WO2021222138A1 - Compounds for mutant ras protein degradation - Google Patents

Compounds for mutant ras protein degradation Download PDF

Info

Publication number
WO2021222138A1
WO2021222138A1 PCT/US2021/029237 US2021029237W WO2021222138A1 WO 2021222138 A1 WO2021222138 A1 WO 2021222138A1 US 2021029237 W US2021029237 W US 2021029237W WO 2021222138 A1 WO2021222138 A1 WO 2021222138A1
Authority
WO
WIPO (PCT)
Prior art keywords
cpd
indol
methyl
phenyl
propyl
Prior art date
Application number
PCT/US2021/029237
Other languages
French (fr)
Inventor
Shih-Hsien Chuang
Ying-Shuan Lailee
Chen-Hsien LIANG
Chun-Liang Lai
Yu-Shiang Lin
Hui-Jan HSU
Yann-Yu Lu
Her-Sheng LIN
Mei-ling HOU
Original Assignee
Development Center For Biotechnology
Dcb-Usa Llc
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 Development Center For Biotechnology, Dcb-Usa Llc filed Critical Development Center For Biotechnology
Publication of WO2021222138A1 publication Critical patent/WO2021222138A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 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

Definitions

  • the present invention relates generally to bifunctional compounds that bind preferably to mutant RAS proteins (e.g., KRAS, NRAS, and HRAS) than bind wild RAS proteins and promote degradation via recruitment of an E3 ubiquitin ligase, and uses of the compounds in the treatment of diseases associated with RAS mutation.
  • mutant RAS proteins e.g., KRAS, NRAS, and HRAS
  • PROTAC is a two-headed heterobifunctional molecule capable of removing unwanted proteins by inducing selective intracellular proteolysis.
  • PROTACs consist of two protein binding moieties, one for binding an E3 ubiquitin ligase and the other for binding a target protein.
  • PROTAC brings the target protein to E3 ligase, resulting in the ubiquitination of the target protein for subsequent degradation by the proteasome (Bondeson et al., "Lessons in PROTAC design from selective degradation with a promiscuous warhead.” Cell Chem Biol. 25(1): 78- 87, 2018).
  • PROTAC technology has been used in several targets: AR, ER, STAT3, BTK, FLT- 3, EGFR, BCR-ABL, BET, BRD7/9, CDK4/6, CK2, ALK, PI3K, MCL-1, PARP1 and c-MET.
  • RAS proteins are proto-oncogenes and are encoded by three RAS genes: HRAS,
  • the RAS proteins function in signal transduction pathways controlling cell growth and differentiation as binary switches, transitioning from an inactive GDP-bound state to an active GTP-bound state. Mutations usually occur at codons 12, 13, and 61 that result in the impairment of the intrinsic GTPase activity of RAS proteins, or that prevent GAP binding, activate downstream signaling pathways and contribute to tumor formation and maintenance.
  • RAS G12C mutant has a cysteine residue that has been exploited to design covalent inhibitors with promising precluded activity.
  • ARS-853 Lito et al., "Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism.” Science. 351(6273): 604-608, 2016).
  • ARS-1620, AMG-510 Canon et al., "The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumor immunity.” Nature.
  • RAS G12C inhibitor is an irreversible inhibitor of oncogenic RAS G12C . It allosterically controls GTP affinity by overturning the native nucleotide preference to favor GDP over GTP.
  • RAS G12C inhibitors are not useful for other kinds of RAS mutations such as RAS G12D , RAS G12V , RAS G12S , RAS G12R , RAS G13D , RAS Q61H , RAS Q611 , etc.
  • Pan-RAS inhibitor such as compound 3144 (Brent R Stockwell et al., Cell 168, 878-889, 2017) was designed to bind multiple sites, enabling sufficient affinity and selectivity for pharmacological RAS inhibition.
  • Compound 3144 has been reported to bind multiple adjacent sites on KRAS G12D protein near D38, A59 and Y32 sites (around SW1, SW2, and the nucleotide binding pocket) (Xu, Ke, et al. "Small molecule KRAS agonist for mutant KRAS Cancer therapy.” Molecular cancer 18(1): 85, 2019).
  • compound 3144 derivatives do not exhibit sufficient potency to move forward with clinical trials.
  • PROTAC relies on a strategy of recruiting a target protein to an E3 ubiquitin ligase and subsequently inducing proteasome-mediated degradation of the target protein.
  • the degradation induced by PROTACs is a catalytic process due to their successful dissociation after promoting polyubiquitination of the protein of interest, thereby providing great potential for allowing PROTAC action at very low doses. Therefore, PROTAC technology' may improve the potency of an inhibitor such as compound 3144 that targets switch I and 11 domain of RAS protein.
  • RAS degraders which were based on compound 3144 derivatives and conjugated with E3 ligase ligand pomalidomide/VHL through different kinds of linkers.
  • Several compounds showed activity for inducing mutant KRAS protein degradation, and thus have potential in treating diseases associated with RAS mutation.
  • tire present disclosure describes a bifunctional compound of formula (I):
  • the bifunctional compounds of formula (I) can induce the ubiquitination of RAS protein and promote its degradation in cells.
  • An advantage of tire bifunctional compounds of formula (I) provided herein is that a broad range of pharmacological activities is possible, consistent with the degradation/inhibition of targeted polypeptides from virtually any protein class or family.
  • the present disclosure provides pharmaceutical compositions comprising an effective amount of the bifunctional compound as described herein, and a pharmaceutically acceptable carrier.
  • the present disclosure provides a method of ubiquitinating and degrading a target protein in a cell comprising contacting the target protein with the bifunctional compound as described herein.
  • the present disclosure provides a method for preventing, ameliorating and/or treating diseases associated with RAS mutation in a subject in need thereof, comprising administering to tire subject a therapeutically effective amount of the bifunctional compound of formula (I) as described herein.
  • Figure 1A is a series of western blot stains showing the effect of exemplary compounds on levels of KRAS protein in MDA-MB-231 (KRAS G13D ) cells after 24 h treatment.
  • Figure IB is a series of western blot stains showing the effect of exemplary compounds on levels of KRAS protein in KLM-1 (KRAS G12D ) cells after 24 h treatment These figures demonstrate the significant KRAS lowering effects of the exemplary compounds.
  • Figure 2 is a series of western blot stains showing the effect of compound 5 on levels of KRAS protein in various human cancer cell lines after 24 h treatment. These figures demonstrate the significant KRAS lowering effects of compound 5.
  • Figure 3A is a series of western blot stains showing the effect of exemplary compounds 7 on levels of KRAS protein in Calu-1 (KRAS G12C ), MIA PaCa-2 (KRAS G12C ) and BT-474 (KRAS WT ) cells after 24h treatment
  • Figure 3B is a series of western blot stains showing the effect of exemplary compounds 43 on levels of KRAS protein in Calu-1 (KRAS G12C ), MIA PaCa-2 (KRAS G12C ), KLM-1 (KRAS G12D ), MDA-MB-231 (KRAS G13D ) and BT-474 (KRAS WT ) cells after 24h treatment.
  • Figure 4A showed tumor growth curve in Calu-1 implanted male NOD SCID mice.
  • Test articles Cpd 5 (10 mg/kg), Cpd 7 (10 mg/kg), and Cpd 39 (10 mg/kg) were given intra-tumor (IT) injection to mice once daily for consecutive 14 days.
  • One-way ANOVA followed by Dunnett’s test was applied for comparison between the vehicle and test article- treated groups. Differences are considered significant at *P ⁇ 0.05.
  • Figure 4B showed body weight changes in Calu-1 implanted male NOD SCID mice.
  • Test articles Cpd 5 (10 mg/kg), Cpd 7 (10 mg/kg), and Cpd 39 (10 mg/kg) were given intra-tumor (IT) injection to mice once daily for consecutive 14 days.
  • Figure 5A showed tumor growth curve in Calu-1 implanted male NOD SCID mice.
  • Test articles Cpd 7 (3 and 10 mg/kg), and Cpd 43 (10 mg/kg) were given intra-tumor injection to mice once daily for consecutive 28 days.
  • One-way ANOVA followed by Dunnett’s test was applied for comparison between the vehicle and test article-treated groups. Differences are considered significant at *P ⁇ 0.05.
  • Figure 5B showed body weight changes in Calu-1 implanted male NOD SCID mice.
  • Test articles Cpd 7 (3 and 10 mg/kg), and Cpd 43 (10 mg/kg) were given intra-tumor injection to mice once daily for consecutive 28 days.
  • ranges are expressed herein as from “about” one particular value and/or to "about” another particular value. When such a range is expressed, an embodiment includes the range from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the word "about,” it will be understood that the particular value forms another embodiment. It will be further understood that the endpoints of each of the ranges are significant both in relation to and independently of the other endpoint As used herein, the term “about” refers to ⁇ 20%, preferably ⁇ 10%, and even more preferably ⁇ 5%.
  • treatment generally refer to obtaining a desired pharmacological and/or physiological effect.
  • the effect maybe preventive in terms of completely or partially preventing a disease, disorder, or symptom thereof, and may be therapeutic in terms of a partial or complete cure for a disease, disorder, and/or symptoms attributed thereto.
  • Treatment used herein covers any treatment of a disease in a mammal, preferably a human, and includes (1) suppressing development of a disease, disorder, or symptom thereof in a subject or (2) relieving or ameliorating the disease, disorder, or symptom thereof in a subject.
  • preventing or “prevention” is recognized in the art, and when used in relation to a condition, it includes administering, prior to onset of the condition, an agent to reduce the frequency or severity of or deity the onset of symptoms of a medical condition in a subject relative to a subject which does not receive the agent.
  • an effective amount of an active ingredient means a sufficient amount of the ingredient to provide the desired regulation of a desired function.
  • the exact amount required will vary from subject to subject, depending on the disease state, physical conditions, age, sex, species and weight of the subject, the specific identity and formulation of the composition, etc. Dosage regimens may be adjusted to induce the optimum therapeutic response. For example, several divided doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation. Thus, it is not possible to specify an exact "effective amount.” However, an appropriate effective amount can be determined by one of ordinary skill in the art using only routine experimentation.
  • alkyl refers to a monovalent, saturated, straight or branched drain hydrocarbon radical containing 1 to 12 carbon atoms.
  • the alkyl is a C 1 -C 8 alkyl group. More preferably, tire alkyl is a C 1 - C 6 alkyl group.
  • the alkyl can be substituted or unsubstituted.
  • Examples of a C 1 -C 6 alkyl group include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, pentyl (including all isomeric forms), and hexyl (including all isomeric forms), heptyl (including all isomeric forms), and octyl (including all isomeric forms).
  • haloalkyl refers to a straight drain or branched chain alkyl group which is partly or totally halogenated.
  • the alkyl portion is as defined above.
  • Representative examples of haloalkyl include, but are not limited to, fluoromethyl, chloromethyl, bromomethyl, difluoromethyl, dichloromethyl, dibromomethyl, trifluoromethyl, trichloromethyl, 2-fluoroethyl, 2-chloroethyl, 2-bromoethyl, 2,2,2-trifluoroethyl, 3- fluoropropyl, 3-chloropropyl, 3-bromopropyl and tire like.
  • haloalkoxy refers to an alkoxy group as defined above, further attached via halo linkage.
  • C 1-6 haloalkoxy refers to an alkoxy group having from 1-6 carbon atoms, or 1-3 carbon atoms further attached via halo linkage.
  • Preferred haloalkoxy groups include, without limitation, -OCH 2 CI, -OCHCl 2 , and the like.
  • alkylene refers to a saturated, branched or straight chain hydrocarbon radical containing 1 to 12 carbon atoms and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane.
  • alkylene radicals include, but are not limited to: methylene ( — CH 2 — ), 1,2-ethylene ( — CH 2 CH 2 — ), 1,3- propylene ( — CH 2 CH 2 CH 2 — ), and 1,4-butylene ( — CH 2 CH 2 CH 2 CH 2 — ).
  • Heteroatoms such as oxygen, sulfur and nitrogen (in the form of tertiary amine moieties) may be present in the alkylene group, to provide a "heteroalkylene" group.
  • heteroalkylene radicals include, but are not limited to, -CH 2 CH 2 N(CH 3 ) 2 and - CH 2 CH 2 OCH 2 CH 3 .
  • alkenylene refers to an unsaturated, straight or branched chain hydrocarbon radical having at least one carbon-carbon double bond, comprising 2 to 12 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkene.
  • the alkenylene is a C 2 -C 8 alkenylene group. More preferably, the alkenylene is a C 2 -C 6 alkenylene group.
  • alkenylene radicals include, but are not limited to, ethenylene, propenylene, and butenylene.
  • alkynylene refers to an unsaturated straight or branched chain hydrocarbon radical having at least one carbon-carbon triple bond, comprising 2 to 12 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkyne.
  • the alkynylene is a C 2 -C 8 alkynylene group. More preferably, the alkynylene is a C 2 -C 6 alkynylene group.
  • alkynylene radicals include, but are not limited to, ethynylene, propynylene, and butynylene.
  • alkoxy refers to radicals of the general formula -O-(alkyl), wherein alkyl is as defined above.
  • exemplary alkoxy includes, but is not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, iso-butoxy, sec-butoxy, tert-butoxy, n-pentoxy, and n-hexoxy.
  • the term “carbocyclyl” refers to a monovalent hydrocarbon radical having cyclic configurations, including monocyclic, bicyclic, tricyclic, and higher multicyclic alkyl radicals (and, when multi cyclic, including fused and bridged bicyclic and spirocyclic moieties) wherein each cyclic moiety has from 3 to 12 carbon atoms.
  • the term “carbocyclyl” includes “aryl” and "cycloalkyl.”
  • aryl refers to all-carbon monocyclic or fused-ring polycyclic aromatic groups having a conjugated pi-electron system.
  • the aryl group may have 6 to 14 carbon atoms in the ring(s).
  • exemplary aryl includes, but is not limited to, phenyl, biphenyl and napthyl.
  • cycloalkyl refers to an all-carbon monocyclic or fused ring (i.e., rings which share an adjacent pair of carbon atoms) group wherein one or more rings does not have a completely conjugated pi-electron system
  • Exemplary cycloalkyl includes, but is not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, adamantanyl, 1-methylcyclopropyl, 2-methylcyclopentyl, and 2-methylcyclooctyl.
  • tire term “carbocyclylene” refers to a hydrocarbon radical having cyclic configurations, including monocyclic, bicyclic, tricyclic, and higher multicyclic alkyl radicals (and, when multicyclic, including fused and bridged bicyclic and spirocyclic moieties) wherein each cyclic moiety has from 3 to 12 carbon atoms and the radical has two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of the parent group.
  • Carbocyclylene includes “arylene” and "cycloalky lore.”
  • arylene includes, but is not limited to, phenylene, biphenylene, napthylene, and the like.
  • cycloalkylene includes, but is not limited to, cyclopropylene, cyclobutylene, cyclopentylene, cyclohexylene, cycloheptylene, cyclooctylene, 1-methylcyclopropylene, 2-methylcyclopentylene, and 2-methylcyclooctylene.
  • heterocyclic ring and “heterocyclyl” are used interchangeably.
  • heterocyclic ring or “heterocyclyl” refers to a mono-, bi-, or polycyclic structure having from 3 to 14 atoms, alternatively 3 to 12 atoms, alternatively 3 to 10 atoms, alternatively 3 to 8 atoms, alternatively 4 to 7 atoms, alternatively 5 or 6 atoms; wherein one or more atoms, for example 1, 2 or 3 atoms, are independently selected from the group consisting of N, 0, and S, the remaining ring-constituting atoms being carbon atoms.
  • the ring structure may be saturated or unsaturated, but is not aromatic.
  • heterocyclic rings include, but are not limited to, imidazolyl, imidazolinoyl, imidazolidinyl, quinolyl, isoqinolyl, indolyl, indazolyl, indazolinolyl, perhydropyridazyl, pyridazyl, pyridyl, pyrrolyl, pyrrolinyl, pyrrolidinyl, pyrazolyl, pyrazinyl, quinoxolyl, piperidinyl, pyranyl, pyrazolinyl, piperazinyl, pyrimidinyl, pyridazinyl, morpholinyl, thiamorpholinyl, furyl, thienyl, triazolyl, thiazolyl, carbolinyl.
  • heteroaryl refers to a mono-, bi- or tri-cy die aromatic radical containing 1 to 4 heteroatoms selected from S, N and 0, and includes radicals having two such monocyclic rings, or one such monocyclic ring and one monocydic aryl ring, which are directly linked by a covalent bond.
  • the heteroaiyl group may have 5 to 14 ring-forming atoms, including 1 to 13 ring-forming carbon atoms, and 1 to 8 ring-forming heteroatoms each independently selected from 0, S and N.
  • heteroaryl includes, but is not limited to, thienyl, benzthienyl, furyl, benzfuryl, pyrrolyl, imidazolyl, benzimidazolyl, thiazolyl, benzthiazolyl, isothiazolyl, benzisothiazolyl, pyrazolyl, oxazolyl, benzoxazolyl, isoxazolyl, benzisoxazolyl, isothiazolyl, triazolyl, benztriazolyl, thiadiazolyl, oxadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, indolyl and indazolyl.
  • heterocycloalkyl refers to a monocyclic or polycyclic (including 2 or more rings that are fused together, including spiro, fused, or bridged systems, for example, a bicyclic ring system), saturated or unsaturated, non-aromatic 4- to 15-membered ring system, including 1 to 14 ring-forming carbon atoms and 1 to 10 ring-forming heteroatoms each independently selected from 0, S and N.
  • heterocycloalkyl include, but are not limited to, azetidinyl, tetrahydrofuran, dihydrofuran, dioxane, morpholine, etc.
  • bifunctional compounds that bind RAS protein and recruit an E3 ligase to promote the degradation of RAS protein.
  • the present disclosure provides a bifunctional compound of formula (I),
  • RB is a RAS protein binding moiety
  • ULM is an E3 ubiquitin ligase binding moiety
  • Linker is a group that covalently binds to the RB and ULM moieties.
  • the therapeutic effect may be a result of degradation, modulation, binding, or modification of RAS protein by a compound described herein.
  • the therapeutic effect may be the result of modulation, targeting, binding, or modification of an E3 ubiquitin ligase by a compound described herein.
  • the therapeutic effect may be a result of recruitment of an E3 ubiquitin ligase by modulation, targeting, binding, or modification of the E3 ubiquitin ligase to an ubiquitinated RAS protein and marking it for proteasomal degradation, by a compound.
  • the bifunctional compound of formula (I) is useful for the treatment and/or prevention of diseases associated with RAS mutation in a subject in need thereof.
  • RB is represented formula RB-I: wherein:
  • An is phenylene substituted with one or more substituents selected from the group consisting of CM alkyl, CM alkoxy, OH, NH 2 , CN, OCF 3 , and halide;
  • Ar 2 is aryl substituted with one to three groups selected from the group consisting of H, CM alkyl, CM alkoxy, OH, NH 2 , CN, OCF 3 , and halide;
  • R 1 is a bond, C 1-6 alkylene, C3-8 cycloalkylene, or C 1-6 heteroalky lene comprising 1 to 3 heteroatoms selected from O and S;
  • R 2 is H or C 1-4 alkyl; t is an integer of 1 ⁇ 10;
  • R 3 is H, halide, or CH 3 ; and the dashed line indicates the site of attachment to Linker.
  • An is phenylene substituted with one or two substituents selected from the group consisting of OMe, NH 2 , OCF 3 , F, or Cl;
  • Ar2 is aryl substituted with one to three groups selected from the group consisting of H, CH 3 , OMe, NH 2 , CN, F, or Cl;
  • R 1 is C 2-4 alkylene, C 3-6 cycloalky lene, or C 1-4 heteroalkylene comprising a heteroatoms selected from 0 and S;
  • R 2 is H or C 1-3 alkyl; tis 1 ⁇ 4;
  • R 3 is H, F, or CH 3 ; and the dashed line indicates the site of attachment to Linker.
  • formula RB-I is selected from the group consisting of:
  • the RB binding moiety binds RAS protein with a KD value of less than about 100 ⁇ , less than about 50 ⁇ , less than about 10 ⁇ , less than about 5 ⁇ , less than about 1 ⁇ , less than about 500 nM, less than about 100 nM, or less than about
  • the RB binding moiety selectively binds mutant RAS protein over wild type RAS protein.
  • the compound of formula (I) selectively binds mutant RAS protein over wild type RAS protein.
  • the selectivity is between about 2-fold and about 5-fold. In certain embodiments, the selectivity is between about 5-fold and about 10-fold. In certain embodiments, the selectivity' is between about 10-fold and about 20-fold.
  • Linker is a divalent moiety linking RB and ULM; preferably, Linker covalently couples RB to ULM.
  • -Linker is -Ai... Aq-, wherein:
  • Aito Aq are each independently selected from the group consisting of a bond, CR L1 R L2 ,
  • R L1 and R L2 each independently can be linked to other Ai to Aq to form a cycloalkyl or heterocyclyl moiety that can be further optionally substituted with 0-4 R L5 groups;
  • R L1 , R L2 , R L3 , R L4 and R L5 are each independently selected from the group consisting of H, C 1-8 alkyl, O(C 1-8 alkyl), S(C 1-8 alkyl), NH(C 1-8 alkyl), N(C 1-8 alkyl)2, C 3-11 cycloalkyl, aryl, heteroaryl, C 3-11 heterocyclyl, O(C 1-8 cycloalkyl), S(C 1-8 cycloalkyl), NH(C 1-8 cycloalkyl),
  • N(C 1-8 cycloalkyl) 2 OH, NH 2, SH, OCH, CO 2 H, halogen, CN, CF 3 , CHF 2 , CH 2 F, and NO 2 ; and q is an integer greater than or equal to 1.
  • Linker comprises up to 30 atoms, excluding hydrogen atoms. In certain embodiments, Linker comprises up to 20 atoms, excluding hydrogen atoms. In certain embodiments, Linker comprises up to 15 atoms, excluding hydrogen atoms. In certain embodiments, Linker comprises up to 10 atoms, excluding hydrogen atoms. In certain embodiments, L comprises up to 5 atoms excluding hydrogen atoms. In certain embodiments, any of the carbon atoms in Linker can be substituted.
  • Linker is selected from the group consisting of:
  • Linker is selected from the group consisting of: wherein the dashed lines, respectively, indicate the sites of attachment to RB and ULM.
  • ULM is an E3 ubiquitin ligase binding moiety, and can be a Cereblon E3 ubiquitin ligase binding moiety (CLM), a Von Hippel-Lindau (VHL) E3 ubiquitin ligase binding moiety (VLM), a DDB1- and CUL4-associated factor 16 (DCAF16) E3 ubiquitin ligase binding moiety (DLM), an IAP E3 ubiquitin ligase binding moiety (ILM), or a mouse double minute 2 (MDM2) homolog E3 ubiquitin ligase binding moiety (MLM), or a Kelch- like ECH-associated protein-1 (KEAP1) E3 ubiquitin ligase binding moiety, or a DCAF15 E3 ubiquitin ligase binding moiety, or an RNF4 E3 ubiquitin ligase binding moiety, or a RNF114 E3 ubiquitin ligase binding moiety,
  • ULM is inclusive of all moieties that bind, can bind, or form covalent bond with any E3 ubiquitin ligase.
  • ULM is capable of binding an E3 ubiquitin ligase, such as Cereblon or von Hippel-Lindau (VHL).
  • E3 ubiquitin ligase such as Cereblon or von Hippel-Lindau (VHL).
  • ULM is capable of forming covalent bond with an E3 ubiquitin ligase, such as DCAF16.
  • ULM is capable of binding to multiple different E3 ubiquitin ligases.
  • ULM binds to Cereblon.
  • ULM binds to VHL.
  • ULM forms covalent bond with DCAF16.
  • Cereblon is an E3 ubiquitin ligase, and it forms an E3 ubiquitin ligase complex with damaged DNA binding protein 1 (DDB1), Cullin-4 A (CUL4A), and a regulator of cullins 1 (ROC1). This complex ubiquitinates a number of proteins.
  • ULM is a Cereblon E3 ligase-binding moiety (CLM) selected from the group consisting of a thalidomide, lenalidomide, pomalidomide, analogs thereof, isosteres thereof, or derivatives thereof; preferably is of the following formula: wherein:
  • Q 1 , Q 2 , Q 3 , and Q 4 are each independently C or N, and wherein one of Q 1 , Q 2 , Q 3 , and Q 4 covalently joins to Linker.
  • ULM is wherein the dashed line indicates the site of attachment to Linker.
  • VHL The von Hippel-Lindau
  • VHL comprises the substrate recognition subunit/E3 ubiquitin ligase complex VCB, which includes elongins B and C, and a complex including Cullin-2 and Rbxl.
  • the primary substrate of VHL is Hypoxia Inducible Factor 1 (HIF-la), a transcription factor that up regulates genes, such as the pro- angiogenic growth factor VEGF and the red blood cell inducing cytokine, erythropoietin, in response to low oxygen levels.
  • HIF-la Hypoxia Inducible Factor 1
  • ULM is a VHL E3 ubiquitin ligase binding moiety, and can be hydroxyproline or a derivative thereof.
  • ULM comprises a peptide backbone structure.
  • ULM is of a chemical structure represented by: wherein:
  • R 4 is H, ethyl, isopropyl, tert-butyl, sec-butyl, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl;
  • R 5 is H, haloalkyl, substituted or unsubstituted alkyl, or substituted or unsubstituted cycloalkyl;
  • R 6 is selected from the group consisting of H, halide, CN, OH, substituted or unsubstituted heteroaryl, substituted or unsubstituted aryl, substituted or unsubstituted alkyl, substituted or unsubstituted haloalkyl, substituted or unsubstituted haloalkoxy, substituted or unsubstituted cydoalkyl, and substituted or unsubstituted heterocycloalkyl; and the dashed line indicates the site of attachment to Linker.
  • R 4 is isopropyl, tert-butyl, cyclopropyl, or cyclobutyl.
  • R 5 is H, methyl, fluoromethyl, or cydopropyl.
  • ULM is of the formula:
  • ULM is of the formula:
  • DCAF16 Zhang et al.,DDBl- and CUL4-associated factor 16 (DCAF16) is a poorly characterized substrate recognition component of CUL4-DDB1 E3 ubiquitin ligases.
  • the DCAF16 protein has eight cysteine residues, and can covalently bound to DCAF16 binding moiety of a heterobifunctional degrader at a cysteine residue and then promote protein degradation.
  • ULM is a DCAF16 ligase-binding moiety (DLM), and is of a chemical structure represented by: wherein:
  • R 7 is H, halide, CM alkyl, CM alkoxy
  • Y is O, or S, orNH
  • R m is a covalent electrophile, and is selected from the following group, the dashed line linked to Y indicates the site of attachment to Linker.
  • the DCAF16 ligase-binding moiety is of the formula:
  • the E3 ligase binding moiety binds an E3 ubiquitin ligase with a KD value of less than about 10,000 nM, less than about 5,000 nM, less than about 1,000 nM, less than about 500 nM, less than about 100 nM, less than about 50 nM.
  • the E3 ligase binding moiety binds Cereblon with a KD value less than about 50 ⁇ , less than about 10,000 nM, less than about 5,000 nM, less than about 1,000 nM, less than about 500 nM, less than about 100 nM, less than about 50 nM.
  • the E3 ligase binding moiety binds VHL with a KD value of less than about 50 ⁇ , less than about 10,000 nM, less than about 5,000 nM, less than about 1,000 nM, less than about 500 nM, less than about 100 nM, less than about 50 nM.
  • the RB group and ULM group may be covalently linked to the linker group through any group which is appropriate and stable to the chemistry of the linker.
  • the linker is independently covalently bonded to the RB group and the ULM group in certain embodiments through an amide, ester, thioester, keto group, carbamate (urethane), carbon or ether, each of which groups may be inserted anywhere on the RB group and ULM group to provide maximum binding of the ULM group on the ubiquitin ligase and the RB group on the target protein to be degraded.
  • substituents for a "substituted" group are those found in the exemplary compounds and embodiments disclosed herein and can include, for example, haloide, -OH, -CF 3 , -CN, -NO 2 , alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, haloalkyl, alkylamino, aminoalkyl, dialkylamino, hydroxylalkyl, alkoxy alkyl, hydroxy alkoxy, alkoxy alkoxy, aminoalkoxy, alkylaminoalkoxy, alkylaminoalkyl, and aryl, and the like.
  • salts refers to compounds according to the invention used in the form of salts derived from inorganic or organic acids and bases. Included among add salts, for example, are the following: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, titrate, camphorate, camphors ulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, flucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxy ethanesulfonate, lactate, maleate, methanesulfonate, 2-naphthalenes ulfonate, nicotinate
  • prodrugs are intended to include any covalently bonded carriers that release the active parent drug according to formula (I) through in vivo physiological action, such as hydrolysis, metabolism and the like, when such prodrug is administered to a subject.
  • the suitability and techniques involved in making and using prodrugs are well known by a person of ordinary skill in the art.
  • Prodrugs of the compounds of formula (I) can be prepared by modifying functional groups presort in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds.
  • Prodrugs include the compounds of formula (I) wherein a hydroxy, amino, or sulfhydryl group is bonded to any group that, when the prodrugs are administered to a subject, cleaves to form a free hydroxyl, free amino, or free sulfliydryl group, respectively.
  • Examples of prodrugs include, but are not limited to, derivatives and metabolites of the compounds of formula (I) that include biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues.
  • prodrugs of the compounds of formula (I) with carboxyl functional groups are the lower alkyl (e.g., C 1 -C 6 ) esters of the carboxylic acid.
  • the carboxylate esters are conveniently formed by esterifying any of the carboxylic add moieties present on the molecule.
  • solvates means a compound of formula (I), or a pharmaceutically acceptable salt thereof, that further includes a stoichiometric or non-stoichiometric amount of a solvent bound by non-covalent intermolecular forces. If the solvent is water, the solvate may be conveniently referred to as a "hydrate,” for example, a hemi-hydrate, a mono-hydrate, a sesqui-hydrate, a di-hydrate, a tri-hydrate, etc.
  • tautomer refers to compounds whose structures differ markedly in the arrangement of atoms, but which exist in easy and rapid equilibrium, and it is to be understood that compounds provided herein may be depicted as different tautomers, and when compounds have tautomeric forms, all tautomeric forms are intended to be within the scope of the invention, and the naming of the compounds does not exclude any tautomer.
  • Exemplary tautomerizations include, but are not limited to, amide-to-imide; enamine-to-imine; enamine-to-(a different) enamine tautomerizations; and keto-to-enol.
  • stereoisomers refer to compounds that have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space. Stereoisomers include diastereomers, enantiomers, conformers and the like.
  • polymorph refers to a crystalline form of a compound (or a salt, hydrate, or solvate thereof). All polymorphs have the same elemental composition. Different crystalline forms usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optical and electrical properties, stability, and solubility. Recrystallization solvent, rate of crystallization, storage temperature, and other factors may cause one crystal form to dominate. Various polymorphs of a compound can be prepared by crystallization under different conditions.
  • isotopically enriched derivatives refers to a compound containing at least one atom having an isotopic composition other than the natural isotopic composition of that atom.
  • isotopic enrichment can be expressed in terms of the percentage of incorporation of an amount of a specific isotope at a given atom in a molecule in the place of the atom's natural isotopic abundance.
  • the bifunctional compound of the present invention may exist in one or more particular geometric, optical, enantiomeric, diasteriomeric, epimeric, atropic, stereoisomeric, tautomeric, conformational, or anomeric forms, including but not limited to, cis- and trans- forms; E- and Z-forms; c-, t-, and r-forms; endo- and exo-forms; R-, S-, and meso-forms; D- and L-forms; d- and I-forms; (+) and (-) forms; keto-, enol-, and enolate-forms; syn- and anti- forms; synclinal- and anticlinal-forms; a- and ⁇ -forms; axial and equatorial forms; boat-, chair- , twist-, envelope-, and halfchair-forms; and combinations thereof.
  • the bifunctional compound is selected from the compounds in Table 1, including:
  • the bifimctional compound as described therein binds RAS protein with a Kn value of less than about 100 ⁇ , less than about 50 ⁇ , less than about 10 ⁇ , less than about 5 ⁇ , less than about 1 ⁇ , less than about 500 nM, less than about 100 nM, less than about 50 nM.
  • the bifimctional compound as described therein selectively binds mutant RAS protein over wild type RAS protein.
  • the compound of Formula I selectively binds mutant RAS protein over wild type RAS protein.
  • the selectivity is between about 2-fold and about 5-fold. In certain embodiments, the selectivity is between about 5-fold and about 10-fold.
  • the selectivity is between about 10-fold and about 20-fold.
  • the bifunctional compound as described therein binds an E3 ubiquitin ligase with a KD value of less than about SO ⁇ , less than about 10,000 nM, less than about 5,000 nM, less than about 1,000 nM, less than about 500 nM, less than about 100 nM, less than about 50 nM.
  • the bifunctional compound as described therein promotes the degradation of mutant RAS protein up to 10%, up to 20%, up to 30%, up to 40%, up to 50%, up to 60%, up to 70%, up to 80%, up to 90%, up to 100% at a concentration of 20 ⁇ or less, 10 ⁇ or less, 5 ⁇ or less, 1,000 nM or less, 500 nM or less, 100 nM or less, 50 nM or less, 10 nM or less.
  • the bifunctional compound as described therein can be therapeutically administered as the neat chemical, but it may be useful to administer the compounds as a pharmaceutical composition or formulation.
  • the present disclosure provides a pharmaceutical composition comprising a therapeutically effective amount of the bifunctional compound as described therein or pharmaceutically acceptable salts, tautomers, stereoisomers, solvates, hydrates, polymorphs, isotopically enriched derivatives, or prodrugs thereof, and one or more pharmaceutically acceptable excipients.
  • the pharmaceutical compositions can be administered in a variety of dosage forms including, but not limited to, a solid dosage form or a liquid dosage form, an oral dosage form, a parenteral dosage form, an intranasal dosage form, a suppository, a lozenge, a troche, buccal, a controlled release dosage form, a pulsed release dosage form, an immediate release dosage form, an intravenous solution, a suspension or combinations thereof.
  • the pharmaceutical compositions can be administered, for example, by oral or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), rectal, vaginal and topical (including buccal and sublingual) administration.
  • excipient generally refers to a substance, often an inert substance, added to a pharmacological composition or otherwise used as a vehicle to further facilitate administration of a compound.
  • excipients include, but are not limited to, inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents, preservatives, effervescent mixtures, and adsorbents.
  • Suitable inert diluents include, but are not limited to, sodium and calcium carbonate, sodium and calcium phosphate, lactose, and the like.
  • Suitable disintegrating agents include, but are not limited to, starches, such as com starch, cross-linked polyvinyl pyrrolidone, agar, alginic acid, or a salt thereof, such as sodium alginate, and the like. Binding agents may include, but are not limited to, magnesium aluminum silicate, starches such as com, wheat or rice starch, gelatin, methylcellulose, sodium caiboxymethylcellulose, polyvinylpyrrolidone, and the like.
  • a lubricating agent if present, will generally be magnesium stearate and calcium stearate, stearic acid, talc, or hydrogenated vegetable oils.
  • the tablet may be coated with a material such as glyceryl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract.
  • the compositions can also be formulated as diewable tablets, for example, by using substances such as mannitol in the formulation.
  • the term "therapeutically effective amount” refers to the amount of the bifunctional compound as described therein, alone or in combination with an anticancer agent which, upon single or multiple dose administration to the subject, provides the desired effect in the subject under treatment
  • the bifunctional compound as described therein is effective in the treatment or amelioration of diseases associated with RAS mutation.
  • Degradation of the target protein will occur when the target protein is placed in proximity to the E3 ubiquitin ligase, thus resulting in degradation/inhibition of the effects of the target protein and the control of protein levels.
  • the control of protein levels afforded by the present disclosure provides treatment of a disease state or condition, which is modulated through the target protein by lowering the level of that protein in the cells of a patient.
  • the "diseases associated with RAS mutation” may be cancers, autoimmune diseases, infectious diseases, or blood vessel proliferative disorders.
  • Cancers may be lung cancer (eg., non-small cell lung cancer), colon cancer, colorectal cancer, breast cancer, prostate cancer, liver cancer, pancreatic cancer, bladder cancer, gastric cancer, renal cancer, salivary gland cancer, ovarian cancer, uterine body cancer, cervical cancer, oral cancer, skin cancer, brain cancer, lymphoma, leukemia, biliary tract malignancies, endometrial cancer, cervical cancer, or myeloid leukemia.
  • lung cancer eg., non-small cell lung cancer
  • colon cancer colorectal cancer
  • breast cancer breast cancer
  • prostate cancer liver cancer
  • pancreatic cancer bladder cancer
  • gastric cancer renal cancer
  • salivary gland cancer ovarian cancer
  • uterine body cancer cervical cancer
  • oral cancer skin cancer
  • brain cancer lymphoma
  • leukemia biliary tract malignancies
  • endometrial cancer cervical cancer
  • myeloid leukemia myeloid leukemia
  • the bifunctional compound as described therein can be administered as the sole active agent, or administered separately, sequentially or together with one or more additional anticancer agents.
  • anticancer agents refers to agents capable of inhibiting or preventing the growth of neoplasms, or checking the maturation and proliferation of malignant (cancer) cells.
  • Anticancer agents suitable for use in combination with the compounds of formula (I) include, but are not limited to, targeted cancer drugs, such as trastuzumab, ramucirumab, vismodegib, sonidegib, bevacizumab, everolimus, tamoxifen, toremifene, fulvestrant, anastrozole, exemestane, lapatinib, letrozole, pertuzumab, ado-trastuzumab emtansine, palbociclib, cetuximab, panitumumab, ziv-aflibercept, regorafenib, lmatinib mesylate, lanreotide acetate, sunitinib, regorafenib, denosumab, alitretinoin, sorafenib, pazopanib, temsirolimus, everolimus, tretinoin, dasatinib
  • the present disclosure provides a therapeutically effective amount of the bifunctional compound for manufacturing a medicament for preventing, ameliorating and/or treating a disease associated with RAS mutation in a subject in need thereof.
  • the present disclosure provides a method of ubiquitinating and degrading a target protein in a cell by contacting the target protein with the bifunctional compound of formula (I) as described herein.
  • N,N-Diisopropylethylamine (DIPEA, 294 ⁇ L, 1.69 mmol, 2 eq) was added to a solution of l-(tert-butoxycarbonyl)piperidine-4-carboxylic acid (184 mg, 0.8 mmol, 0.95 eq) and (2- (1H-benzotriazol- 1 -y 1)- 1 ,1,3,3-tetramethyluronium hexafluorophosphate(HBTU,385 mg, 0.39 mmol, 1.2 eq) in DMF (8 ml) and stirred for 30 minutes.
  • DIPEA 1,N-Diisopropylethylamine
  • Example compounds 1-4, 6-16, 19-24, 27-28, 32, 37-40 were prepared in an analogous manner to compound 5, employing the corresponding amine starting materials, like RB-1, and carboxylic acid, like L.
  • RB-2 (44 mg, 0.063 mmol) was treated with HBTU (40 mg, 0.10 mmol) in DMF (4 mL) at 0°C. After DIPEA (24 uL, 0.14 mmol) was added, the solution was stirred for 30 min at room temperature.
  • VHL.HC1 29 mg, 0.067 mmol was added, then the resulting mixture was stirred for 2 h at room temperature. Water was added to the solution and the solution was extracted with EA The organic layer was collected, dried with MgSO*, then concentrated.
  • Example compounds 18, 25-26, 29-31, 33, 35 were prepared in an analogous manner to compound 17 or 34, employing the corresponding amine starting materials, like RB- 1 or RB-2, and carboxylic acid, like compound M, or tosyloxy tert-butyl ester, like compound
  • Example compounds 41, 42, 44, 45, 47 were prepared in an analogous manner to compound 43, employing the corresponding amine starting materials, like RB-2, and carboxylic acid, like compound P.
  • RAS genes are the most frequently mutated oncogenes in human cancer. Its gain-of-fimction mutations occur in approximately 30% of all human cancers. As the most frequently mutated RAS isoform, mutated KRAS is appearing in 90% of pancreatic ductal adenocarcinoma, 30% of lung adenocarcinoma and in 40% of colorectal adenocarcinoma. These high occurrences make KRAS one of the most important targets in oncology for drug development.
  • RAS is a small GTPase transductor protein that initiates the activation of downstream signaling molecules, allowing the transmission of transducing signals from the cell surface to the nucleus, and plays a key role in essential cellular processes such as cell differentiation, growth, chemotaxis and apoptosis.
  • the RAS G12C mutant has a cysteine residue that has been exploited to design covalent inhibitors.
  • Preclinical data have shown that the KRAS G12C inhibitor blocks tumor growth for G12C KRAS gene mutations. This led to KRAS as a promising therapeutic target for cancer treatment. However, the KRAS G12C inhibitor can’t block tumor growth for other G12 and G13 KRAS gene mutations.
  • a RAS-PROTAC refers to a PROTAC that includes a target binder for the RAS protein.
  • Various compounds of RAS-PROTACs were tested in cellular RAS protein degradation via western blotting assay. The following uses RAS- PROTACs to illustrate the benefits of embodiments of the invention.
  • BT-474(KRAS WT ) and MIA PaCa-2(KRAS G12C ) cells were cultured in DMEM medium with 10% FBS.
  • BxPC3 (KRAS WT ) and KLM-1 (KRAS G12D ) cells were cultured in DMEM medium with 10% FBS.
  • MDA-MB-231 (KRAS G13D ) cells were cultured in L15 medium with 10% FBS.
  • BT-474, MIA PaCa-2, BxPC3 and MDA- MB-231 were established from the American Type Culture Collection.
  • KLM-1 were established from the ExPASy.
  • Anti-rabbit IgG, HRP-linked secondary antibodies was purchased from Cell Signaling Technology (Danvers, MA). Antibody against actin was purchased from Millipore (Burlington, MA). Immunoblots were revealed by chemiluminescence (SuperSignalTM West Femto Maximum Sensitivity Substrate, Thermo Fisher, Waltham, MA) and detected by ChemiDocTM MP Imaging System (Bio-Rad, Hercules, CA). Band intensities of western blot were also quantified by ChemiDocTM MP Imaging System. Relative intensities of bands corresponding to the drug treatment group were compared to those of the untreated group.
  • FIG. 1 shows the results of the assay.
  • Compounds 5, 10, 11, 12 and 16 are hetero-bifunctional molecule comprising a RAS binding moiety linked to a Cereblon E3 ligase binding moiety.
  • compounds 5, 11, 12 and 16 are effective in promoting the degradation of KRAS G13D in breast cancer cell line MDA-MB-231 (KRAS G13D ).
  • FIG. IB shows that compounds 5, 11, 12 and 16 are effective in promoting the degradation of KRAS G12D in pancreatic cancer cell line KLM-1 (KRAS G12D ).
  • FIG. 1 shows the results of the assay.
  • compound 5 exhibits specific KRAS mutation protein degradation activity in cancer cell lines with mutated KRAS gene such as MDA-MB-231, KLM-1, and MiaPaCa-2, instead of cell lines without mutated KRAS gene such as BT474 and BxPC-3.
  • a covalent inhibitor can only target KRAS G12C , and compound 5 can not only promote the degradation of KRAS G12C in MiaPaCa-2 cells but also KRAS G12D in KLM-1 cells and KRAS G13D in MDA-MB-231 cells.
  • Compound 5 does not cause degradation of either AKT protein or actin protein.
  • FIG. 3A shows that compound 7 exhibits specific KRAS mutation protein degradation activity in cancer cell lines Calu-1 (KRAS G12C ) and Mi A PaCa-2 (KRAS G12C ), instead of cell lines without mutated KRAS gene such as BT474 (KRAS WT ).
  • FIG. 3B shows that compound 43 exhibits specific KRAS mutation protein degradation activity in cancer cell lines MDA-MB-231 (KRAS G13D ), KLM-1 (KRAS G12D ), Calu-1 (KRAS G12C ) and MiA PaCa-2 (KRAS G12C ), instead of cell lines without mutated KRAS gene such as BT474 (KRAS* 7 ).
  • RAS-PROTACs of the invention may be used to treat diseases or disorders harboring a specific RAS mutation
  • the diseases may be cancers, autoimmune diseases, infectious diseases, or blood vessel proliferative disorders.
  • the cancers may be lung cancer (e.g., non-small cell lung cancer), colon cancer, colorectal cancer, breast cancer, prostate cancer, liver cancer, pancreatic cancer, bladder cancer, gastric cancer, renal cancer, salivary gland cancer, ovarian cancer, uterine body cancer, cervical cancer, oral cancer, skin cancer, brain cancer, lymphoma, leukemia, biliary tract malignancies, endometrial cancer, cervical cancer, or myeloid leukemia.
  • lung cancer e.g., non-small cell lung cancer
  • colon cancer colorectal cancer
  • breast cancer prostate cancer
  • liver cancer pancreatic cancer
  • bladder cancer gastric cancer
  • renal cancer salivary gland cancer
  • ovarian cancer uterine body cancer
  • cervical cancer oral cancer, skin cancer, brain cancer, lymphoma, leuk
  • RAS-PROTACs of the invention were measured using CellTiterTM-96 assay.
  • the cytotoxicities of RAS-PROTACs were evaluated in lung cancer cell lines, breast cancer cell lines and pancreatic cancer cell lines with different RAS mutations.
  • the results given in Table 9 show that RAS-PROTACs of the present invention are more toxic to cancer cells with KRAS mutations.
  • Table 10 shows the ICso of compound 5, 7, 30, 37, 39, 41, 43, 46 in the Calu-1, MIA PaCa-2 and HCT-116 cell lines. 00118] Table 9
  • the aim of this study was to evaluate the in vivo anti-tumor efficacy of Ras- PROTAC in Calu-1 human lung cancer xenograft model in male NOD SCID mice.
  • Test articles Cpd 5, Cpd 7, Cpd 39 and corresponding vehicle were formulated and given intra-tumor (IT) injection to mice once daily for consecutive 14 days.
  • the Calu-1 cells were maintained in vitro as a monolayer culture in RPMI-1640 medium supplemented with 10% fetal bovine serum at 37°C in an atmosphere of 5% CO2 in air.
  • the tumor cells were routinely sub-cultured twice weekly by trypsin-EDTA treatment.
  • the cells growing in an exponential growth phase were harvested and counted for tumor inoculation.
  • mice at age of 6-7 weeks were purchased from BioLasco Taiwan Co., LTD. and quarantined for one week. Five mice were housed in each cage. All animals were hosted in the animal facility with a 12-h light/12-h dark cycle at 19-25°C. Animals had free access to rodent pellet foods and water ad libitum. Calu-1 cells were subcutaneously (SC) implanted (5 x 10 6 cells in 1:1 PBS/matrigel mixture at 0.1 mL per mouse) into the right flank of male NOD SCID mice.
  • SC subcutaneously
  • TGI tumor growth inhibition
  • Test articles Cpd 5 (10 mg/kg), Cpd 7 (10 mg/kg), and Cpd 39 (10 mg/kg) were given intra-tumor (IT) injection to mice once daily for consecutive 14 days.
  • One-way ANOVA followed by Dunnett’s test was applied for comparison between the vehicle and test article- treated groups. Differences are considered significant at *P ⁇ 0.05.
  • Cpd 5 at 10 mg/kg showed low anti-tumor activity, with a TGI value of 25% (D28).
  • Cpd 7 at 10 mg/kg significantly reduced Calu-1 tumor growth from Day 8 to Day 28, with the TGI values ranged from 38% to 55%, and was considered as moderate efficacious to Calu-1 xenografts.
  • Cpd 39 at 10 mg/kg significantly reduced Calu-1 tumor growth from Day 8 to Day 28, with the TGI values ranged from 23% to 52%, and was considered as moderate efficacious to Calu-1 xenografts.
  • FIG. 4B showed body weight changes in Calu-1 implanted male NOD SCID mice.
  • Test articles Cpd 5 (10 mg/kg), Cpd 7 (10 mg/kg), and Cpd 39 (10 mg/kg) were given intra-tumor (IT) injection to mice once daily for consecutive 14 days. No body weight loss was observed throughout tire experiment.
  • the aim of this study was to evaluate the in vivo anti-tumor efficacy of Ras- PROTAC in Calu-1 human lung cancer xenograft model in male NOD SCID mice.
  • Test articles Cpd 7 (3 mg/kg and 10 mg/kg), Cpd 43 (10 mg/kg), and corresponding vehicle were formulated and given intra-tumor (IT) injection to mice once daily for consecutive 28 days.
  • Cpd 37 at 5 mg/kg was given intra-tumor (IT) injection to mice once daily for consecutive 14 days.
  • the Calu-1 cells were maintained in vitro as a monolayer culture in RPMI-1640 medium supplemented with 10% fetal bovine serum at 37°C in an atmosphere of 5% CO2 in air.
  • the tumor cells were routinely sub-cultured twice weekly by trypsin-EDTA treatment. The cells growing in an exponential growth phase were harvested and counted for tumor inoculation.
  • SC subcutaneously
  • TGI tumor growth inhibition
  • FIG. 5A showed tumor growth curve in Calu-1 implanted male NOD SCID mice.
  • Test articles Cpd 7 (3 and 10 mg/kg), and Cpd 43 (10 mg/kg) were given intra-tumor injection to mice once daily for consecutive 28 days.
  • One-way ANOVA followed by Dunnett’s test was applied for comparison between the vehicle and test article-treated groups. Differences are considered significant at *P ⁇ 0.05.
  • Cpd 7 at 3 mg/kg showed low anti-tumor activity, with a TGI value of 24% (D28).
  • Cpd 7 at 10 mg/kg significantly reduced Calu-1 tumor growth, with a TGI value of 51% (D28).
  • FIG. 5B showed body weight changes in Calu-1 implanted male NOD SCID mice. Test articles Cpd 7 (3 and 10 mg/kg), and Cpd 43 (10 mg/kg) were given intra-tumor injection to mice once daily for consecutive 28 days. Cpd 37 at 5 mg/kg was given intra-tumor injection to mice once daily for consecutive 14 days. No body weight loss was observed among the treatment groups.
  • the RAS-PROTACs of the invention are promising new therapeutics for cancer patients with RAS mutation.

Abstract

The present invention provides a compound of formula (I): RB-Linker-ULM which binds wild-type and mutant RAS proteins and promotes degradation via recruitment of an E3 ubiquitin ligase. Pharmaceutical compositions comprising the compound and uses thereof are also provided.

Description

COMPOUNDS FOR MUTANT RAS PROTEIN DEGRADATION
FIELD OF THE INVENTION
[0001] The present invention relates generally to bifunctional compounds that bind preferably to mutant RAS proteins (e.g., KRAS, NRAS, and HRAS) than bind wild RAS proteins and promote degradation via recruitment of an E3 ubiquitin ligase, and uses of the compounds in the treatment of diseases associated with RAS mutation.
BACKGROUND OF THE INVENTION
[0002] A proteolysis targeting chimera (PROTAC) technology was first described in
2001 (Sakamoto et al., “Protacs: chimeric molecules that target proteins to the Skpl-Cullin-F box complex for ubiquitination and degradation,” Proceedings of the National Academy of Sciences of the United States of America. 98 (15): 8554-9). PROTAC is a two-headed heterobifunctional molecule capable of removing unwanted proteins by inducing selective intracellular proteolysis. PROTACs consist of two protein binding moieties, one for binding an E3 ubiquitin ligase and the other for binding a target protein. By binding both proteins, PROTAC brings the target protein to E3 ligase, resulting in the ubiquitination of the target protein for subsequent degradation by the proteasome (Bondeson et al., "Lessons in PROTAC design from selective degradation with a promiscuous warhead." Cell Chem Biol. 25(1): 78- 87, 2018). PROTAC technology has been used in several targets: AR, ER, STAT3, BTK, FLT- 3, EGFR, BCR-ABL, BET, BRD7/9, CDK4/6, CK2, ALK, PI3K, MCL-1, PARP1 and c-MET. [0003] RAS proteins are proto-oncogenes and are encoded by three RAS genes: HRAS,
KRAS, and NRAS. The RAS proteins function in signal transduction pathways controlling cell growth and differentiation as binary switches, transitioning from an inactive GDP-bound state to an active GTP-bound state. Mutations usually occur at codons 12, 13, and 61 that result in the impairment of the intrinsic GTPase activity of RAS proteins, or that prevent GAP binding, activate downstream signaling pathways and contribute to tumor formation and maintenance. (Chang et al., "Detection of N-, H-, and KRAS codons 12, 13, and 61 mutations with universal RAS primer multiplex PCR and N-, H-, and KRAS-specific primer extension" Clin Biodiem 43(3):296-301, 2010.) Mutations of RAS genes are commonly found in numerous malignancies, including pancreatic (90%), colon (45%), and lung cancers (35%). Many tumor types have been shown to be dependent on the continued expression of oncogenic mutant RAS in cell and animal models. Pharmacologically targeting RAS is difficult because of its picomolar affinity for GTP and GDP, and because it lacks well-defined pockets for high- affinity small-molecule binding, and has therefore often been referred to as “undruggable” (Cox et al., "Drugging the undruggable Ras: mission possible?" Nat Rev Drug Discov. 13(11): 828-851, 2014.). [0004] The RASG12C mutant has a cysteine residue that has been exploited to design covalent inhibitors with promising precluded activity. To date, available RASG12C covalent inhibitors have resulted in the development of ARS-853 (Lito et al., "Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism." Science. 351(6273): 604-608, 2016). ARS-1620, AMG-510 (Canon et al., "The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumor immunity." Nature. 575: 217-223, 2019) and MRTX-849 (Christensen et al., "The KRASG12C Inhibitor, MRTX849, Provides Insight Toward Therapeutic Susceptibility of KRAS Mutant Cancers in Mouse Models and Patients." Cancer Discov. 10(1):54-71, 2020). RASG12C inhibitor is an irreversible inhibitor of oncogenic RASG12C. It allosterically controls GTP affinity by overturning the native nucleotide preference to favor GDP over GTP. However,
RASG12C inhibitors are not useful for other kinds of RAS mutations such as RASG12D, RASG12V, RASG12S, RASG12R, RASG13D, RASQ61H, RASQ611, etc.
[0005] To overcome the limitation of RASG12C inhibitor that only can target RASG12C, several small molecule binding sites on RAS have been explored for RAS inhibitor development. Pan-RAS inhibitor such as compound 3144 (Brent R Stockwell et al., Cell 168, 878-889, 2017) was designed to bind multiple sites, enabling sufficient affinity and selectivity for pharmacological RAS inhibition. Compound 3144 has been reported to bind multiple adjacent sites on KRAS G12D protein near D38, A59 and Y32 sites (around SW1, SW2, and the nucleotide binding pocket) (Xu, Ke, et al. "Small molecule KRAS agonist for mutant KRAS Cancer therapy." Molecular cancer 18(1): 85, 2019). However, compound 3144 derivatives do not exhibit sufficient potency to move forward with clinical trials.
[0006] PROTAC relies on a strategy of recruiting a target protein to an E3 ubiquitin ligase and subsequently inducing proteasome-mediated degradation of the target protein. The degradation induced by PROTACs is a catalytic process due to their successful dissociation after promoting polyubiquitination of the protein of interest, thereby providing great potential for allowing PROTAC action at very low doses. Therefore, PROTAC technology' may improve the potency of an inhibitor such as compound 3144 that targets switch I and 11 domain of RAS protein. In this invention, we present our design, synthesis and biological evaluation of RAS degraders, which were based on compound 3144 derivatives and conjugated with E3 ligase ligand pomalidomide/VHL through different kinds of linkers. Several compounds showed activity for inducing mutant KRAS protein degradation, and thus have potential in treating diseases associated with RAS mutation.
SUMMARY OF THE INVENTION [0007] In one aspect, tire present disclosure describes a bifunctional compound of formula (I):
RB— Linker— ULM (I) or pharmaceutically acceptable salts, tautomers, stereoisomers, solvates, hydrates, polymorphs, isotopically enriched derivatives, or prodrugs thereof. The bifunctional compounds of formula (I) can induce the ubiquitination of RAS protein and promote its degradation in cells. An advantage of tire bifunctional compounds of formula (I) provided herein is that a broad range of pharmacological activities is possible, consistent with the degradation/inhibition of targeted polypeptides from virtually any protein class or family. [0008] In an additional aspect, the present disclosure provides pharmaceutical compositions comprising an effective amount of the bifunctional compound as described herein, and a pharmaceutically acceptable carrier.
[0009] In another aspect, the present disclosure provides a method of ubiquitinating and degrading a target protein in a cell comprising contacting the target protein with the bifunctional compound as described herein.
[0010] In still another aspect, the present disclosure provides a method for preventing, ameliorating and/or treating diseases associated with RAS mutation in a subject in need thereof, comprising administering to tire subject a therapeutically effective amount of the bifunctional compound of formula (I) as described herein.
BRIEF DESCRIPTION OF THE DRAWINGS
[0011] Figure 1A is a series of western blot stains showing the effect of exemplary compounds on levels of KRAS protein in MDA-MB-231 (KRASG13D) cells after 24 h treatment. Figure IB is a series of western blot stains showing the effect of exemplary compounds on levels of KRAS protein in KLM-1 (KRASG12D) cells after 24 h treatment These figures demonstrate the significant KRAS lowering effects of the exemplary compounds. [0012] Figure 2 is a series of western blot stains showing the effect of compound 5 on levels of KRAS protein in various human cancer cell lines after 24 h treatment. These figures demonstrate the significant KRAS lowering effects of compound 5.
[0013] Figure 3A is a series of western blot stains showing the effect of exemplary compounds 7 on levels of KRAS protein in Calu-1 (KRASG12C), MIA PaCa-2 (KRASG12C) and BT-474 (KRASWT) cells after 24h treatment Figure 3B is a series of western blot stains showing the effect of exemplary compounds 43 on levels of KRAS protein in Calu-1 (KRASG12C), MIA PaCa-2 (KRASG12C), KLM-1 (KRASG12D), MDA-MB-231 (KRASG13D) and BT-474 (KRASWT) cells after 24h treatment. [0014] Figure 4A showed tumor growth curve in Calu-1 implanted male NOD SCID mice. Test articles Cpd 5 (10 mg/kg), Cpd 7 (10 mg/kg), and Cpd 39 (10 mg/kg) were given intra-tumor (IT) injection to mice once daily for consecutive 14 days. One-way ANOVA followed by Dunnett’s test was applied for comparison between the vehicle and test article- treated groups. Differences are considered significant at *P<0.05. Figure 4B showed body weight changes in Calu-1 implanted male NOD SCID mice. Test articles Cpd 5 (10 mg/kg), Cpd 7 (10 mg/kg), and Cpd 39 (10 mg/kg) were given intra-tumor (IT) injection to mice once daily for consecutive 14 days.
[0015] Figure 5A showed tumor growth curve in Calu-1 implanted male NOD SCID mice. Test articles Cpd 7 (3 and 10 mg/kg), and Cpd 43 (10 mg/kg) were given intra-tumor injection to mice once daily for consecutive 28 days. Cpd 37 at 5 mg/kg was given intra-tumor injection to mice once daily for consecutive 14 days. One-way ANOVA followed by Dunnett’s test was applied for comparison between the vehicle and test article-treated groups. Differences are considered significant at *P<0.05. Figure 5B showed body weight changes in Calu-1 implanted male NOD SCID mice. Test articles Cpd 7 (3 and 10 mg/kg), and Cpd 43 (10 mg/kg) were given intra-tumor injection to mice once daily for consecutive 28 days. Cpd 37 at 5 mg/kg was given intra-tumor injection to mice once daily for consecutive 14 days.
DETAILED DESCRIPTION
Definitions [0016] In order for the present disclosure to be fully understood, the following detailed description is set forth. In the description, the following terms are employed:
[0017] It must be noted that, as used herein, the singular forms "a" " an" and "the" include plural referents unless the context clearly dictates otherwise. Thus, unless otherwise required by context, singular terms shall include the plural and plural terms shall include the singular.
[0018] Often, ranges are expressed herein as from "about" one particular value and/or to "about" another particular value. When such a range is expressed, an embodiment includes the range from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the word "about," it will be understood that the particular value forms another embodiment. It will be further understood that the endpoints of each of the ranges are significant both in relation to and independently of the other endpoint As used herein, the term "about" refers to ± 20%, preferably ± 10%, and even more preferably ± 5%.
[0019] The term "and/or" is used to refer to both things or either one of the two mentioned. [0020] The terms "treatment," "treating," and "treat" generally refer to obtaining a desired pharmacological and/or physiological effect. The effect maybe preventive in terms of completely or partially preventing a disease, disorder, or symptom thereof, and may be therapeutic in terms of a partial or complete cure for a disease, disorder, and/or symptoms attributed thereto. "Treatment" used herein covers any treatment of a disease in a mammal, preferably a human, and includes (1) suppressing development of a disease, disorder, or symptom thereof in a subject or (2) relieving or ameliorating the disease, disorder, or symptom thereof in a subject.
[0021] The term "preventing" or "prevention" is recognized in the art, and when used in relation to a condition, it includes administering, prior to onset of the condition, an agent to reduce the frequency or severity of or deity the onset of symptoms of a medical condition in a subject relative to a subject which does not receive the agent.
[0022] The terms "individual," "subject," and "patient" herein are used interchangeably and refer to any mammalian subject for whom diagnosis, treatment, or therapy is desired.
[0023] The term "effective amount" of an active ingredient as provided herein means a sufficient amount of the ingredient to provide the desired regulation of a desired function. As will be pointed out below, the exact amount required will vary from subject to subject, depending on the disease state, physical conditions, age, sex, species and weight of the subject, the specific identity and formulation of the composition, etc. Dosage regimens may be adjusted to induce the optimum therapeutic response. For example, several divided doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation. Thus, it is not possible to specify an exact "effective amount." However, an appropriate effective amount can be determined by one of ordinary skill in the art using only routine experimentation.
[0024] Unless otherwise specified, the term "alkyl" as used herein refers to a monovalent, saturated, straight or branched drain hydrocarbon radical containing 1 to 12 carbon atoms. Preferably, the alkyl is a C1-C8 alkyl group. More preferably, tire alkyl is a C1- C6 alkyl group. The alkyl can be substituted or unsubstituted. Examples of a C1-C6 alkyl group include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, pentyl (including all isomeric forms), and hexyl (including all isomeric forms), heptyl (including all isomeric forms), and octyl (including all isomeric forms).
[0025] The term "haloalkyl" as used herein refers to a straight drain or branched chain alkyl group which is partly or totally halogenated. The alkyl portion is as defined above. Representative examples of haloalkyl include, but are not limited to, fluoromethyl, chloromethyl, bromomethyl, difluoromethyl, dichloromethyl, dibromomethyl, trifluoromethyl, trichloromethyl, 2-fluoroethyl, 2-chloroethyl, 2-bromoethyl, 2,2,2-trifluoroethyl, 3- fluoropropyl, 3-chloropropyl, 3-bromopropyl and tire like. [0026] The term "haloalkoxy " as used herein refers to an alkoxy group as defined above, further attached via halo linkage. For example, C1-6 haloalkoxy refers to an alkoxy group having from 1-6 carbon atoms, or 1-3 carbon atoms further attached via halo linkage. Preferred haloalkoxy groups include, without limitation, -OCH2CI, -OCHCl2, and the like. [0027] Unless otherwise specified, the term "alkylene" as used herein refers to a saturated, branched or straight chain hydrocarbon radical containing 1 to 12 carbon atoms and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane. Examples of alkylene radicals include, but are not limited to: methylene ( — CH2 — ), 1,2-ethylene ( — CH2CH2 — ), 1,3- propylene ( — CH2CH2CH2 — ), and 1,4-butylene ( — CH2CH2CH2CH2 — ).
[0028] Heteroatoms such as oxygen, sulfur and nitrogen (in the form of tertiary amine moieties) may be present in the alkylene group, to provide a "heteroalkylene" group. Examples of heteroalkylene radicals include, but are not limited to, -CH2CH2N(CH3)2 and - CH2CH2OCH2CH3. [0029] Unless otherwise specified, the term "alkenylene" as used herein refers to an unsaturated, straight or branched chain hydrocarbon radical having at least one carbon-carbon double bond, comprising 2 to 12 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkene. Preferably, the alkenylene is a C2-C8 alkenylene group. More preferably, the alkenylene is a C2-C6 alkenylene group. Examples of alkenylene radicals include, but are not limited to, ethenylene, propenylene, and butenylene.
[0030] Unless otherwise specified, the term "alkynylene" used herein refers to an unsaturated straight or branched chain hydrocarbon radical having at least one carbon-carbon triple bond, comprising 2 to 12 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkyne. Preferably, the alkynylene is a C2-C8 alkynylene group. More preferably, the alkynylene is a C2-C6 alkynylene group. Examples of alkynylene radicals include, but are not limited to, ethynylene, propynylene, and butynylene.
[0031] Unless otherwise specified, the term "alkoxy" as used herein refers to radicals of the general formula -O-(alkyl), wherein alkyl is as defined above. Exemplary alkoxy includes, but is not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, iso-butoxy, sec-butoxy, tert-butoxy, n-pentoxy, and n-hexoxy.
[0032] As used herein, the term "carbocyclyl" refers to a monovalent hydrocarbon radical having cyclic configurations, including monocyclic, bicyclic, tricyclic, and higher multicyclic alkyl radicals (and, when multi cyclic, including fused and bridged bicyclic and spirocyclic moieties) wherein each cyclic moiety has from 3 to 12 carbon atoms. The term "carbocyclyl" includes "aryl" and "cycloalkyl." [0033] As used herein, the term "aryl" refers to all-carbon monocyclic or fused-ring polycyclic aromatic groups having a conjugated pi-electron system. The aryl group may have 6 to 14 carbon atoms in the ring(s). Exemplary aryl includes, but is not limited to, phenyl, biphenyl and napthyl. [0034] As used herein, the term "cycloalkyl" refers to an all-carbon monocyclic or fused ring (i.e., rings which share an adjacent pair of carbon atoms) group wherein one or more rings does not have a completely conjugated pi-electron system Exemplary cycloalkyl includes, but is not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, adamantanyl, 1-methylcyclopropyl, 2-methylcyclopentyl, and 2-methylcyclooctyl. [0035] As used herein, tire term "carbocyclylene" refers to a hydrocarbon radical having cyclic configurations, including monocyclic, bicyclic, tricyclic, and higher multicyclic alkyl radicals (and, when multicyclic, including fused and bridged bicyclic and spirocyclic moieties) wherein each cyclic moiety has from 3 to 12 carbon atoms and the radical has two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of the parent group. The term "carbocyclylene" includes "arylene" and "cycloalky lore." Exemplary arylene includes, but is not limited to, phenylene, biphenylene, napthylene, and the like. Exemplary cycloalkylene includes, but is not limited to, cyclopropylene, cyclobutylene, cyclopentylene, cyclohexylene, cycloheptylene, cyclooctylene, 1-methylcyclopropylene, 2-methylcyclopentylene, and 2-methylcyclooctylene. [0036] As used herein, the terms "heterocyclic ring" and "heterocyclyl" are used interchangeably. The term "heterocyclic ring" or "heterocyclyl" refers to a mono-, bi-, or polycyclic structure having from 3 to 14 atoms, alternatively 3 to 12 atoms, alternatively 3 to 10 atoms, alternatively 3 to 8 atoms, alternatively 4 to 7 atoms, alternatively 5 or 6 atoms; wherein one or more atoms, for example 1, 2 or 3 atoms, are independently selected from the group consisting of N, 0, and S, the remaining ring-constituting atoms being carbon atoms. The ring structure may be saturated or unsaturated, but is not aromatic. Exemplary heterocyclic rings include, but are not limited to, imidazolyl, imidazolinoyl, imidazolidinyl, quinolyl, isoqinolyl, indolyl, indazolyl, indazolinolyl, perhydropyridazyl, pyridazyl, pyridyl, pyrrolyl, pyrrolinyl, pyrrolidinyl, pyrazolyl, pyrazinyl, quinoxolyl, piperidinyl, pyranyl, pyrazolinyl, piperazinyl, pyrimidinyl, pyridazinyl, morpholinyl, thiamorpholinyl, furyl, thienyl, triazolyl, thiazolyl, carbolinyl. tetrazolyl, thiazolidinyl, benzofuranoyl, thiamorpholinyl sulfone, oxazolyl, benzoxazolyl, oxopiperidinyl, oxopyrrolidinyl, oxoazepinyl, azepinyl, isoxozolyl, isothiazolyl, furazanyl, tetrahydropyranyl, tetrahydrofuranyl, thiazolyl, thiadiazoyl, dioxolyl, dioxinyl, oxathiolyl, benzodioxolyl, dithiolyl, thiophenyl, tetrahydrothiophenyl, sulfolanyl, dioxanyl, dioxolanyl, tetahydrofurodihydrofuranyl, tetrahydropyranodihydrofuranyl, dihydropyranyl, tetradyrofurofuranyl, and tetrahydropyranofuranyl.
[0037] As used herein, tire term "heteroaryl" refers to a mono-, bi- or tri-cy die aromatic radical containing 1 to 4 heteroatoms selected from S, N and 0, and includes radicals having two such monocyclic rings, or one such monocyclic ring and one monocydic aryl ring, which are directly linked by a covalent bond. The heteroaiyl group may have 5 to 14 ring-forming atoms, including 1 to 13 ring-forming carbon atoms, and 1 to 8 ring-forming heteroatoms each independently selected from 0, S and N. Exemplary heteroaryl includes, but is not limited to, thienyl, benzthienyl, furyl, benzfuryl, pyrrolyl, imidazolyl, benzimidazolyl, thiazolyl, benzthiazolyl, isothiazolyl, benzisothiazolyl, pyrazolyl, oxazolyl, benzoxazolyl, isoxazolyl, benzisoxazolyl, isothiazolyl, triazolyl, benztriazolyl, thiadiazolyl, oxadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, indolyl and indazolyl.
[0038] As used herein, the term "heterocycloalkyl" refers to a monocyclic or polycyclic (including 2 or more rings that are fused together, including spiro, fused, or bridged systems, for example, a bicyclic ring system), saturated or unsaturated, non-aromatic 4- to 15-membered ring system, including 1 to 14 ring-forming carbon atoms and 1 to 10 ring-forming heteroatoms each independently selected from 0, S and N. Examples of heterocycloalkyl include, but are not limited to, azetidinyl, tetrahydrofuran, dihydrofuran, dioxane, morpholine, etc. [0039] As used herein, the terms "halide" and "halo" are used interchangeably and include fluoro, chloro, bromo and iodo.
Compounds
[0040] Provided herein are bifunctional compounds that bind RAS protein and recruit an E3 ligase to promote the degradation of RAS protein. In one aspect, the present disclosure provides a bifunctional compound of formula (I),
RB— Linker— ULM (I) or pharmaceutically acceptable salts, solvates, hydrates, polymorphs, tautomers, stereoisomers, isotopically enriched derivatives, or prodrugs thereof, wherein:
RB is a RAS protein binding moiety;
ULM is an E3 ubiquitin ligase binding moiety; and
Linker is a group that covalently binds to the RB and ULM moieties.
[0041] The bifunctional compound described herein interacts with RAS protein and an
E3 ubiquitin ligase. As described herein, the therapeutic effect may be a result of degradation, modulation, binding, or modification of RAS protein by a compound described herein. Without wishing to be bound by any particular theory, the therapeutic effect may be the result of modulation, targeting, binding, or modification of an E3 ubiquitin ligase by a compound described herein. The therapeutic effect may be a result of recruitment of an E3 ubiquitin ligase by modulation, targeting, binding, or modification of the E3 ubiquitin ligase to an ubiquitinated RAS protein and marking it for proteasomal degradation, by a compound. Specifically, the bifunctional compound of formula (I) is useful for the treatment and/or prevention of diseases associated with RAS mutation in a subject in need thereof.
RB
[0042] In certain embodiments, RB is represented formula RB-I:
Figure imgf000011_0001
wherein:
An is phenylene substituted with one or more substituents selected from the group consisting of CM alkyl, CM alkoxy, OH, NH2, CN, OCF3, and halide;
Ar2 is aryl substituted with one to three groups selected from the group consisting of H, CM alkyl, CM alkoxy, OH, NH2, CN, OCF3, and halide;
R1 is a bond, C1-6 alkylene, C3-8 cycloalkylene, or C1-6 heteroalky lene comprising 1 to 3 heteroatoms selected from O and S;
R2 is H or C1-4 alkyl;
Figure imgf000011_0002
Figure imgf000012_0001
t is an integer of 1~10;
R3 is H, halide, or CH3; and the dashed line indicates the site of attachment to Linker. [0043] Preferably, in formula RB-I, wherein
An is phenylene substituted with one or two substituents selected from the group consisting of OMe, NH2, OCF3, F, or Cl;
Ar2 is aryl substituted with one to three groups selected from the group consisting of H, CH3, OMe, NH2, CN, F, or Cl;
R1 is C2-4 alkylene, C3-6 cycloalky lene, or C1-4 heteroalkylene comprising a heteroatoms selected from 0 and S;
R2 is H or C1-3 alkyl;
Figure imgf000012_0002
tis 1~4;
R3 is H, F, or CH3; and the dashed line indicates the site of attachment to Linker.
[0044] In certain embodiments, formula RB-I is selected from the group consisting of:
Figure imgf000013_0001
Figure imgf000014_0001
wherein the dashed line indicates the site of attachment to Linker.
[0045] In certain embodiments, the RB binding moiety binds RAS protein with a KD value of less than about 100 μΜ, less than about 50 μΜ, less than about 10 μΜ, less than about 5 μΜ, less than about 1 μΜ, less than about 500 nM, less than about 100 nM, or less than about
50 nM.
[0046] In certain embodiments, the RB binding moiety selectively binds mutant RAS protein over wild type RAS protein. In some embodiments, the compound of formula (I) selectively binds mutant RAS protein over wild type RAS protein. In certain embodiments, the selectivity is between about 2-fold and about 5-fold. In certain embodiments, the selectivity is between about 5-fold and about 10-fold. In certain embodiments, the selectivity' is between about 10-fold and about 20-fold.
Linker
[0047] Linker is a divalent moiety linking RB and ULM; preferably, Linker covalently couples RB to ULM. In certain embodiments, -Linker is -Ai... Aq-, wherein:
Aito Aq are each independently selected from the group consisting of a bond, CRL1RL2,
O, S, S=O, S(=O)2, NRL3, C(=O), C(=O)NRL3, NRL3C(=O), C≡C, C3-11 cycloalkyl optionally substituted with 0-6 substituents selected from the group consisting of RL1 and RL2, C3.11 heterocyclyl optionally substituted with 0-6 substituents selected from the group consisting of RL1 and RL2, aryl optionally substituted with 0-6 substituents selected from the group consisting of RL1 and RL2, and heteroaryl optionally substituted with 0-6 substituents selected from the group consisting of RL1 and RL2, wherein:
RL1 and RL2 each independently can be linked to other Ai to Aq to form a cycloalkyl or heterocyclyl moiety that can be further optionally substituted with 0-4 RL5 groups; RL1, RL2, R L3 , RL4 and RL5 are each independently selected from the group consisting of H, C1-8 alkyl, O(C1-8 alkyl), S(C1-8 alkyl), NH(C1-8 alkyl), N(C1-8 alkyl)2, C3-11 cycloalkyl, aryl, heteroaryl, C3-11 heterocyclyl, O(C1-8 cycloalkyl), S(C1-8 cycloalkyl), NH(C1-8 cycloalkyl),
N(C1-8 cycloalkyl)2, OH, NH2, SH, OCH, CO2H, halogen, CN, CF3, CHF2, CH2F, and NO2; and q is an integer greater than or equal to 1.
[0048] In certain embodiments, Linker comprises up to 30 atoms, excluding hydrogen atoms. In certain embodiments, Linker comprises up to 20 atoms, excluding hydrogen atoms. In certain embodiments, Linker comprises up to 15 atoms, excluding hydrogen atoms. In certain embodiments, Linker comprises up to 10 atoms, excluding hydrogen atoms. In certain embodiments, L comprises up to 5 atoms excluding hydrogen atoms. In certain embodiments, any of the carbon atoms in Linker can be substituted.
[0049] In certain embodiments, Linker is selected from the group consisting of:
Figure imgf000015_0001
Figure imgf000016_0001
wherein: k is an integer of 0 or 1; mis an integer selected from 0~12; n is an integer selected from 0—8; and the dashed lines, respectively, indicate the sites of attachment to RB and ULM.
[0050] In certain embodiments, Linker is selected from the group consisting of:
Figure imgf000017_0001
wherein the dashed lines, respectively, indicate the sites of attachment to RB and ULM. ULM
[0051] ULM is an E3 ubiquitin ligase binding moiety, and can be a Cereblon E3 ubiquitin ligase binding moiety (CLM), a Von Hippel-Lindau (VHL) E3 ubiquitin ligase binding moiety (VLM), a DDB1- and CUL4-associated factor 16 (DCAF16) E3 ubiquitin ligase binding moiety (DLM), an IAP E3 ubiquitin ligase binding moiety (ILM), or a mouse double minute 2 (MDM2) homolog E3 ubiquitin ligase binding moiety (MLM), or a Kelch- like ECH-associated protein-1 (KEAP1) E3 ubiquitin ligase binding moiety, or a DCAF15 E3 ubiquitin ligase binding moiety, or an RNF4 E3 ubiquitin ligase binding moiety, or a RNF114 E3 ubiquitin ligase binding moiety, or a arylhydrocarbon receptor (AhR) E3 ubiquitin ligase binding moiety, or others E3 ubiquitin ligase binding moiety described in SLAS Discovery, 1- 19, 2020. ULM is inclusive of all moieties that bind, can bind, or form covalent bond with any E3 ubiquitin ligase. For example, in certain embodiments, ULM is capable of binding an E3 ubiquitin ligase, such as Cereblon or von Hippel-Lindau (VHL). In certain embodiments, ULM is capable of forming covalent bond with an E3 ubiquitin ligase, such as DCAF16. In certain embodiments, ULM is capable of binding to multiple different E3 ubiquitin ligases. In certain embodiments, ULM binds to Cereblon. In certain embodiments, ULM binds to VHL. In certain embodiments, ULM forms covalent bond with DCAF16.
[0052] Cereblon is an E3 ubiquitin ligase, and it forms an E3 ubiquitin ligase complex with damaged DNA binding protein 1 (DDB1), Cullin-4 A (CUL4A), and a regulator of cullins 1 (ROC1). This complex ubiquitinates a number of proteins.
[0053] In certain embodiments, ULM is a Cereblon E3 ligase-binding moiety (CLM) selected from the group consisting of a thalidomide, lenalidomide, pomalidomide, analogs thereof, isosteres thereof, or derivatives thereof; preferably is of the following formula:
Figure imgf000018_0001
wherein:
W is selected from CH2 and C=O; and
Q1, Q2, Q3, and Q4 are each independently C or N, and wherein one of Q1, Q2, Q3, and Q4 covalently joins to Linker.
[0054] In certain embodiments, ULM is
Figure imgf000018_0002
wherein the dashed line indicates the site of attachment to Linker.
[0055] The von Hippel-Lindau (VHL) is an E3 ubiquitin ligase. VHL comprises the substrate recognition subunit/E3 ubiquitin ligase complex VCB, which includes elongins B and C, and a complex including Cullin-2 and Rbxl. The primary substrate of VHL is Hypoxia Inducible Factor 1 (HIF-la), a transcription factor that up regulates genes, such as the pro- angiogenic growth factor VEGF and the red blood cell inducing cytokine, erythropoietin, in response to low oxygen levels. VCB is a known target in cancer, chronic anemia, and ischemia In an embodiment, ULM is a VHL E3 ubiquitin ligase binding moiety, and can be hydroxyproline or a derivative thereof. [0056] In certain embodiments, ULM comprises a peptide backbone structure. In certain embodiments, ULM is of a chemical structure represented by:
Figure imgf000019_0001
wherein:
R4 is H, ethyl, isopropyl, tert-butyl, sec-butyl, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl;
R5 is H, haloalkyl, substituted or unsubstituted alkyl, or substituted or unsubstituted cycloalkyl;
R6 is selected from the group consisting of H, halide, CN, OH, substituted or unsubstituted heteroaryl, substituted or unsubstituted aryl, substituted or unsubstituted alkyl, substituted or unsubstituted haloalkyl, substituted or unsubstituted haloalkoxy, substituted or unsubstituted cydoalkyl, and substituted or unsubstituted heterocycloalkyl; and the dashed line indicates the site of attachment to Linker. [0057] In a preferred embodiment, R4 is isopropyl, tert-butyl, cyclopropyl, or cyclobutyl. In another preferred embodiment, R5 is H, methyl, fluoromethyl, or cydopropyl.
[0058] In certain embodiments, ULM is of the formula:
Figure imgf000019_0002
[0059] In certain embodiments, ULM is of the formula:
Figure imgf000020_0001
[0060] As described in Nature Chemical Biology, VOL 15, 2019, p73791og by Xiaoyu
Zhang et al.,DDBl- and CUL4-associated factor 16 (DCAF16) is a poorly characterized substrate recognition component of CUL4-DDB1 E3 ubiquitin ligases. The DCAF16 protein has eight cysteine residues, and can covalently bound to DCAF16 binding moiety of a heterobifunctional degrader at a cysteine residue and then promote protein degradation.
[0061] In certain embodiments, ULM is a DCAF16 ligase-binding moiety (DLM), and is of a chemical structure represented by:
Figure imgf000020_0002
wherein:
R7 is H, halide, CM alkyl, CM alkoxy;
Y is O, or S, orNH;
Rm is a covalent electrophile, and is selected from the following group,
Figure imgf000020_0003
the dashed line linked to Y indicates the site of attachment to Linker.
[0062] In certain embodiments, the DCAF16 ligase-binding moiety is of the formula:
Figure imgf000021_0001
[0063] In certain embodiments, the E3 ligase binding moiety binds an E3 ubiquitin ligase with a KD value of less than about 10,000 nM, less than about 5,000 nM, less than about 1,000 nM, less than about 500 nM, less than about 100 nM, less than about 50 nM. [0064] In certain embodiments, the E3 ligase binding moiety binds Cereblon with a KD value less than about 50 μΜ, less than about 10,000 nM, less than about 5,000 nM, less than about 1,000 nM, less than about 500 nM, less than about 100 nM, less than about 50 nM. [0065] In certain embodiments, the E3 ligase binding moiety binds VHL with a KD value of less than about 50 μΜ, less than about 10,000 nM, less than about 5,000 nM, less than about 1,000 nM, less than about 500 nM, less than about 100 nM, less than about 50 nM.
[0066] The RB group and ULM group may be covalently linked to the linker group through any group which is appropriate and stable to the chemistry of the linker. In exemplary aspects of the present invention, the linker is independently covalently bonded to the RB group and the ULM group in certain embodiments through an amide, ester, thioester, keto group, carbamate (urethane), carbon or ether, each of which groups may be inserted anywhere on the RB group and ULM group to provide maximum binding of the ULM group on the ubiquitin ligase and the RB group on the target protein to be degraded.
[0067] As used herein, the phrase "substituted or unsubstituted" means that substitution is optional. In the event a substitution is desired, then such substitution means that any number of hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the normal valence of the designated atom is not exceeded, and that the substitution results in a stable compound. For example, when a substituent is keto (i.e., =O), then 2 hydrogens on the atom are replaced. Examples of substituents for a "substituted" group are those found in the exemplary compounds and embodiments disclosed herein and can include, for example, haloide, -OH, -CF3, -CN, -NO2, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, haloalkyl, alkylamino, aminoalkyl, dialkylamino, hydroxylalkyl, alkoxy alkyl, hydroxy alkoxy, alkoxy alkoxy, aminoalkoxy, alkylaminoalkoxy, alkylaminoalkyl, and aryl, and the like.
[0068] The term "pharmaceutically acceptable salts" as used herein refers to compounds according to the invention used in the form of salts derived from inorganic or organic acids and bases. Included among add salts, for example, are the following: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, titrate, camphorate, camphors ulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, flucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxy ethanesulfonate, lactate, maleate, methanesulfonate, 2-naphthalenes ulfonate, nicotinate, oxalate, pamoate, pectianate, persulfate, phenylproprionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate and undecanoate. Salts derived from appropriate bases include alkali metal (e.g. sodium), alkaline earth metal (e.g., magnesium), ammonium and NW4+ (wherein W is CM alkyl).
[0069] As used herein, "prodrugs" are intended to include any covalently bonded carriers that release the active parent drug according to formula (I) through in vivo physiological action, such as hydrolysis, metabolism and the like, when such prodrug is administered to a subject. The suitability and techniques involved in making and using prodrugs are well known by a person of ordinary skill in the art. Prodrugs of the compounds of formula (I) (parent compounds) can be prepared by modifying functional groups presort in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds. "Prodrugs" include the compounds of formula (I) wherein a hydroxy, amino, or sulfhydryl group is bonded to any group that, when the prodrugs are administered to a subject, cleaves to form a free hydroxyl, free amino, or free sulfliydryl group, respectively. Examples of prodrugs include, but are not limited to, derivatives and metabolites of the compounds of formula (I) that include biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues. In certain embodiments, prodrugs of the compounds of formula (I) with carboxyl functional groups are the lower alkyl (e.g., C1-C6) esters of the carboxylic acid. The carboxylate esters are conveniently formed by esterifying any of the carboxylic add moieties present on the molecule.
[0070] The compounds of the invention can exist as solvates. As used herein and unless otherwise indicated, the term "solvate" means a compound of formula (I), or a pharmaceutically acceptable salt thereof, that further includes a stoichiometric or non-stoichiometric amount of a solvent bound by non-covalent intermolecular forces. If the solvent is water, the solvate may be conveniently referred to as a "hydrate," for example, a hemi-hydrate, a mono-hydrate, a sesqui-hydrate, a di-hydrate, a tri-hydrate, etc.
[0071] The term "tautomer" as used herein refers to compounds whose structures differ markedly in the arrangement of atoms, but which exist in easy and rapid equilibrium, and it is to be understood that compounds provided herein may be depicted as different tautomers, and when compounds have tautomeric forms, all tautomeric forms are intended to be within the scope of the invention, and the naming of the compounds does not exclude any tautomer. Exemplary tautomerizations include, but are not limited to, amide-to-imide; enamine-to-imine; enamine-to-(a different) enamine tautomerizations; and keto-to-enol.
[0072] The term "stereoisomers" refer to compounds that have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space. Stereoisomers include diastereomers, enantiomers, conformers and the like. [0073] The term "polymorph" refers to a crystalline form of a compound (or a salt, hydrate, or solvate thereof). All polymorphs have the same elemental composition. Different crystalline forms usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optical and electrical properties, stability, and solubility. Recrystallization solvent, rate of crystallization, storage temperature, and other factors may cause one crystal form to dominate. Various polymorphs of a compound can be prepared by crystallization under different conditions.
[0074] As used herein, "isotopically enriched derivatives" refers to a compound containing at least one atom having an isotopic composition other than the natural isotopic composition of that atom. "Isotopic enrichment" can be expressed in terms of the percentage of incorporation of an amount of a specific isotope at a given atom in a molecule in the place of the atom's natural isotopic abundance.
[0075] The bifunctional compound of the present invention may exist in one or more particular geometric, optical, enantiomeric, diasteriomeric, epimeric, atropic, stereoisomeric, tautomeric, conformational, or anomeric forms, including but not limited to, cis- and trans- forms; E- and Z-forms; c-, t-, and r-forms; endo- and exo-forms; R-, S-, and meso-forms; D- and L-forms; d- and I-forms; (+) and (-) forms; keto-, enol-, and enolate-forms; syn- and anti- forms; synclinal- and anticlinal-forms; a- and β-forms; axial and equatorial forms; boat-, chair- , twist-, envelope-, and halfchair-forms; and combinations thereof. [0076] In certain embodiments, the bifunctional compound is selected from the compounds in Table 1, including:
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(12-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-12-oxododecanoyl)piperidine-4-carboxamide (cpd 1); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(2-(2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)ethoxy)ethoxy)ethoxy)ethyl)piperidine-4-carboxamide (cpd 2); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol- 1 -yl)propyl)- 1 -(4-(2-((2-(2,6-dioxopiperidin-3-yl)-l ,3-dioxoisoindolin-4- yl)oxy)acetamido)butanoyl)piperidine-4-carboxamide (cpd 3); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(3-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)ethoxy)ethoxy)propanoyl)piperidine-4-carboxamide (cpd 4); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-3,6,9,12-tetraoxatetradecanoyl)piperidine-4-carboxamide (cpd 5); 4-amino-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- ΙΗ-indol- 1 -yl)propyl)- 1 -(14-((2-(2,6-dioxopiperidin-3-yl)- 1,3- dioxoisoindolin-4-yl)amino)-3,6,9,12-tetraoxatetradecanoyl)piperidine-4-carboxamide (cpd 6);
4-amino-N-(3-(5-((4-(2.6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- ΙΗ-indol- 1 -yl)propyl)- 1 -(1 -((2-(2,6-dioxopiperidin-3-yl)- 1,3- dioxoisoindolin-4-yl)amino)-3,6,9,12-tetraoxapentadecan-15-oyl)piperidine-4- carboxamide (cpd 7);
4-amino-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- 1 H-indol- 1 -y l)propy 1)- 1 -(3-(2-(2-(2-((2-(2,6-dioxopiperidin-3 - yl)- 1 ,3-dioxoisoindolin-4-yl)amino)ethoxy)ethoxy)ethoxy)propanoyl)piperidine-4- carboxamide (cpd 8);
4-amino-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- ΙΗ-indol- 1 -yl)propyl)- 1 -(1 -((2-(2,6-dioxopiperidin-3-yl)- 1,3- dioxoisoindolin-4-yl)amino)-3,6,9,12,15-pentaoxaoctadecan-18-oyl)piperidine-4- carboxamide (cpd 9);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-phenyl-lH-indol-l-yl)propyl)- l-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)amino)-3,6,9,12- tetraoxatetradecanoyl)piperidine-4-carboxamide (cpd 10);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(l-((2-(2,6-dioxopiperidm-3-yl)-l,3-dioxoisoindolin-4- yl)amino>3,6,9,12,15-pentaoxaoctadecan-18-oyl)piperidine-4-carboxamide (cpd 11); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-4-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolm-4- yl)amino)-3,6,9,12-tetraoxatetradecanamido)piperidine-4-carboxamide (cpd 12); N-(4-((3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- 1 H-indol- 1 -y l)propy l)amino)-4-oxobutyl)- 14-((2-(2,6 - dioxopiperidin-3-yl)-l ,3-dioxoisoindolin-4-yl)amino)-3,6,9, 12-tetraoxatetradecanamide (cpd 13);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-3,6,9,12-tetraoxatetradecanoyl)piperidine-3-carboxamide (cpd 14);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(3-(2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)ethoxy)ethox>)ethoxy)propanoyl)piperidine-4-carboxamide (cpd 15);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(l-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-3,6,9.12-tetraoxapentadecan- 15-oyl)piperidine-4-carboxamide (cpd 16);
4-amino-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-
(trifluoromethoxy )pheny 1)- 1 H-indol- 1 -yl)propyl)-l -((R)- 18-((2R,4S)-4-hy droxy-2-((4-(4- methy lthiazol-5-y l)benzy l)carbamoy l)pyrrolidine- 1 -carbony 1)- 19,19-dimethyl- 16-oxo- 4,7,10,13-tetraoxa-17-azaicosanoyl)piperidine-4-carboxamide (cpd 17); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol- 1 -yl)propyl)- 1 -((R)- 18-((2R,4S)-4-hydroxy-2-((4-(4-methylthiazol-5- yl)benzyl)carbamoyl)pyrrohdine-l -carbonyl)- 19.19-dimethyl- 16-oxo-4,7,l 0, 13-tetraoxa- 17-azaicosanoyl)piperidine-4-carboxamide (cpd 18); N-(3-(5-((4-((2,6-dichilorobenzyl)amino)piperidin- 1 -yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- ΙΗ-indol- 1 -yl)propyl)- 1 -(14-((2-(2,6-dioxopiperidin-3-yl)- 1,3- dioxoisoindolin-4-yl)amino)-3,6,9, 12-tetraoxatetradecanoyl)piperidine-4- carboxamide (cpd 19);
N-(3-(5-((4-(2,6-dichlorobenzyl)-2-methylpiperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- ΙΗ-indol- 1 -yl)propyl)- 1 -(14-((2-(2,6-dioxopiperidin-3-yl)- 1,3- dioxoisoindolin-4-yl)amino)-3,6,9,12-tetraoxatetradecanoyl)piperidine-4-carboxamide (cpd 20);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-3,6,9.12-tetraoxatetradecanoyl)piperidine-2-carboxamide (cpd 21); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino>3,6,9,12-tetraoxatetradecanoyl)pyrrolidine-2-carboxamide (cpd 22); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-3,6,9,12-tetraoxatetradecanoyl)-4-methylpiperidine-4-carboxamide (cpd 23); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-4-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-3,6.9,12-tetraoxatetradecanoyl)piperazine-2-carboxamide (cpd 24); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- 1 H-indol- 1 -y l)propyl)- 1 -((S)-l 8-((2S,4R)-4-hy droxy-2-((4-(4-methy lthiazol-5 - yl)benzyl)carbamoyl)pyrrolidine-l -carbonyl)-19, 19-dimethyl- 16-oxo-4,7,l 0, 13-tetraoxa- 17-azaicosan-l-oyl)piperidine-3-carboxamide (cpd 25);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-((S)-18-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5- yl)benzyl)carbamoyl)pynohdine-l -carbonyl)- 19, 19-dimethyl- 16-oxo-4,7,l 0, 13-tetraoxa- 17-azaicosan-l-oyl)piperidine-2-carboxamide (cpd 26);
N-(3-(6-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(l-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-3,6,9, 12-tetraox^entadecan- 15-oyl)piperidine-4-carboxamide (cpd 27); N-(3-(7-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(l-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-3,6,9,12-tetraoxapentadecan-15-oyl)piperidine-4-carboxamide (cpd 28); N-(3-(6-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-((S)-18-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5- yl)benzyl)carbamoyl)pynolidine-l -carbonyl)- 19.19-dimethyl- 16-oxo-4,7,l 0, 13-tetraoxa- 17-azaicosanoyl)piperidine-4-carboxamide (cpd 29); Nl-(4-((3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- 1 H-indol- 1 -yl)propyl)amino)-4-oxobutyl)-N 16-((R)-l - ((2R,4S)-4-hydroxy-2-((4-(4-rnethylthiazol-5-yl)benzyl)carbamoyl)pynOlidin- 1 -yl)-3,3- dimethyl-l-oxobutan-2-yl)-4,7,10,13-tetraoxahexadecanediamide (cpd 30); N-(3-(7-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- 1 H-indol- 1 -y l)propyl)- 1 -((S)-l 8-((2S,4R)-4-hydroxy-2-((4-(4-methy lthiazol-5 - yl)benzyl)carbamoyl)pyrrolidine-l -carbonyl)- 19, 19-dimethyl- 16-oxo-4,7,l 0, 13-tetraoxa- 17-azaicosanoyl)piperidine-4-carboxamide (cpd 31);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(9-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)nonanoyl)piperidine-4-carboxamide (cpd 32);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- 1 H-indol- 1 -yl)propyl)-l -(12-(((R)- 1 -((2R,4S)-4-hydroxy-2-((4-(4-methylthiazol-5- yl)benzyl)carbamoyl)pyrrolidin-l-yl)-3,3-dimethyl-l-oxobutan-2-yl)amino)-12- oxododecanoyl)piperidine-4-carboxamide (cpd 33);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- 1 H-indol- 1 -y l)propyl)- 1 -((S)-l 4-((2S,4R)-4-hy droxy-2-((4-(4-methy lthiazol-5 - yl)benzyl)carbamoyl)pynOlidine-l -carbonyl)- 15, 15-dimethyl-12-oxo-3,6,9-trioxa- 13- azahexadecyl)piperidine-4-carboxamide (cpd 34);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(2-(3-(((S)-l-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5- yl)benzyl)carbamoyl)pyrrohdin- l-yl)-3, 3-dimethyl- 1 -oxobutan-2-yl)amino)-3- oxopropoxy)ethyl)piperidine-4-carboxamide (cpd 35);
N-(3-(5-((4-(2,6-dichilorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(3-(4-(2-(((S)-l-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5- yl)benzyl)carbamoyl)pyrrohdin-l-yl)-3,3-dimethyl-l-oxobutan-2-yl)amino)-2- oxoethyl)piperazin-l-yl)propyl)piperidine-4-carboxamide (cpd 36);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(3-(2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5- yl)amino)ethoxy)ethoxy)ethoxy)propanoyl)piperidine-4-carboxamide (cpd 37);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(l-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5- yl)amino)-3,6,9, 12-tetraoxapentadecan- 15-oyl)piperidine-4-carboxamide (cpd 38); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- ΙΗ-indol- 1 -yl)propyl)- 1 -(1 -((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)amino)- 3, 6, 9, 12-tetraoxapentadecan-15-oyl)piperidine-4- carboxamide (cpd 39); 4-amino-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- ΙΗ-indol- 1 -yl)propyl)- 1 -(1 -((2-(2,6-dioxopiperidin-3-yl)- 1 - oxoisoindolin-4-y l)amino)-3 ,6,9,12-tetraoxapentadecan- 15 -oy l)piperidine-4-carboxamide (cpd 40);
1 -( 1 -(( 1 -(2-chloroacety 1)- 1 ,2,3,4-tetrahydroquinolin-6-yl)oxy)-2-oxo-6,9, 12, 15-tetraoxa-
3-azaoctadecan-18-oyl)-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)-lH-indol-l-yl)propyl)piperidine-4-carboxamide (cpd 41);
1 -( 1 -(( 1 -(2-chloroacety 1)- 1 ,2,3,4-tetrahydroquinolin-6-yl)oxy)-2-oxo-6,9, 12, 15, 18- pentaoxa-3-azahenicosan-21 -oyl)-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l - yl)mefliyl)-3 -(4-(trifluoromethoxy )phenyl)- ΙΗ-indol- 1 -yl)propy l)piperidine-4- carboxamide (cpd 42);
4-amino- 1 -(1 -(( 1 -(2-chloroacety 1)- 1 ,2,3,4-tetrahydroquinolin-6-yl)oxy)-2-oxo-6,9, 12,15- tetraoxa-3-azaoctadecan-18-oyl)-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-
3-(4-(trifluoromethoxy)phenyl)-lH-indol-l-yl)propyl)piperidine-4-carboxamide (cpd 43); 1 -( 1 -(( 1 -(2-chloroacetyl)-l ,2,3,4-tetrahy droquinolin-6-yl)oxy)-2-oxo-6,9, 12-trioxa-3- azapentadecan- 15-oyl)-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin- 1 -yl)methyl)-3-(4- (trifluoromethoxy)phenyl)-lH-indol-l-yl)propyl)piperidine-4-carboxamide (cpd 44);
4-amino- 1 -(1 -(( 1 -(2-chloroacety 1)- 1 ,2,3,4-tetrahydroquinolin-6-yl)oxy)-2-oxo-6,9, 12- trioxa-3-azapentadecan-15-oyl)-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)- 3-(4-(trifluoromethoxy)phenyl)- lH-indol- l-yl)propyl)piperidine-4-carboxamide (cpd 45); and
4-amino-l-(2-((l-(2-chloroacetyl)-l,2,3,4-tetrahydroquinolin-6-yl)oxy)acetyl)-N-(3-(5-
((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)-lH-indol- l-yl)propyl)piperidine-4-carboxamide (cpd 46); or pharmaceutically acceptable salts, solvates, hydrates, polymorphs, tautomers, stereoisomers, and isotopically enriched derivatives, prodrugs thereof.
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
[0077] In certain embodiments, the bifimctional compound as described therein binds RAS protein with a Kn value of less than about 100 μΜ, less than about 50 μΜ, less than about 10 μΜ, less than about 5 μΜ, less than about 1 μΜ, less than about 500 nM, less than about 100 nM, less than about 50 nM. [0078] In certain embodiments, the bifimctional compound as described therein selectively binds mutant RAS protein over wild type RAS protein. In some embodiments, the compound of Formula I selectively binds mutant RAS protein over wild type RAS protein. In certain embodiments, the selectivity is between about 2-fold and about 5-fold. In certain embodiments, the selectivity is between about 5-fold and about 10-fold. In certain embodiments, the selectivity is between about 10-fold and about 20-fold. [0079] In certain embodiments, the bifunctional compound as described therein binds an E3 ubiquitin ligase with a KD value of less than about SO μΜ, less than about 10,000 nM, less than about 5,000 nM, less than about 1,000 nM, less than about 500 nM, less than about 100 nM, less than about 50 nM. [0080] In certain embodiments, the bifunctional compound as described therein promotes the degradation of mutant RAS protein up to 10%, up to 20%, up to 30%, up to 40%, up to 50%, up to 60%, up to 70%, up to 80%, up to 90%, up to 100% at a concentration of 20 μΜ or less, 10 μΜ or less, 5 μΜ or less, 1,000 nM or less, 500 nM or less, 100 nM or less, 50 nM or less, 10 nM or less. Phannaceutical Compositions and Use
[0081] The bifunctional compound as described therein can be therapeutically administered as the neat chemical, but it may be useful to administer the compounds as a pharmaceutical composition or formulation. Thus, the present disclosure provides a pharmaceutical composition comprising a therapeutically effective amount of the bifunctional compound as described therein or pharmaceutically acceptable salts, tautomers, stereoisomers, solvates, hydrates, polymorphs, isotopically enriched derivatives, or prodrugs thereof, and one or more pharmaceutically acceptable excipients.
[0082] The pharmaceutical compositions can be administered in a variety of dosage forms including, but not limited to, a solid dosage form or a liquid dosage form, an oral dosage form, a parenteral dosage form, an intranasal dosage form, a suppository, a lozenge, a troche, buccal, a controlled release dosage form, a pulsed release dosage form, an immediate release dosage form, an intravenous solution, a suspension or combinations thereof. The pharmaceutical compositions can be administered, for example, by oral or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), rectal, vaginal and topical (including buccal and sublingual) administration.
[0083] An "excipient" generally refers to a substance, often an inert substance, added to a pharmacological composition or otherwise used as a vehicle to further facilitate administration of a compound. Examples of excipients include, but are not limited to, inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents, preservatives, effervescent mixtures, and adsorbents. Suitable inert diluents include, but are not limited to, sodium and calcium carbonate, sodium and calcium phosphate, lactose, and the like. Suitable disintegrating agents include, but are not limited to, starches, such as com starch, cross-linked polyvinyl pyrrolidone, agar, alginic acid, or a salt thereof, such as sodium alginate, and the like. Binding agents may include, but are not limited to, magnesium aluminum silicate, starches such as com, wheat or rice starch, gelatin, methylcellulose, sodium caiboxymethylcellulose, polyvinylpyrrolidone, and the like. A lubricating agent, if present, will generally be magnesium stearate and calcium stearate, stearic acid, talc, or hydrogenated vegetable oils. If desired, the tablet may be coated with a material such as glyceryl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract. The compositions can also be formulated as diewable tablets, for example, by using substances such as mannitol in the formulation. [0084] The term "therapeutically effective amount" refers to the amount of the bifunctional compound as described therein, alone or in combination with an anticancer agent which, upon single or multiple dose administration to the subject, provides the desired effect in the subject under treatment
[0085] The bifunctional compound as described therein is effective in the treatment or amelioration of diseases associated with RAS mutation. Degradation of the target protein will occur when the target protein is placed in proximity to the E3 ubiquitin ligase, thus resulting in degradation/inhibition of the effects of the target protein and the control of protein levels. The control of protein levels afforded by the present disclosure provides treatment of a disease state or condition, which is modulated through the target protein by lowering the level of that protein in the cells of a patient. The "diseases associated with RAS mutation" may be cancers, autoimmune diseases, infectious diseases, or blood vessel proliferative disorders. Cancers may be lung cancer (eg., non-small cell lung cancer), colon cancer, colorectal cancer, breast cancer, prostate cancer, liver cancer, pancreatic cancer, bladder cancer, gastric cancer, renal cancer, salivary gland cancer, ovarian cancer, uterine body cancer, cervical cancer, oral cancer, skin cancer, brain cancer, lymphoma, leukemia, biliary tract malignancies, endometrial cancer, cervical cancer, or myeloid leukemia.
[0086] The bifunctional compound as described therein can be administered as the sole active agent, or administered separately, sequentially or together with one or more additional anticancer agents. The term "anticancer agents" as used herein, unless otherwise indicated, refers to agents capable of inhibiting or preventing the growth of neoplasms, or checking the maturation and proliferation of malignant (cancer) cells. Anticancer agents suitable for use in combination with the compounds of formula (I) include, but are not limited to, targeted cancer drugs, such as trastuzumab, ramucirumab, vismodegib, sonidegib, bevacizumab, everolimus, tamoxifen, toremifene, fulvestrant, anastrozole, exemestane, lapatinib, letrozole, pertuzumab, ado-trastuzumab emtansine, palbociclib, cetuximab, panitumumab, ziv-aflibercept, regorafenib, lmatinib mesylate, lanreotide acetate, sunitinib, regorafenib, denosumab, alitretinoin, sorafenib, pazopanib, temsirolimus, everolimus, tretinoin, dasatinib, nilotinib, bosutinib, rituximab, alemtuzumab, ofatumumab, obinutuxumab, ibrutinib, idelalisib, blinatumomab, soragenib, crizotinib, erlotinib, gefitinib, afatinib dimaleate, ceritnib, ramucirumab, nivolumab, pembrolizumab, osimertinib, and necitumumab; an alkylating agent, such as busulfan, chlorambucil, cyclophosphamide, iphosphamide, melphalan, nitrogen mustard, streptozodn, thiotepa, uracil nitrogen mustard, triethylenemelamine, temozolomide, and 2-chloroethyl-3-sarcosinamide-l-nitrosourea (SarCNU); an antibiotic or plant alkaloid, such as actinomycin-D, bleomycin, cryptophydns, daunorubicin, doxorubicin, idarubidn, irinotecan, L-asparaginase, mitomycin-C, mitramycin, navelbine, paclitaxel, docetaxel, topotecan, vinblastine, vincristine, teniposide (VM-26), and etoposide (VP-16); a hormone or steroid, such as 5a-reductase inhibitor, aminoglutethimide, anastrozole, bicalutamide, chlorotrianisene, diethylstilbestrol (DBS), dromostanolone, estramustine, ethinyl estradiol, flutamide, fluoxymesterone, goserelin, hydroxyprogesterone, letrozole, leuprolide, medroxyprogesterone acetate, megestrol acetate, methyl prednisolone, methyltestosterone, mitotane, nilutamide, prednisolone, arzoxifene (SERM-3), tamoxifen, testolactone, testosterone, triamicnolone, and zoladex; a synthetic, such as all-trans retinoic acid, carmustine (BCNU), carboplatin (CBDCA), lomustine (CCNU), ds-diaminedichloroplatinum (cisplatin), dacarbazine, gliadel, hexamethylmelamine, hydroxyurea, levamisole, mitoxantrone, ο,ρ'- dichlorodiphenyldichloroethane (ο,ρ'-DDD) (also known as lysodren or mitotane), oxaliplatin, porfimer sodium, procarbazine, and imatinib mesylate (Gleevec®); an antimetabolite, such as chlorodeoxyadenosine, cytosine arabinoside, 2'-deoxycoformycin, fludarabine phosphate, 5- fluorouracil (5-FU), 5-fluoro-2'-deoxy uridine (5-FUdR), gemcitabine, camptothedn, 6- mercaptopurine, methotrexate, 4-methylthioamphetamine (4-MTA), and thioguanine; and a biologic, such as alpha interferon, BCG (Bacillus Calmette-Guerin), granulocyte colony stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-2, and herceptin. [0087] Furthermore, the present disclosure provides a method for preventing, ameliorating and/or treating diseases associated with RAS mutation in a subject in need thereof comprising administering the bifunctional compound as described therein or a pharmaceutical composition comprising the same, to the subject.
[0088] Furthermore, the present disclosure provides a therapeutically effective amount of the bifunctional compound for manufacturing a medicament for preventing, ameliorating and/or treating a disease associated with RAS mutation in a subject in need thereof.
[0089] In an additional aspect, the present disclosure provides a method of ubiquitinating and degrading a target protein in a cell by contacting the target protein with the bifunctional compound of formula (I) as described herein. [0090] In order for the invention described herein to be more fully understood, the following examples are set forth. The examples described in this application are offered to illustrate the compounds, pharmaceutical compositions, and methods provided herein and are not to be construed in any way as limiting their scope.
EXAMPLES Example 1 [0091] Compounds of formula (I) can be prepared using the synthetic schemes and procedures described in detail below.
Preparation of synthetic intermediates
Figure imgf000044_0001
[0092] Synthesis of tert-butyl-4-((3-(5-((4-(2,6-dichlorobenzyl)piperazin-l - yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- 1H-indol- 1 -yl)propy l)carbamoy l)piperidine- 1 - carboxylate (B)
N,N-Diisopropylethylamine (DIPEA, 294 μL, 1.69 mmol, 2 eq) was added to a solution of l-(tert-butoxycarbonyl)piperidine-4-carboxylic acid (184 mg, 0.8 mmol, 0.95 eq) and (2- (1H-benzotriazol- 1 -y 1)- 1 ,1,3,3-tetramethyluronium hexafluorophosphate(HBTU,385 mg, 0.39 mmol, 1.2 eq) in DMF (8 ml) and stirred for 30 minutes. 3-(5-((4-(2,6- dichlorobenzyl)piperazin- 1 -y l)methy l)-3-(4-(trifluoromethoxy )pheny 1)- 1H-indol- 1 - yl)propan-l -amine (compound A, prepared as the procedures described in Cell, 2017, 168, 878-889) (0.5 g, 0.85mmol, 1 eq) was added and stirred for an additional 4 hours. Upon completion, the reaction was quenched with saturated aqueous NaHCO3 and extracted 3 times with EtOAc. The combined organic layers were washed with brine, dried with MgSO*, concentrated, and the crude material was purified by silica gel chromatography (0 to 5 % MeOH in CH2CI2), to yield tert-butyl 4-((3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)- 3-(4-(trifluoromethoxy)phenyl)- 1H-indol- 1 -yl)propyl)carbamoyl)piperidine- 1 -carboxylate
(564mg, 83 %). 1 H NMR (600 MHz, CDCl3) δ 7.78 (s, 1H), 7.63 (d, J= 8.7 Hz, 2H), 7.41 (m, 1H), 7.33 (m, 6H), 7.16 (m, 1H), 4.25 (m, 2H), 4.00 (m, 4H), 3.80 (s, 2H), 3.5(m, 2H), 3.32 (m, 2H), 2.70 (m, 9H), 2.12 (m, 3H), 1.66 (m, 4H), 1.4 (s, 9H). LC-MS (m/z): [M]+ cald for C41H48C12F3N5O4, 802.76, found 803.2 [0093] Synthesis of N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- 1H-indol- 1 -yl)propyl)piperidine-4-carboxamide (RB-1 )
[0094] To a solution of the compound B (564 mg, 0.7 mmol) in CH2CI2 (1.5 mL) was added TFA (1 mL, 14 mmol). The reaction mixture was stirred at RT for 2 h. The mixture was neutralized with NaHC03(sat) and diluted with CH2CI2. The CH2CI2 was dried with MgSO*, concentrated, and the crude material was purified by silica gel chromatography (0 to 30% MeOH in DCM) to yield N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- 1H-indol- 1 -yl)propyl)piperidine-4-carboxamide(0.4 g, 81%). 1 H NMR (600 MHz, DMSO) δ 9.52 (s, 1H), 8.55 (s, 1H), 8.27 (s, 1H), 8.05 (d, J= 9.0 Hz, 2H), 7.88 (s, 1H), 7.79 (d ,J= 8.64 Hz,2H), 7.65 (d, J= 8.4 Hz, 1H), 7.49 (d ,J= 8.0 Hz, 2H), 7.46
(d, J= 8.28 Hz, , 2H), 7.37 (t, J= 8.04 Hz, 1H), 7.32(d, J= 8.46 Hz, 1H),4.44 (d, J= 4.8 Hz, 2H), 4.26 (t, J= 6.69 Hz, 2H), 3.75 (s, 2H), 3.3 (m, 4H), 3.06 (m, 2H), 3.0 (m, 2H), 2.89(m, 4H), 2.53 (m, 2H), 2.41 (m, 1H), 1.93 (m, 2H), 1.83 (m,2H). 1.70 (m, 2H). LC-MS (m/z): [M]+ cald for C36H40CI2F3N5O2, 702.64, found 702.6 [0095] RB-4, 5, 6, 7, 8, 9, 10, 11, 12 were prepared in an analogous manner to compound RB-1, employing the corresponding materials.
Preparation of Exemplary Compounds
Figure imgf000045_0001
[0096] Synthesis of N-(3-(5-((4-(2,6-didilorobenzyl)piperazin-l-yl)methyl)-3-(4-
(trifluoromethoxy)phenyl)-lH-indol-l-yl)propyl)-l-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3- dioxoisoindolin-4-yl)amino)-3,6,9,12-tetraoxatetradecanoyl)piperidine-4-carboxamide (5) [0097] DIPEA (50 μL, 0.28 mmol, 2 eq) was added to a solution of 14-((2-(2,6- dioxopiperidin-3-yl)-l ,3-dioxoisoindolin-4-yl)aniino)-3,6,9, 12-tetraoxatetradecanoic acid (74 mg, 0.15 mmol, 1 eq) and HBTU (62 mg, 0.16 mmol, 1.2 eq) in DMF (1.5 ml) and stirred for
30 minutes. N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin- 1 -yl)methyl)-3-(4- (trifluoromethoxy)phenyl)-lH-indol-l-yl)propyl)piperidine-4-carboxamide (RB-1) (100 mg, 0.14 mmol) was added and stirred for an additional 1 hour. Upon completion, the reaction was quenched with NaHCO3(aq) and extracted 3 times with EtOAc. The combined organic layers were washed with brine, dried with MgSO*, concentrated, and the crude material was purified by silica gel chromatography (0 to 10 % MeOH in CH2CI2), to yield compound 5 (100 mg, 59 %). 1H NMR (600 MHz, DMSO-d6) δ 11.11 (s, 1H), 9.28 (s, 1H), 8.03 (s, 1H), 7.92 (t, J = 5.6
Hz, 1H), 7.88 (s, 1H), 7.81 - 7.76 (m, 1H), 7.63 (d, J= 8.4 Hz, 1H), 7.60 - 7.55 (m, 1H), 7.47 (dd, J= 14.7, 8.1 Hz, 3H), 7.38 - 7.34 (m, 1H), 7.31 (d, J= 8.4 Hz, 1H), 7.14 (d, J= 8.6 Hz, 1H), 7.04 (d, J= 7.0 Hz, 1H), 6.60 (t, J= 5.9 Hz, 1H), 5.06 (dd, J= 12.8, 5.4 Hz, 1H), 4.43 (s, 1H), 4.32 - 4.22 (m, 2H), 3.76 (d, J= 14.0 Hz, 2H), 3.61 (t, J= 5.5 Hz, 1H), 3.57 - 3.44 (m, 10H), 3.08 - 2.84 (m, 6H), 2.34 (tt, J= 11.4, 3.8 Hz, 1H), 2.02 (ddq, J= 10.9, 5.3, 2.7, 2.2 Hz,
1H), 1.93 (p, J= 6.9 Hz, 1H), 1.68 (d, J= 13.1 Hz, 1H), 1.50 (q, J= 11.9 Hz, 1H), 1.36 (p, J= 10.0 Hz, 1H). LC-MS (m/z): [M]+ cald for C59H67CI2F3N8O11, 1192.13, found 1193.47.
[0098] Example compounds 1-4, 6-16, 19-24, 27-28, 32, 37-40 were prepared in an analogous manner to compound 5, employing the corresponding amine starting materials, like RB-1, and carboxylic acid, like L.
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Example 2
[0099] Synthesis of compound 17
Figure imgf000058_0001
RB-2 (44 mg, 0.063 mmol) was treated with HBTU (40 mg, 0.10 mmol) in DMF (4 mL) at 0°C. After DIPEA (24 uL, 0.14 mmol) was added, the solution was stirred for 30 min at room temperature. Compound M ((R)- 18-((2R,4S)-4-hydroxy-2-((4-(4-methylthiazol-5- y l)benzy l)carbamoy l)py rrolidine- 1 -carbonyl)- 19, 19-dimethy 1-16-oxo-4,7, 10,13-tetraoxa- 17- azaicosanoic acid) (50 mg, 0.070 mmol) was added, then tiie resulting mixture was stirred for 2 h at room temperature. Water was added to the solution and the solution was extracted with EA. The organic layer was collected, dried with MgSCH, then concentrated. The crude product was purified by column chromatography (CH2Cl2:MeOH, 100:0 to 90: 10) to yield compound
17 (4-amino-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)-lH-indol-l -yl)propyl)-l-((R)-18-((2R,4S)-4-hydroxy-2-((4-(4- methylthiazol-5-yl)ben2yl)carbamoyl)pyrrolidine-l -carbonyl)-! 9, 19-dimethyl- 16-oxo- 4,7, 10, 13-tetraoxa- 17-azaicosanoyl)piperidine-4-carboxamide) (20 mg, 23%) as a white solid. LC-MS (m/z): [M] + calcd for C70H89CI2F3N10O11S, 1406.50, found 1406.64; 1H NMR (600 MHz, DMSO) δ 8.97 (s, 1H), 8.56 (t , J= 6.0 Hz, 1H), 7.90 (d , J= 9.4 Hz, 1H), 7.79 (s, 1H), 7.75 (d, J= 8.6 Hz, 3H), 7.48 (d, J= 8.1 Hz, 1H), 7.45 - 7.39 (m, 6H), 7.38 (d, J= 8.2 Hz, 2H), 7.34 - 7.28 (m, 2H), 7.20 (s, 1H), 5.14 - 5.11 (m, 1H), 4.54 (d, J= 9.4 Hz, 2H), 4.43 (td, J= 8.3, 7.5, 4.1 Hz, 3H), 4.34 (s, 2H), 4.21 (q ,J= 7.7, 6.7 Hz, 4H), 4.10 (q, J= 5.2 Hz, 2H), 3.66 (s, 4H), 3.64 - 3.57 (m, 6H), 3.47 (t, J= 3.8 Hz, 12H), 3.17 (d, J= 5.2 Hz, 2H), 3.12 (d,
J= 6.3 Hz, 3H), 3.00 (s, 2H), 2.55 (d, J= 6.6 Hz, 3H), 2.44 (s, 3H), 2.05 - 1.85 (m, 8H), 1.23 (s, 2H), 0.92 (s, 9H). Example 3
[00100] Synthesis of 34.
Figure imgf000059_0001
[00101] Step 1
After tot-butyl 3-(2-(2-(2-(tosyloxy)ethoxy)ethoxy)e1hoxy)propanoate (135 mg, 0.3 mmol, compound O) and RB-1 (20 mg, 0.3 mmol) were dissolved in DMF (2 mL), cesium carbonate (185 mg, 0.57 mmol) was added, and the mixture was stirred at rt for 16 hours. Upon completion, the reaction was quenched with water and extracted 3 times with EtOAc. The combined organic layers were washed with brine, dried with MgSO*, concentrated, and the crude material was purified by silica gel chromatography (0 to 10 % MeOH in CH2CI2), to yield tert-butyl 3-(2-(2-(2-(4-((3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)-lH-indol-l-yl)propyl)carbamoyl)piperidin-l- yl)ethoxy)ethoxy)ethoxy)propanoate (90 mg, 33 %). 1H NMR (600 MHz, Chloroform-d) δ 7.77 (dd, J= 1.5, 0.7 Hz, 1H), 7.67 - 7.64 (m, 2H), 7.34 - 7.29 (m, 7H), 7.14 (dd, J= 8.4, 7.7 Hz, 1H), 5.44 (s, 1H), 4.23 (t, /= 6.7 Hz, 2H), 3.77 (s, 2H), 3.72 (t , J= 6.6 Hz, 2H), 3.67 - 3.59 (m, 12H), 3.33 (q, J= 6.5 Hz, 2H), 2.98 - 2.93 (m, 2H), 2.65 (bs, 4H), 2.58 (t, J= 5.9 Hz, 2H), 2.52 (t ,J= 6.6 Hz, 3H), 2.20 (s, 10H), 2.11 (p,J= 6.7 Hz, 2H), 2.03 - 1.91 (m, 3H), 1.89 - 1.58 (m, 11H), 1.46 (s, 9H).LC-MS (m/z): [M]+ cald for C49H64CI2F3N5O7, 962.97, found 964.07. [00102] Step 2
To a solution of the compound tert-butyl 3-(2-(2-(2-(4-((3-(5-((4-(2,6- dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- ΙΗ-indol- 1 - yl)propyl)carbamoyl)piperidin- 1 -yl)ethoxy)ethoxy)ethoxy )propanoate (106 mg, 0.1 mmol) in CH2CI2 (0.5 mL) was added TFA(0.5 mL, 6.6 mmol). The reaction mixture was stirred at RT for 2 h. The mixture was neutralized withNaHCOsiaq) and diluted with CH2CI2. The CH2CI2 phase was dried with MgS04, concentrated to provide the product 3-(2-(2-(2-(4-((3-(5-((4-(2,6- dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- ΙΗ-indol- 1 - yl)propyl)carbamoyl)piperidin- 1 -yl)ethoxy)ethoxy)ethoxy)propanoic add (compound N, 100 mg) for tiie next step without purification. LC-MS (m/z): [M]+ cald for C45H56CI2F3N5O7, 906.87, found 906.7
[00103] Step 3
3-(2-(2-(2-(4-((3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)metiiyl)-3-(4- (trifluoromethoxy)phenyl)-lH-indol-l-yl)propyl)carbamoyl)piperidin-l- yl)ethoxy)ethoxy)ethoxy)propanoic add (67 mg, 0.074 mmol) was treated with HBTU (28 mg, 0.074 mmol) in DMF (4 mL) at 0°C. After DIPEA (23 uL, 0.13 mmol) was added, and the solution was stirred for 30 min at room temperature. VHL.HC1 (29 mg, 0.067 mmol) was added, then the resulting mixture was stirred for 2 h at room temperature. Water was added to the solution and the solution was extracted with EA The organic layer was collected, dried with MgSO*, then concentrated. The crude product was purified by column chromatography (CmCkMeOH, 100:0 to 90:10) to yield compound 34, N-(3-(5-((4-(2,6- dichlorobenzy l)piperazin- 1 -yl)methyl)-3 -(4- (trifluoromethoxy)phenyl)- 1 H-indol- 1 - yl)propyl)-l-((S)-14-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5- yl)benzyl)carbamoyl)py rrolidine- 1 -carbonyl)- 15, 15 -dimethyl- 12-oxo-3 ,6,9-trioxa- 13- azahexadecyl)piperidine-4-carboxamide (27 mg, 28%). ¾ NMR (600 MHz, DMSO-de) δ 8.98 (s, 1H), 8.59 (t, J= 6.1 Hz, 1H), 7.93 (d, J= 9.4 Hz, 1H), 7.82 (t ,J= 5.6 Hz, 1H), 7.79 - 7.73
(m, 3H), 7.72 (d, J= 1.5 Hz, 1H), 7.50 - 7.36 (m, 10H), 7.32 (dd, J= 8.5, 7.6 Hz, 1H), 7.17 (dd, J= 8.5, 1.5 Hz, 1H), 5.15 (d, J= 3.5 Hz, 1H), 4.55 (d, J= 9.4 Hz, 1H), 4.47 - 4.40 (m, 2H), 4.35 (s, 1H), 4.21 (td, J= 8.7, 7.3, 4.0 Hz, 3H), 3.70 - 3.54 (m, 9H), 3.53 - 3.44 (m, 11H), 3.05 (q, J= 6.5 Hz, 2H), 2.89 (d, J= 10.8 Hz, 2H), 2.44 (s, 6H), 2.35 (dt, J= 14.6, 6.1 Hz, 3H), 2.08 -2.01 (m, 2H), 1.95 - 1.87 (m, 4H), 1.62 (d,J= 11.2 Hz, 2H), 1.59 - 1.50 (m, 2H), 0.93 (s, 9H). LC-MS (m/z): [M]+ cald for C67H84CI2F3N9O9S, 1319.42, found 1319.44.
[00104] Example compounds 18, 25-26, 29-31, 33, 35 were prepared in an analogous manner to compound 17 or 34, employing the corresponding amine starting materials, like RB- 1 or RB-2, and carboxylic acid, like compound M, or tosyloxy tert-butyl ester, like compound
O.
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Example 4
[00105] Synthesis of compound 43
Figure imgf000065_0001
l-((l-(2-cchoroacetyl)-l,2,3,4-tetrahydroquinolin-6-yl)oxy)-2-oxo-6,9,12,15-tetraoxa-3- azaoctadecan-18-oic acid(Compound Ρ, prepared as the procedures described in Nature Chemical Biology, VOL 15, 2019, 7379 15X108 mg, 0.204 mmol) was added with HBTU (89 mg, 0.3234 mmol), DMF (2.0 mL) and DIPEA (40mg, 0.306 mmol), and the solution was stirred under Ar atmosphere for 15 min at 25°C. Then,RB-2 (146 mg, 0.204 mmol)was added, then the resulting mixture was stirred for 2 h at 25°C.The reaction mixture was diluted to 10 mL with acetonitrile and purified via preparative HPLC and lyophilized to provide the title compound (61.0 mg, 24%) as a white TFA salt. LC-MS (m/z): [M]+ calcd for
C60H74CI3F3N8O10, 1230.64, found 1229.63. 1H NMR (600 MHz, DMSO-d6) δ 9.65 (s, 1H), 8.47 (t , J= 5.6 Hz, 1H), 8.41 (s, 3H), 8.07 (t , J= 6.0 Hz, 1H), 8.03 (s, 1H), 7.87 (s, 1H), 7.80 - 7.74 (m, 2H), 7.63 (d , J= 8.5 Hz, 1H), 7.48 (d, J= 8.0 Hz, 2H), 7.45 (d, J= 8.2 Hz, 2H), 7.39 - 7.33 (m, 1H), 7.31 (d, j= 8.0 Hz, 1H), 6.78 (m, 2H), 4.44 (m, 5H), 4.26 (t, J= 6.9 Hz, 2H), 4.15 (m, 1H), 3.79 - 3.72 (m, 12H), 3.67 - 3.59 (m, 10H), 3.43 (t, J= 6.0 Hz, 3H), 3.28
(m, 4H), 3.17 (q, J = 6.6 Hz, 2H), 3.01 (m, 3H), 2.92 (m, 2H), 2.66 (m, 3H), 2.16 - 2.09 (m, 1H), 2.08 - 2.03 (m, 1H), 1.97 (p, J= 6.9 Hz, 2H), 1.87 (s, 2H), 1.73 (t,J= 15.1 Hz, 2H).
[00106] Example compounds 41, 42, 44, 45, 47 were prepared in an analogous manner to compound 43, employing the corresponding amine starting materials, like RB-2, and carboxylic acid, like compound P.
Figure imgf000065_0002
Figure imgf000066_0001
Figure imgf000067_0001
Example 5
Biological activity
[00107] Various of RAS-PROTACs were tested for their specificity and abilities for degrading the targeted protein. Brief descriptions of different assays are described below.
Western blot to evaluate the cellular potencies of RAS-PROTACs in KRAS protein degradation
[00108] RAS genes (KRAS, NRAS and HRAS) are the most frequently mutated oncogenes in human cancer. Its gain-of-fimction mutations occur in approximately 30% of all human cancers. As the most frequently mutated RAS isoform, mutated KRAS is appearing in 90% of pancreatic ductal adenocarcinoma, 30% of lung adenocarcinoma and in 40% of colorectal adenocarcinoma. These high occurrences make KRAS one of the most important targets in oncology for drug development.
[00109] RAS is a small GTPase transductor protein that initiates the activation of downstream signaling molecules, allowing the transmission of transducing signals from the cell surface to the nucleus, and plays a key role in essential cellular processes such as cell differentiation, growth, chemotaxis and apoptosis. The RASG12C mutant has a cysteine residue that has been exploited to design covalent inhibitors. Preclinical data have shown that the KRASG12C inhibitor blocks tumor growth for G12C KRAS gene mutations. This led to KRAS as a promising therapeutic target for cancer treatment. However, the KRASG12C inhibitor can’t block tumor growth for other G12 and G13 KRAS gene mutations.
[00110] In the current invention, a RAS-PROTAC refers to a PROTAC that includes a target binder for the RAS protein. Various compounds of RAS-PROTACs were tested in cellular RAS protein degradation via western blotting assay. The following uses RAS- PROTACs to illustrate the benefits of embodiments of the invention. [00111] For western blot experiments, BT-474(KRASWT) and MIA PaCa-2(KRASG12C) cells were cultured in DMEM medium with 10% FBS. BxPC3 (KRASWT) and KLM-1 (KRASG12D) cells were cultured in DMEM medium with 10% FBS. MDA-MB-231 (KRASG13D) cells were cultured in L15 medium with 10% FBS. BT-474, MIA PaCa-2, BxPC3 and MDA- MB-231 were established from the American Type Culture Collection. KLM-1 were established from the ExPASy. On the assay day, two hundred thousand cells were pretreated with each of the test compounds for 24 hours. After 24 hours, the whole cell lysate was harvested by adding 2x SDS Sample Buffer. Proteins were separated by SDS-PAGE electrophoresis and transferred to PVDF membrane. Protein expression was detected using immunoblot with various primary antibodies and secondary antibodies following standard protocols. Antibody against KRAS was purchased from Abeam (Cambridge, UK). Anti-rabbit IgG, HRP-linked secondary antibodies was purchased from Cell Signaling Technology (Danvers, MA). Antibody against actin was purchased from Millipore (Burlington, MA). Immunoblots were revealed by chemiluminescence (SuperSignal™ West Femto Maximum Sensitivity Substrate, Thermo Fisher, Waltham, MA) and detected by ChemiDoc™ MP Imaging System (Bio-Rad, Hercules, CA). Band intensities of western blot were also quantified by ChemiDoc™ MP Imaging System. Relative intensities of bands corresponding to the drug treatment group were compared to those of the untreated group.
[00112] FIG. 1 shows the results of the assay. Compounds 5, 10, 11, 12 and 16 are hetero-bifunctional molecule comprising a RAS binding moiety linked to a Cereblon E3 ligase binding moiety. As shown in FIG. 1A, compounds 5, 11, 12 and 16 are effective in promoting the degradation of KRASG13D in breast cancer cell line MDA-MB-231 (KRASG13D). FIG. IB shows that compounds 5, 11, 12 and 16 are effective in promoting the degradation of KRASG12D in pancreatic cancer cell line KLM-1 (KRASG12D). [00113] FIG. 2 shows that compound 5 exhibits specific KRAS mutation protein degradation activity in cancer cell lines with mutated KRAS gene such as MDA-MB-231, KLM-1, and MiaPaCa-2, instead of cell lines without mutated KRAS gene such as BT474 and BxPC-3. In comparison with KRASG12C, a covalent inhibitor can only target KRASG12C, and compound 5 can not only promote the degradation of KRASG12C in MiaPaCa-2 cells but also KRASG12D in KLM-1 cells and KRASG13D in MDA-MB-231 cells. Compound 5 does not cause degradation of either AKT protein or actin protein.
[00114] FIG. 3A shows that compound 7 exhibits specific KRAS mutation protein degradation activity in cancer cell lines Calu-1 (KRASG12C) and Mi A PaCa-2 (KRASG12C), instead of cell lines without mutated KRAS gene such as BT474 (KRASWT). FIG. 3B shows that compound 43 exhibits specific KRAS mutation protein degradation activity in cancer cell lines MDA-MB-231 (KRASG13D), KLM-1 (KRASG12D), Calu-1 (KRASG12C) and MiA PaCa-2 (KRASG12C), instead of cell lines without mutated KRAS gene such as BT474 (KRAS*7). [00115] The primary screening of bifimctional compounds 1-46 degraded KRAS protein in MDA-MB-231 cell line were as shown in Table 2 which tested under the conditions described above, and some of them were took further degradation test at 1 uM and 0.3 μΜ as shown in Table 3. Table 4 showed the degradation of compounds in KLM-1 cell line, and Table 5 showed the degradation at 1 μΜ and 0.3 μΜ. Table 6 showed the degradation of compounds in MIA PaCa-2 cell line, and Table 7 showed the degradation of compounds in Calu-1 cell line. In Tables 2 and 7, “ND” means not determined, “A” means >70%, B means in the range of
35-70%, C means <35%, means no degradation, means actin remains < 70%.
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000070_0002
Figure imgf000071_0001
Figure imgf000071_0002
Figure imgf000071_0003
Figure imgf000072_0001
Figure imgf000072_0002
Figure imgf000072_0003
Figure imgf000073_0002
Comparative examples:
[00116] Two RAS binders, abd-7 and CH2, were reported in NATURE
COMMUNICATIONS (2018) 9:3169, 1~12 and PNAS, 2019, vol. 116, no. 7, 2545-2550, respectively. These two binders were conjugated with linker-E3 ligase ligand to form PROTAC molecules, as Comparative examples 1~4. Further, the degradation of Comparative examples 1~4 in the Calu-1 cell line was measured and the results are shown in Table 8 below, which showed almost no degradation effect. This shows that not all of the PROTAC molecules formed by any of the RAS binders are effective in degrading RAS protein, as revealed by WO20 18092723 A1.
Figure imgf000073_0001
Comparative example 2
Figure imgf000074_0001
Comparative example 3
Figure imgf000074_0002
Comparative example 4
Example 6
Figure imgf000074_0003
Anti-proliferation activity
[00117] As noted above, RAS-PROTACs of the invention may be used to treat diseases or disorders harboring a specific RAS mutation The diseases may be cancers, autoimmune diseases, infectious diseases, or blood vessel proliferative disorders. The cancers may be lung cancer (e.g., non-small cell lung cancer), colon cancer, colorectal cancer, breast cancer, prostate cancer, liver cancer, pancreatic cancer, bladder cancer, gastric cancer, renal cancer, salivary gland cancer, ovarian cancer, uterine body cancer, cervical cancer, oral cancer, skin cancer, brain cancer, lymphoma, leukemia, biliary tract malignancies, endometrial cancer, cervical cancer, or myeloid leukemia. Inhibitions of cell growth by RAS-PROTACs of the invention were measured using CellTiter™-96 assay. The cytotoxicities of RAS-PROTACs were evaluated in lung cancer cell lines, breast cancer cell lines and pancreatic cancer cell lines with different RAS mutations. The results given in Table 9 show that RAS-PROTACs of the present invention are more toxic to cancer cells with KRAS mutations. Table 10 shows the ICso of compound 5, 7, 30, 37, 39, 41, 43, 46 in the Calu-1, MIA PaCa-2 and HCT-116 cell lines. 00118] Table 9
Figure imgf000075_0001
Table 10
Figure imgf000075_0002
Figure imgf000076_0001
Example 7
Xenograft model of Ras-PROTAC (Lung cancer)
[00119] The aim of this study was to evaluate the in vivo anti-tumor efficacy of Ras- PROTAC in Calu-1 human lung cancer xenograft model in male NOD SCID mice. [00120] Test articles Cpd 5, Cpd 7, Cpd 39 and corresponding vehicle were formulated and given intra-tumor (IT) injection to mice once daily for consecutive 14 days. The Calu-1 cells were maintained in vitro as a monolayer culture in RPMI-1640 medium supplemented with 10% fetal bovine serum at 37°C in an atmosphere of 5% CO2 in air. The tumor cells were routinely sub-cultured twice weekly by trypsin-EDTA treatment. The cells growing in an exponential growth phase were harvested and counted for tumor inoculation. Male NOD SCID mice at age of 6-7 weeks were purchased from BioLasco Taiwan Co., LTD. and quarantined for one week. Five mice were housed in each cage. All animals were hosted in the animal facility with a 12-h light/12-h dark cycle at 19-25°C. Animals had free access to rodent pellet foods and water ad libitum. Calu-1 cells were subcutaneously (SC) implanted (5 x 106 cells in 1:1 PBS/matrigel mixture at 0.1 mL per mouse) into the right flank of male NOD SCID mice.
When tiie average tumor volume had reached 170 mm3, the mice were randomly divided into 5 groups (N=5-7 per group). Vehicle, Cpd 5 (10 mg/kg), Cpd 7 (10 mg/kg), and Cpd 39 (10 mg/kg) were given intra-tumor (IT) injection once daily for consecutive 14 days. The tumor volumes, body weights, mortality, and signs of overt toxicity were monitored and recorded three times weekly for 28 days. Tumor volumes (mm3) were measured three times per week using calipers and calculated according to the formula: Tumor Volume = (w2 x/)/2, where w = width and l = length in diameter (mm) of the tumor. The percentages of tumor growth inhibition (TGI) were calculated using the following formula: %TGI = [1 - (T/C)] x 100%, where T and C represent the mean tumor volumes of the treatment group and the control group, respectively. One-way ANOVA followed by Dunnett’s test was applied for comparison between the vehicle and test article-treated groups. Differences are considered significant at *P<0.05. Animals were weighed three times weekly until the completion of the study. The body weight changes were calculated as percentage increases in the body weights, as compared with the initial body weights. [00121] FIG. 4A showed tumor growth curve in Calu-1 implanted male NOD SCID mice. Test articles Cpd 5 (10 mg/kg), Cpd 7 (10 mg/kg), and Cpd 39 (10 mg/kg) were given intra-tumor (IT) injection to mice once daily for consecutive 14 days. One-way ANOVA followed by Dunnett’s test was applied for comparison between the vehicle and test article- treated groups. Differences are considered significant at *P<0.05. Cpd 5 at 10 mg/kg showed low anti-tumor activity, with a TGI value of 25% (D28). Cpd 7 at 10 mg/kg significantly reduced Calu-1 tumor growth from Day 8 to Day 28, with the TGI values ranged from 38% to 55%, and was considered as moderate efficacious to Calu-1 xenografts. Cpd 39 at 10 mg/kg significantly reduced Calu-1 tumor growth from Day 8 to Day 28, with the TGI values ranged from 23% to 52%, and was considered as moderate efficacious to Calu-1 xenografts.
[00122] FIG. 4B showed body weight changes in Calu-1 implanted male NOD SCID mice. Test articles Cpd 5 (10 mg/kg), Cpd 7 (10 mg/kg), and Cpd 39 (10 mg/kg) were given intra-tumor (IT) injection to mice once daily for consecutive 14 days. No body weight loss was observed throughout tire experiment.
Example 8
[00123] The aim of this study was to evaluate the in vivo anti-tumor efficacy of Ras- PROTAC in Calu-1 human lung cancer xenograft model in male NOD SCID mice. Test articles Cpd 7 (3 mg/kg and 10 mg/kg), Cpd 43 (10 mg/kg), and corresponding vehicle were formulated and given intra-tumor (IT) injection to mice once daily for consecutive 28 days. Cpd 37 at 5 mg/kg was given intra-tumor (IT) injection to mice once daily for consecutive 14 days. The Calu-1 cells were maintained in vitro as a monolayer culture in RPMI-1640 medium supplemented with 10% fetal bovine serum at 37°C in an atmosphere of 5% CO2 in air. The tumor cells were routinely sub-cultured twice weekly by trypsin-EDTA treatment. The cells growing in an exponential growth phase were harvested and counted for tumor inoculation. Male NOD SCID mice at age of 6-7 weeks were purchased from BioLasco Taiwan Co., LTD. and quarantined for one week. Five mice were housed in each cage. All animals were hosted in the animal facility with a 12-h light/ 12-h dark cycle at 19-25°C. Animals had free access to rodent pellet foods and water ad libitum Calu-1 cells were subcutaneously (SC) implanted (5 x 106 cells in 1:1 PBS/matrigel mixture at 0.1 mL per mouse) into the right flank of male
NOD SCID mice. When the average tumor volume had reached 160 mm3, the mice were randomly divided into 5 groups (N=5-6 per group). Vehicle, Cpd 7 (3 mg/kg and 10 mg/kg), Cpd 43 (10 mg/kg) were given intra-tumor (IT) injection once daily for consecutive 28 days. Cpd 37 at 5 mg/kg was given intra-tumor (IT) injection once daily for consecutive 14 days. The tumor volumes, body weights, mortality, and signs of overt toxicity were monitored and recorded three times weekly for 28 days. Tumor volumes were measured three times per week using calipers and calculated according to the formula: Tumor Volume = (w2 x/)/2, where w = width and l = length in diameter (mm) of the tumor. The percentages of tumor growth inhibition (TGI) were calculated using the following formula: %TGI = [1 - (T/C)] χ 100%, where T and C represent the mean tumor volumes of the treatment group and the control group, respectively. One-way ANOVA followed by Dunnett’s test was applied for comparison between the vehicle and test article-treated groups. Differences are considered significant at *P<0.05. Animals were weighed three times weekly until the completion of the study. The body weight changes were calculated as percentage increases in the body weights, as compared with the initial body weights.
[00124] FIG. 5A showed tumor growth curve in Calu-1 implanted male NOD SCID mice. Test articles Cpd 7 (3 and 10 mg/kg), and Cpd 43 (10 mg/kg) were given intra-tumor injection to mice once daily for consecutive 28 days. Cpd 37 at 5 mg/kg was given intra-tumor injection to mice once daily for consecutive 14 days. One-way ANOVA followed by Dunnett’s test was applied for comparison between the vehicle and test article-treated groups. Differences are considered significant at *P<0.05. Cpd 7 at 3 mg/kg showed low anti-tumor activity, with a TGI value of 24% (D28). Cpd 7 at 10 mg/kg significantly reduced Calu-1 tumor growth, with a TGI value of 51% (D28). Cpd 37 at 5 mg/kg showed low anti-tumor activity against Calu-1 xenografts, with a TGI value of 37% (D28). Cpd 43 at 10 mg/kg significantly reduced Calu-1 tumor growth from Day 2 to Day 28, with the TGI values ranged from 50% to 86%, and was considered as pronounced efficacious to Calu-1 xenografts. [00125] FIG. 5B showed body weight changes in Calu-1 implanted male NOD SCID mice. Test articles Cpd 7 (3 and 10 mg/kg), and Cpd 43 (10 mg/kg) were given intra-tumor injection to mice once daily for consecutive 28 days. Cpd 37 at 5 mg/kg was given intra-tumor injection to mice once daily for consecutive 14 days. No body weight loss was observed among the treatment groups. [00126] The RAS-PROTACs of the invention are promising new therapeutics for cancer patients with RAS mutation.
[00127] This application refers to various issued patents, published patent applications, journal articles, and other publications, all of which are incorporated herein by reference. If there is a conflict between any of the incorporated references and the instant specification, the specification shall control. In addition, any particular embodiment of the presort invention that falls within the prior art may be explicitly excluded from any one or more of the claims. Because such embodiments are deemed to be known to one of ordinary skill in the art, they may be excluded, even if the exclusion is not set forth explicitly herein. Ary particular embodiment of the invention can be excluded from any claim, for any reason, whether or not related to the existence of prior art.
[00128] Those skilled in the art will recognize or be able to ascertain using no more than routine experimentation many equivalents to the specific embodiments described herein. The scope of the present embodiments described herein is not intended to be limited to the above description, but rather is as set forth in the appended claims. Those of ordinary skill in the art will appreciate that various changes and modifications to this description may be made without departing from the spirit or scope of tire present invention, as defined in the following claims.

Claims

CLAIMS What is claimed is:
1. A bifunctional compound of formula (I):
RB— Linker— ULM (I) or pharmaceutically acceptable salts, solvates, hydrates, polymorphs, tautomers, stereoisomers, isotopically enriched derivatives, prodrugs thereof, wherein:
RB is a RAS protein binding moiety;
ULM is an E3 ubiquitin ligase binding moiety; and Linker is a group covalently binding to the RB and ULM moieties; wherein RB is represented by formula RB-I:
Figure imgf000079_0001
wherein:
An is phenylene substituted with one or more substituents selected from the group consisting of CM alkyl, CM alkoxy, OH, NH2, CN, OCF3, and halide;
Ar2 is aryl substituted with one to three groups selected from the group consisting of H, CM alkyl, CM alkoxy, OH, NH2, CN, OCF3, and halide;
Ri is a bond, C1-6 alkylene, C3-8 cycloalkylene, or C1-6 heteroalkylene comprising 1 to 3 heteroatoms selected from 0 and S;
R2 is H or CM alkyl; is selected from the group consisting of none, amine,
Figure imgf000079_0003
Figure imgf000079_0002
Figure imgf000079_0004
t is an integer of 1~10;
R3 is H, halide, or CH3; and the dashed lme indicates the site of attachment to Linker.
2. The bifunctional compound according to claim 1 , wherein in the formula of RB-I:
Ar1 is phenylene substituted with one or two substituents selected from the group consisting ofOMe, NH2, OCF3, F, or Cl;
Arz is aryl substituted with one to three groups selected from the group consisting of H, CH3, OMe, NH2, CN, F, or Cl;
R1 is C2-4 alkylene, C3-6 cycloalkylene, or CM heteroalkylene comprising a heteroatom selected from O and S;
R2 is H or C1-3 alkyl;
Figure imgf000080_0001
t is 1~4;
R3 is H, F, or CH3; and tiie dashed line indicates the site of attachment to Linker.
3. The bifunctional compound according to claim 1 , wherein the formula RB-I is selected for tiie group consisting of:
Figure imgf000080_0002
Figure imgf000081_0001
wherein the dashed line indicates the site of attachment to Linker.
4. The bifunctional compound according to claim 1 , wherein ULM is a Cereblon E3 ubiquitin ligase binding moiety (CLM), a Von Hippel-Lindau (VHL) E3 ubiquitin ligase binding moiety (VLM), a DDB1- and CUL4-assodated factor 16 (DCAF16) E3 ubiquitin ligase binding moiety (DLM), an IAP E3 ubiquitin ligase binding moiety (ILM), or a mouse double minute 2 (MDM2) homolog E3 ubiquitin ligase binding moiety (MLM), or a Kelch- like ECH-associated protein-1 (KEAP1) E3 ubiquitin ligase binding moiety, or a DCAF15 E3 ubiquitin ligase binding moiety, or an RNF4 E3 ubiquitin ligase binding moiety, or an RNF114 E3 ubiquitin ligase binding moiety, or an aiylhydrocarbon receptor (AhR) E3 ubiquitin ligase binding moiety.
5. The bifunctional compound according to claim 4, wherein ULM is the CLM selected from tiie group consisting of a thalidomide, lenalidomide, pomalidomide, analogs thereof, or derivatives thereof.
6. The bifunctional compound according to claim 4, wherein the CLM has a chemical structure represented by:
Figure imgf000082_0001
wherein:
W is selected from CH2 and C=O; and
Q1, Q2 Q3, and Q4 are each independently C or N, and wherein one of Q1, Q2, Q3, and Q4 covalently join to Linker.
7. The bifunctional compound according to claim 6, wherein the CLM is selected from the group consisting of:
Figure imgf000082_0002
wherein the dashed line indicates the site of attachment to Linker.
8. The bifunctional compound according to claim 4, wherein ULM is the VLM with a chemical structure represented by:
Figure imgf000082_0003
wherein:
R4 is H, ethyl, isopropyl, tert-butyl, sec-butyl, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl;
Rs is H, haloalkyl, substituted or unsubstituted alkyl, or substituted or unsubstituted cycloalkyl;
Re is selected from the group consisting of H, halide, CN, OH, substituted or unsubstituted heteroaiyl, substituted or unsubstituted aryl, substituted or unsubstituted alkyl, substituted or unsubstituted haloalkyl, substituted or unsubstituted haloalkoxy, substituted or unsubstituted cycloalkyl, and substituted or unsubstituted heterocycloalkyl; and the dashed line indicates the site of attachment to Linker.
9. The bifunctional compound according to claim 8, wherein: R4 is isopropyl, tert-butyl, cyclopropyl, or cyclobutyl.
10. The bifunctional compound according to claim 8, wherein: Rs is H, methyl, fluoromethyl, or cyclopropyl.
11. The bifunctional compound according to claim 4, wherein ULM is the DLM with a chemical structure represented by:
Figure imgf000083_0001
wherein:
R7 is H, halide, C1-4 alkyl, or C1-4 alkoxy;
Y is O, or S, orNH;
Rm is a covalent electrophile, and is selected from
Figure imgf000083_0002
the dashed line linked to Y indicates the site of attachment to Linker.
12. The bifunctional compound according to claim 1, wherein Linker covalently couples RB to ULM.
13. The bifunctional compound according to claim 1, wherein Linker is -A1... Aq-; Ai to Aq are each independently selected from the group consisting of a bond, CRL1RL2, O,
S, S=O, S(=O)2, NRL3, C(=O), C(=O)NRL3, NRL3C(=O), C≡C, C3-11 cycloalkyl substituted with 0-6 substituents selected from the group consisting of RL1 and RL2, C3-11 heterocyclyl substituted with 0-6 substituents selected from the group consisting of RL1 and RL2, aryl substituted with 0-6 substituents selected from the group consisting of RL1 and RL2, and heteroaryl substituted with 0-6 substituents selected from the group consisting of RL1 and RL2, wherein:
RL1 and RL2 are each independentiy linked to other A1 to Aq to form a cycloalkyl or heterocyclyl moiety substituted with 0-4 RL5 groups;
RL1, RL2,RL3 ,RM and RL5 are each independently selected from the group consisting of H, C1-8 alkyl, O(C1-8 alkyl), S(C1-8 alkyl), NH(C1-8 alkyl), N(C1-8 alkyls, C3-11 cycloalkyl, aryl, heteroaryl, C3-11 heterocyclyl, O(C1-8 cycloalkyl), S(Cn cycloalkjd), NH(CM cycloalkyl), N(C1-8 cycloalkyl)2, OH,NH2, SH, C≡CH, CO2H, halogen, CN, CF3, CHF2, CH2F, and NO2; and q is an integer greater than or equal to 1.
14. The bifunctional compound according to claim 1, wherein Linker is selected from the group consisting of:
Figure imgf000084_0001
10
Figure imgf000085_0001
wherein: k is an integer of 0 or 1; m is an integer selected from 0—12; n is an integer selected from 0~8; and the dashed lines, respectively, indicate the sites of attachment to RB and ULM. 15. A bifunctional compound selected from the group consisting of:
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(12-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino>12-oxododecanoyl)piperidine-4-carboxamide (cpd 1); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(2-(2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)ethoxy)ethoxy)ethoxy)ethyl)piperidine-4-carboxamide (cpd 2); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol- 1 -yl)propyl)- 1 -(4-(2-((2-(2,6-dioxopiperidin-3-yl)-l ,3-dioxoisoindolin-4- yl)oxy)acetaniido)butanoyl)piperidine-4-carboxamide (cpd 3);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(3-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)ethoxy)ethoxy)propanoyl)piperidine-4-carboxamide (cpd 4); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino>3,6,9,12-tetraoxatetradecanoyl)piperidine-4-carboxamide (cpd 5); 4-amino-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (tiifluoromethoxy)phenyl)- ΙΗ-indol- 1 -yl)propyl)- 1 -(14-((2-(2,6-dioxopiperidin-3-yl)- 1,3- dioxoisoindolin-4-yl)amino)-3,6,9, 12-tetraoxatetradecanoyl)piperidine-4- carboxamide (cpd 6);
4-amino-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (tiifluoromethoxy)phenyl)- ΙΗ-indol- 1 -yl)propyl)- 1 -(1 -((2-(2,6-dioxopiperidin-3-yl)- 1,3- dioxoisoindolin-4-yl)amino)-3,6,9,12-tetraoxapentadecan-15-oyl)piperidine-4- carboxamide (cpd 7);
4-amino-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- 1 H-indol- 1 -y l)propy 1)- 1 -(3-(2-(2-(2-((2-(2,6-dioxopiperidin-3 - yl)-l,3-dioxoisoindolin-4-yl)amino)ethoxy)ethoxy)e1hoxy)propanoyl)piperidine-4- carboxamide (cpd 8);
4-amino-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- ΙΗ-indol- 1 -yl)propyl)- 1 -(1 -((2-(2,6-dioxopiperidin-3-yl)- 1,3- dioxoisoindolin-4-yl)amino)-3,6,9, 12, 15-pentaoxaoctadecan- 18-oyl)piperidine-4- carboxamide (cpd 9);
N-(3-(5-((4-(2,6-didilorobenzyl)piperazin-l-yl)methyl)-3-phenyl-lH-indol-l-yl)propyl)- l-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)amino)-3, 6,9,12- tetraoxatetradecanoyl)piperidine-4-carboxamide (cpd 10);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(l-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino>3,6,9,12,15-paitaoxaoctadecan-18-qyl)piperidine-4-caiboxamide (cpd 11); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-4-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-3,6,9,12-tetraoxatetradecanamido)piperidine-4-carboxamide (cpd 12); N-(4-((3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- 1 H-indol- 1 -yl)propyl)amino)-4-oxobutyl)- 14-((2-(2,6- dioxopiperidin-3-yl)-l ,3-dioxoisoindolin-4-yl)amino)-3,6,9, 12-tetraoxatetradecanamide (cpd 13);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-3,6,9,12-tetraoxatetradecanoyl)piperidine-3-carboxamide (cpd 14); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(3-(2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)ethoxy)ethoxy)ethoxy)propanoyl)piperidine-4-carboxamide (cpd 15); N-(3-(5-((4-(2,6-clichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(l-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-3,6,9,12-tetraoxapentadecan-15-oyl)piperidine-4-carboxamide (cpd 16); 4-amino-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- 1 H-indol- 1 -yl)propyl)-l -((R)- 18-((2R,4S)-4-hydroxy-2-((4-(4- methy lthiazol-5-y l)benzy l)carbampy l)py rrolidine- 1 -caibony 1)- 19,19-dimethyl- 16-oxo- 4,7,10,13-tetraoxa-17-azaicosanoyl)piperidine-4-caiboxamide (cpd 17); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- 1 H-indol- 1 -yl)propyl)- 1 -((R)- 18-((2R,4S)-4-hydroxy-2-((4-(4-methylthiazol-5- yl)benzyl)carbamoyl)py rrolidine- 1 -carbonyl)- 19, 19-dimethyl- 16-oxo-4,7,l 0, 13-tetraoxa- 17-azaicosanoyl)piperidine-4-carboxamide (cpd 18); N-(3-(5-((4-(2,6-didilorobenzyl)-2-methylpiperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)pheny 1)- ΙΗ-indol- 1 -y l)propy 1)- 1 -( 14-((2-(2,6-dioxopiperidin-3 -yl)- 1,3- dioxoisoindolin-4-yl)amino)-3,6,9, 12-tetraoxatetradecanoyl)piperidine-4- carboxamide (cpd 20);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-3,6,9,12-tetraoxatetradecanoyl)piperidine-2-carboxamide (cpd 21); N-(3-(5-((4-(2,6-didilorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-3,6,9,12-tetraoxatetradecanoyl)pynOlidine-2-carboxamide (cpd 22); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-3,6,9,12-tetraoxatetradecanoyl)-4-methylpiperidine-4-carboxaniide (cpd 23); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazm-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-4-(14-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-3,6,9,12-tetraoxatetradecanoyl)piperazine-2-carboxamide (cpd 24); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-((S)-18-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5- yl)benzyl)carbamoyl)pynolidine-l -carb(xiyl)-19, 19-dimethyl- 16-oxo-4,7,l 0, 13-tetraoxa- 17-azaicosan-l-oyl)piperidine-3-carboxamide (cpd 25);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- 1 H-indol- 1 -y l)propyl)- 1 -((S)-l 8-((2S,4R)-4-hy droxy-2-((4-(4-methy lthiazol-5 - yl)benz>l)carbamoyl)pyrrolidine-l -carbonyl)- 19, 19-dimethyl- 16-oxo-4,7,l 0, 13-tetraoxa- 17-azaicosan-l-oyl)piperidine-2-carboxamide (cpd 26);
N-(3-(6-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(l-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)-3,6,9.12-tetraoxapentadecan- 15-oyl)piperidine-4-carboxamide (cpd 27); N-(3-(7-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(l-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino>3,6,9,12-tetraox¾)entadecan-15-oyl)piperidine-4-carboxamide (cpd 28); N-(3-(6-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- 1 H-indol- 1 -y l)propyl)- 1 -((S)-l 8-((2S,4R)-4-hydroxy-2-((4-(4-methy lthiazol-5 - yl)benzyl)carbamoyl)pynohdine-l -carbonyl)- 19, 19-dimethyl- 16-oxo-4,7,l 0, 13-tetraoxa- 17-azaicosanoyl)piperidine-4-carboxamide (cpd 29); Nl-(4-((3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- 1 H-indol- 1 -yl)propyl)amino)-4-oxobutyl)-N 16-((R)-l - ((2R,4S)-4-hydroxy-2-((4-(4-methylthiazol-5-yl)benzyl)carbamoyl)pynOlidin-l-yl)-3,3- dimethyl-l-oxobutan-2-yl)-4,7,10,13-tetraoxahexadecanediamide (cpd 30); N-(3-(7-((4-(2,6-didilorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- 1 H-indol- 1 -y l)propyl)- 1 -((S)-l 8-((2S,4R)-4-hy droxy-2-((4-(4-methy lthiazol-5 - yl)benzyl)carbamoyl)pynohdine-l -carbonyl)- 19.19-dimethyl- 16-oxo-4,7,l 0, 13-tetraoxa- 17-azaicosanoyl)piperidine-4-carboxamide (cpd 31);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(9-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)nonanoyl)piperidine-4-carboxamide (cpd 32);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- 1 H-indol- 1 -yl)propyl)-l -(12-(((R)- 1 -((2R,4S)-4-hy droxy-2-((4-(4-methyltiiiazol-5- yl)benzyl)carbamoyl)pynohdin-l-yl)-3,3-dimethyl-l-oxobutan-2-yl)amino)-12- oxododecanoyl)piperidine-4-carboxamide (cpd 33);
N-(3-(5-((4-(2,6-didilorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- 1 H-indol- 1 -y l)propyl)- 1 -((S)-l 4-((2S,4R)-4-hy droxy-2-((4-(4-methy lthiazol-5 - yl)benzyl)carbamoyl)pyrrolidine-l -carbonyl)- 15, 15-dimethyl- 12-oxo-3, 6, 9-trioxa- 13- azahexadecyl)piperidine-4-carboxamide (cpd 34);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- ΙΗ-indol- 1 -yl)propyl)- 1 -(2-(3-(((S)- l-((2S,4R)-4-hy droxy-2-((4-(4-methylthiazol-5- yl)benzyl)carbamoyl)pynoUdm-l-yl)-3,3-dimethyl-l-oxobutan-2-yl)amino)-3- oxopropoxy)ethyl)piperidine-4-carboxaniide (cpd 35);
N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(3-(4-(2-(((S)-l-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5- yl)benzyl)carbamoyl)pynoliclin- l-yl)-3, 3-dimethyl- 1 -oxobutan-2-yl)amino)-2- oxoethyl)piperazin-l-yl)propyl)piperidine-4-carboxamide (cpd 36); N-(3-(5-((4-(2,6-dicMorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(3-(2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5- yl)amino)ethoxy)ethoxy)ethoxy)propanoyl)piperidine-4-carboxamide (qxl 37); N-(3-(5-((4-(2,6-dicMorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(l-((2-(2,6-dioxopiperidm-3-yl)-l,3-dioxoisoindolin-5- yl)amino)-3,6.9,12-tetraoxapentadecan-15-oyl)piperidine-4-carboxamide (cpd 38); N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)- lH-indol-l-yl)propyl)-l-(l-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)amino)- 3,6,9, 12-tetraoxapentadecan- 15-oyl)piperidine-4-carboxamide (cpd 39); 4-amino-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- ΙΗ-indol- 1 -yl)propyl)- 1 -(1 -((2-(2,6-dioxopiperidin-3-yl)- 1 - oxoisoindolin-4-y l)amino)-3 ,6,9,12-tetraoxapentadecan- 15 -oy l)piperidine-4-caiboxamide (cpd 40);
1 -( 1 -(( 1 -(2-chloroacety 1)- 1 ,2,3,4-tetrahydroquinolin-6-yl)oxy)-2-oxo-6,9, 12, 15-tetraoxa-
3-azaoctadecan-18-oyl)-N-(3-(5-((4-(2,6-dichloroben2yl)pipeiazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)-lH-indol-l-yl)propyl)piperidine-4-carboxamide (cpd 41);
1 -( 1 -(( 1 -(2-diloroacety 1)- 1 ,2,3,4-tetrahydroquinolin-6-yl)oxy)-2-oxo-6,9, 12, 15, 18- pentaoxa-3-azahenicosan-21-oyl)-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l- yl)mefliyl)-3-(4-(trifluoromethoxy)phenyl)- ΙΗ-indol- 1 -yl)propyl)piperidine-4- carboxamide (cpd 42);
4-amino- 1 -(1 -(( 1 -(2-chloroacety 1)- 1 ,2,3,4-tetrahydroquinolin-6-yl)oxy)-2-oxo-6,9, 12,
15- tetraoxa-3-azaoctadecan-18-pyl)-N-(3-(5-((4-(2,6-didilorobenzyl)piperazin-l-yl)methyl)- 3-(4-(trifluoromethoxy)phenyl)-lH-indol-l-yl)propyl)piperidine-4-cait>oxamide (cpd 43); 1 -( 1 -(( 1 -(2-chloroacetyl)-l ,2,3,4-tetrahy droquinolin-6-yl)oxy)-2-oxo-6,9, 12-trioxa-3- azzpentadecan-15-oyl)-N-(3-(5-((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4- (trifluoromethoxy)phenyl)- lH-indol- 1 -yl)propyl)piperidine-4-carboxamide (cpd 44); 4-amino- 1 -(1 -(( 1 -(2-chloroacetyl)- 1 ,2,3,4-tetrahydroquinolin-6-yl)oxy)-2-oxo-6,9, 12- trioxa-3-azapentadecan-15-oyl)-N-(3-(5-((4-(2.6-dichlorobenzyl)piperazin-l-yl)methyl)- 3-(4-(trifluoromethoxy)phenyl)-lH-indol-l-yl)propyl)piperidine-4-carboxamide (cpd 45); and
4-amino-l-(2-((l-(2-chloroacetyl)-l,2,3,4-tetrahydroqiiinolin-6-yl)oxy)acetyl)-N-(3-(5- ((4-(2,6-dichlorobenzyl)piperazin-l-yl)methyl)-3-(4-(trifluoromethoxy)phenyl)-lH-indol- 1 -yl)propyl)piperidine-4-caiboxainide (q)d 46); or pharmaceutically acceptable salts, solvates, hydrates, polymorphs, tautomers, stereoisomers, isotopically enriched derivatives, or prodrugs thereof.
16. A pharmaceutical composition comprising an effective amount of the bifunctional compound according to any one of claims 1 to 15, and one or more pharmaceutically acceptable excipients.
17. Use of a therapeutically effective amount of the bifunctional compound according to any one of claims 1 to 15 for manufacturing a medicament for preventing, ameliorating and/or treating a disease associated with RAS mutation in a subject in need thereof.
18. The use according to claim 17, wherein the bifunctional compound is administered separately, sequentially or together with one or more additional anticancer agents.
19. The use according to claim 18, wherein the additional anticancer agents are selected from tiie group consisting of trastuzumab, ramucirumab, vismodegib, sonidegib, bevacizumab, everolimus, tamoxifen, toremifene, fulvestrant, anastrozole, exemestane, lapatinib, letrozole, pertuzumab, ado-trastuzumab emtansine. palbociclib, cetuximab, panitumumab, ziv-aflibercept, regorafenib, lmatinib mesylate, lanreotide acetate, sunitinib, regorafenib, denosumab, alitretinoin, sorafenib, pazopanib, temsirolimus, everolimus, tretinoin, dasatinib, nilotinib, bosutinib, rituximab, alemtuzumab, ofatumumab, obinutuxumab, ibrutinib, idelalisib, blinatumomab, soragenib, crizotinib, erlotinib, gefitinib, afatinib dimaleate, ceritnib, ramucirumab, nivolumab, pembrolizumab, osimertinib, necitumumab, busulfan, chlorambucil, cyclophosphamide, iphosphamide, melphalan, nitrogen mustard, streptozodn, thiotepa, uracil nitrogen mustard, trietiiylenemelamine, temozolomide, 2- chloroethyl-3-sarcosinamide-l -nitrosourea (SarCNU), actinomycin-D, bleomycin, cryptophycins, daunorubicin, doxorubicin, idambicin, irinotecan, L-asparaginase, mitomycin-C, mitramycin, navelbine, paclitaxel, docetaxel, topotecan, vinblastine, vincristine, teniposide (VM-26), etoposide (VP-16), 5a-reductase inhibitor, aminoglutethimide, anastrozole, bicalutamide, dilorotrianisene, diethylstilbestrol (DBS), dromostanolone, estramustine, ethinyl estradiol, flutamide, fluoxymesterone, goserelin, hydroxyprogesterone, letrozole, leuprolide, medroxyprogesterone acetate, megestrol acetate, methyl prednisolone, methyltestosterone, mitotane, nilutamide, prednisolone, arzoxifene (SERM-3), tamoxifen, testolactone, testosterone, triamicnolone, zoladex, all- trans retinoic acid, carmustine (BCNLJ), carboplatin (CBDCA), lomustine (CCNU), ds- diaminedichloroplatinum (dsplatin), dacarbazine, gliadel, hexamethylmelamine, hydroxyurea, levamisole, mitoxantrone, ο,ρ'- dichlorodiphenyldichloroethane (o,p'-DDD) (also known as lysodren or mitotane), oxaliplatin, porfimer sodium, procarbazinejmalinib mesylate, chlorodeoxy adenosine, cytosine arabinoside, 2'-deoxycoformydn, fludarabine phosphate, 5-fluorouracil (5-FU), 5-fluoro-2'-deoxy uridine (5-FUdR), gemcitabine, camptothecin, 6-mercaptopurine, methotrexate, 4-methylthioamphetamine (4-MTA), thioguanine, alpha interferon, BCG (Bacillus Calmett e-Guerin), granulocyte colony stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM- CSF), interleukin-2, and herceptin.
20. The use according to claim 17, wherein the disease is cancer, autoimmune disease, infectious disease, or blood vessel proliferative disorder.
21. The use according to claim 20, wherein the cancer is selected from the group consisting of lung cancer, colon cancer, colorectal cancer, breast cancer, prostate cancer, liver cancer, pancreatic cancer, bladder cancer, gastric cancer, renal cancer, salivary gland cancer, ovarian cancer, uterine body cancer, cervical cancer, oral cancer, skin cancer, brain cancer, lymphoma, leukemia, biliary tract malignancies, endometrial cancer, cervical cancer, and myeloid leukemia.
22. The use according to claim 21, wherein the cancer is pancreatic cancer, colon cancer, lung cancer, or non-small cell lung cancer.
PCT/US2021/029237 2020-04-27 2021-04-26 Compounds for mutant ras protein degradation WO2021222138A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063015848P 2020-04-27 2020-04-27
US63/015,848 2020-04-27

Publications (1)

Publication Number Publication Date
WO2021222138A1 true WO2021222138A1 (en) 2021-11-04

Family

ID=78332142

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/029237 WO2021222138A1 (en) 2020-04-27 2021-04-26 Compounds for mutant ras protein degradation

Country Status (2)

Country Link
TW (1) TW202206423A (en)
WO (1) WO2021222138A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115403561A (en) * 2022-08-22 2022-11-29 西安交通大学 Intracellular self-assembly protein degradation agent based on thalidomide analogue and preparation method and application thereof
WO2022266206A1 (en) * 2021-06-16 2022-12-22 Erasca, Inc. Kras inhibitor conjugates
WO2023099620A1 (en) * 2021-12-01 2023-06-08 Boehringer Ingelheim International Gmbh Kras degrading compounds comprising annulated 2-amino-3-cyano thiophenes
WO2023116934A1 (en) * 2021-12-24 2023-06-29 苏州泽璟生物制药股份有限公司 Krasg12d protein hydrolysis regulator, and preparation method therefor and use thereof
WO2023130012A1 (en) * 2021-12-29 2023-07-06 Development Center For Biotechnology Compounds for mutant kras protein degradation and uses thereof
WO2023215906A1 (en) * 2022-05-06 2023-11-09 Hangzhou Jijing Pharmaceuticaltechnology Limited Kras g12d proteolysis targeting chimeras

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160229836A1 (en) * 2014-05-30 2016-08-11 The Trustees Of Columbia University In The City Of New York Multivalent ras binding compounds
WO2017024317A2 (en) * 2015-08-06 2017-02-09 Dana-Farber Cancer Institute, Inc. Methods to induce targeted protein degradation through bifunctional molecules
US20170158702A1 (en) * 2015-12-02 2017-06-08 Kyras Therapeutics, Inc. Multivalent ras binding compounds
US20200000927A1 (en) * 2016-11-15 2020-01-02 Tokyo University Of Science Foundation Ras protein degradation inducing molecule and pharmaceutical composition

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160229836A1 (en) * 2014-05-30 2016-08-11 The Trustees Of Columbia University In The City Of New York Multivalent ras binding compounds
WO2017024317A2 (en) * 2015-08-06 2017-02-09 Dana-Farber Cancer Institute, Inc. Methods to induce targeted protein degradation through bifunctional molecules
US20170158702A1 (en) * 2015-12-02 2017-06-08 Kyras Therapeutics, Inc. Multivalent ras binding compounds
US20200000927A1 (en) * 2016-11-15 2020-01-02 Tokyo University Of Science Foundation Ras protein degradation inducing molecule and pharmaceutical composition

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022266206A1 (en) * 2021-06-16 2022-12-22 Erasca, Inc. Kras inhibitor conjugates
WO2023099620A1 (en) * 2021-12-01 2023-06-08 Boehringer Ingelheim International Gmbh Kras degrading compounds comprising annulated 2-amino-3-cyano thiophenes
WO2023116934A1 (en) * 2021-12-24 2023-06-29 苏州泽璟生物制药股份有限公司 Krasg12d protein hydrolysis regulator, and preparation method therefor and use thereof
WO2023130012A1 (en) * 2021-12-29 2023-07-06 Development Center For Biotechnology Compounds for mutant kras protein degradation and uses thereof
WO2023215906A1 (en) * 2022-05-06 2023-11-09 Hangzhou Jijing Pharmaceuticaltechnology Limited Kras g12d proteolysis targeting chimeras
CN115403561A (en) * 2022-08-22 2022-11-29 西安交通大学 Intracellular self-assembly protein degradation agent based on thalidomide analogue and preparation method and application thereof
CN115403561B (en) * 2022-08-22 2024-03-08 西安交通大学 Intracellular self-assembled protein degradation agent based on thalidomide analogue, and preparation method and application thereof

Also Published As

Publication number Publication date
TW202206423A (en) 2022-02-16

Similar Documents

Publication Publication Date Title
WO2021222138A1 (en) Compounds for mutant ras protein degradation
EP3063143B1 (en) Aminoheteroaryl benzamides as kinase inhibitors
US10905665B2 (en) Chemical modulators of signaling pathways and therapeutic use
EP3423443B1 (en) Cyano-substituted indole compounds and uses thereof as lsd1 inhibitors
JP2022502385A (en) Method for producing a compound for inhibiting the activity of SHP2
KR102653190B1 (en) Highly active STING protein agonist compounds
JP2022523916A (en) Bifunctional compounds and methods for ubiquitination targeting androgen receptors
WO2018133795A1 (en) Ezh2 inhibitor and use thereof
US11702423B2 (en) BRM targeting compounds and associated methods of use
WO2016210247A1 (en) New methods of use for an anti-diarrhea agent
EP3350181A1 (en) Her3 ligands and uses thereof
KR20200054046A (en) Novel piperidine-2,6-dione derivatives and use thereof
WO2023130012A1 (en) Compounds for mutant kras protein degradation and uses thereof
WO2020092662A1 (en) Proteolysis-targeting chimeras
WO2017025493A1 (en) Quinoline ezh2 inhibitors
KR20180085814A (en) Preparation of Substituted 5,6-dihydro-6-phenylbenzo [f] isoquinolin-2-amine
EP4006033A1 (en) Adenosine receptor antagonist
JP6257835B2 (en) Novel triazolopyrimidinone or triazolopyridinone derivatives and their uses
EP3114114B1 (en) Tricyclic prodrugs
WO2022081842A1 (en) Substituted acyl sulfonamides for treating cancer
JP2022549353A (en) Diterpenoid compounds acting on protein kinase C (PKC)
JP2021134218A (en) Pharmaceuticals comprising optically active azabicyclo ring derivative
CN112341390A (en) Compound for preparing targeted histone methyltransferase EZH2 covalent inhibitor and preparation method and application thereof
EP3750894A1 (en) Urea-substituted aromatic ring-linked dioxazoline compound, preparation method therefor, and uses thereof
WO2023185864A1 (en) Compounds for Targeted Degradation of KRAS

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: 21795250

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21795250

Country of ref document: EP

Kind code of ref document: A1