domain (tead) transcription factor inhibitors and uses thereof
WO2020097389A1 (en) 2018-11-09 2020-05-14 Vivace Therapeutics, Inc. Bicyclic compounds
WO2020190774A1 (en) 2019-03-15 2020-09-24 The General Hospital Corporation Novel small molecule inhibitors of tead transcription factors
WO2020214734A1 (en) 2019-04-16 2020-10-22 Vivace Therapeutics, Inc. Bicyclic compounds
WO2020243423A1 (en) 2019-05-31 2020-12-03 Ikena Oncology, Inc. Tead inhibitors and uses thereof
WO2020243415A2 (en) 2019-05-31 2020-12-03 Ikena Oncology, Inc. Tead inhibitors and uses thereof
US11274082B2 (en) 2019-05-31 2022-03-15 Ikena Oncology, Inc. Tead inhibitors and uses thereof
WO2022051565A1 (en) 2020-09-04 2022-03-10 Ikena Oncology, Inc. Substituted 4-piperidinyl-quinazolines, 4-piperidinyl-pyrimidine-2-amines, and related compounds and their use in treating medical conditions
WO2022051569A1 (en) 2020-09-04 2022-03-10 Ikena Oncology, Inc. Substituted 3-piperidinyl-pyrrolo[2,3-b]pyridines and related compounds and their use in treating medical conditions
WO2022051567A1 (en) 2020-09-04 2022-03-10 Ikena Oncology, Inc. Substituted pyrido[2,3-b]pyrazinones and reuated compounds and their use in treating medicau conditions
WO2022051568A1 (en) 2020-09-04 2022-03-10 Ikena Oncology, Inc. Substituted 4-piperidinyl-pyrrolo[2,3-b]pyridines and related compounds and their use in treating medical conditions
WO2022120354A1 (en) 2020-12-02 2022-06-09 Ikena Oncology, Inc. Tead inhibitors and uses thereof
WO2022120353A1 (en) 2020-12-02 2022-06-09 Ikena Oncology, Inc. Tead inhibitors and uses thereof
WO2022120355A1 (en) 2020-12-02 2022-06-09 Ikena Oncology, Inc. Tead degraders and uses thereof

Non-Patent Citations (16)

* Cited by examiner, † Cited by third party
Title
"March's Advanced Organic Chemistry", 2001, JOHN WILEY & SONS
BUM-ERDENE ET AL.: "Small-Molecule Covalent Modification of Conserved Cysteine Leads to Allosteric Inhibition of the TEADoYap Protein-Protein Interaction", CELL CHEMICAL BIOLOGY, vol. 26, 2019, pages 1 - 12
CONNOLLY ET AL., INT'L J. BIOLOGICAL SCIENCES, vol. 8, 2012, pages 964 - 978
GIBAULT ET AL.: "Targeting Transcriptional Enhanced Associate Domains (TEADs", J. MED. CHEM., vol. 61, 2018, pages 5057 - 5072
HELLMAN ET AL.: "Principles of Radiation Therapy, Cancer, in Principles and Practice of Oncology", vol. 1, 1993, pages: 248 - 275
HOLDEN ET AL.: "Small Molecule Dysregulation of TEAD Lipidation Induces a Dominant-Negative Inhibition of HippoPathway Signaling", CELL REPORTS, vol. 31, 2020, pages 107809
J. PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19
JERRY L. ADAMS ET AL.: "Big opportunities for small molecules in immuno-oncology", CANCER THERAPY, vol. 14, 2015, pages 603 - 622
MARTIN: "Remington's Pharmaceutical Sciences", 1975, MACK PUBL. CO.
OKAZAKI, T ET AL., NAT. IMMUNOL, vol. 14, 2013, pages 1212 - 1218
PETER L. TOOGOOD: "Small molecule immuno-oncology therapeutic agents", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 28, 2018, pages 319 - 329, XP055669954, DOI: 10.1016/j.bmcl.2017.12.044
POBBATI ET AL.: "Targeting the Central Pocket in Human Transcription Factor TEAD as a Potential Cancer Therapeutic Strategy", STRUCTURE, vol. 23, 2015, pages 2076 - 2086, XP055570312, DOI: 10.1016/j.str.2015.09.009
PRICE: "Putative allosteric MEK1 and MEK2 inhibitors", EXPERT OPIN. THER. PATENTS, vol. 18, no. 6, June 2008 (2008-06-01), pages 603 - 627, XP002494157, ISSN: 1354-3776, DOI: 10.1517/13543776.18.6.603 *
SANDRA L. ROSS ET AL.: "Bispecific T cell engager (BITE®) antibody constructs can mediate bystander tumor cell killing", PLOS ONE, vol. 12, no. 8, pages e0183390, XP055629868, DOI: 10.1371/journal.pone.0183390
THOMAS SORRELL: "Handbook of Chemistry and Physics", 1999, UNIVERSITY SCIENCE BOOKS
ZOU ET AL., SCI. TRANSL. MED, 2016, pages 8

Similar Documents

Publication Publication Date Title
CN109562106B (en) CXCR4 inhibitors and uses thereof
KR20220034739A (en) TEAD inhibitors and uses thereof
JP2022534426A (en) TEAD inhibitors and uses thereof
EP3472129A1 (en) Cxcr4 inhibitors and uses thereof
WO2022120355A1 (en) Tead degraders and uses thereof
AU2021392040A1 (en) Tead inhibitors and uses thereof
WO2022051565A1 (en) Substituted 4-piperidinyl-quinazolines, 4-piperidinyl-pyrimidine-2-amines, and related compounds and their use in treating medical conditions
WO2022120353A1 (en) Tead inhibitors and uses thereof
WO2020014465A1 (en) Polymorphic compounds and uses thereof
EP3846793B1 (en) Eif4e inhibitors and uses thereof
CA3217792A1 (en) Cdk2 degraders and uses thereof
CN116133692A (en) Methods of treating mutant lymphomas
KR20230172548A (en) MEK inhibitors and uses thereof
WO2022187518A1 (en) Substituted 4-piperidinyl-imidazo[4,5-b]pyridines and related compounds and their use in treating medical conditions
US11878958B2 (en) MEK inhibitors and uses thereof
WO2023173057A1 (en) Mek inhibitors and uses thereof
WO2023173053A1 (en) Mek inhibitors and uses thereof
US20240092779A1 (en) Usp1 inhibitors and uses thereof
US20230233546A1 (en) Methods of treating cancer
WO2022187849A1 (en) Salinomycin derivatives and uses thereof
TW202404581A (en) Mek inhibitors and uses thereof
WO2022187520A1 (en) Substituted 3-piperidinyl-pyrazolo[3,4-b]pyridines and related compounds and their use in treating medical conditions
WO2023211889A1 (en) Polymorphic compounds and uses thereof
CA3144650A1 (en) Cxcr4 inhibitors and uses 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: 23715373

Country of ref document: EP

Kind code of ref document: A1