WO2024054625A2 - Bifunctional compounds for degrading kras g12d via ubiquitin proteasome pathway - Google Patents

Bifunctional compounds for degrading kras g12d via ubiquitin proteasome pathway Download PDF

Info

Publication number
WO2024054625A2
WO2024054625A2 PCT/US2023/032284 US2023032284W WO2024054625A2 WO 2024054625 A2 WO2024054625 A2 WO 2024054625A2 US 2023032284 W US2023032284 W US 2023032284W WO 2024054625 A2 WO2024054625 A2 WO 2024054625A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkylene
hydrogen
alkyl
compound
heterocyclylene
Prior art date
Application number
PCT/US2023/032284
Other languages
French (fr)
Other versions
WO2024054625A3 (en
Inventor
Yongfeng SUN
Jiping Fu
Haiqiang ZENG
Original Assignee
Nikang Therapeutics, Inc.
Shanghai Blueray Biopharma Co., Ltd.
Nikang Therapeutics (Shanghai) Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/CN2022/117697 external-priority patent/WO2024050742A1/en
Application filed by Nikang Therapeutics, Inc., Shanghai Blueray Biopharma Co., Ltd., Nikang Therapeutics (Shanghai) Ltd. filed Critical Nikang Therapeutics, Inc.
Publication of WO2024054625A2 publication Critical patent/WO2024054625A2/en
Publication of WO2024054625A3 publication Critical patent/WO2024054625A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present disclosure provides certain bifunctional compounds that cause degradation of K-ras G12D via ubiquitin proteasome pathway and are therefore useful for the treatment of diseases mediated by K-ras G12D. Also provided are pharmaceutical compositions containing such compounds and processes for preparing such compounds.
  • Kirsten Rat Sarcoma 2 Viral Oncogene Homolog (KRAS) gene is a prevalent oncogene that encodes a small GTPase transductor protein called K-Ras.
  • K-Ras can serve as a molecular switch by cycling between active GTP-bound and inactive GDP-bound forms (see Science 2001;294: 1299-304.).
  • K-Ras signaling is activated by RAS guanine nucleotide exchange factors (GEFs), e.g., Son of Sevenless homologue (SOS) protein, that facilitate the GDP to GTP exchange of K-Ras (see Curr Biol 2005;15:563-74.).
  • GEFs RAS guanine nucleotide exchange factors
  • SOS Son of Sevenless homologue
  • GAPs GTPase-activating proteins
  • K-Ras plays a crucial role in the regulation of cell proliferation, differentiation and survival by signaling through several major downstream pathways, including the MAPK, the PI3K and the Ral-GEFs pathways (see Lung Cancer 2018; 124: 53-64), among them the MAPK pathway is the best characterized (see Mol. Cell Biol.l995;15:6443-6453.).
  • K-Ras-GTP binds to and activates RAF kinases, which phosphorylates MEK and subsequently phosphorylates ERK. Phospho-ERK can further activate downstream cytosolic proteins and which then translocate to the nucleus to drive the expression of diverse genes, propagating the growth signal.
  • PI3K pathway is also involved in RAS-mediated tumorigenesis (see Cell 2007; 129:957- 968.).
  • PI3K phosphorylates PIP2 to form PIP3, activates PDK1 and then phosphorylates AKT.
  • pAKT yields phosphorylation of several physiological substrates, e.g., mTOR, FOXO and NF-KB that promote metabolism, cell-cycle progression, resistance to apoptosis, cell survival and migration.
  • the Ral-GEFs signaling pathway plays a key role in RAS- mediated oncogenesis as well (see Proc. Natl. Acad. Sci. U. S. A. 1994; 91:11089-11093.).
  • the K-Ras effector, RALGDS stimulates the RAS family RAL-A/B small GTPases for the subsequent signaling cascades.
  • RALGDS can also promote the JNK pathway to stimulate transcription of pro-survival and cell-cycle progression genes for cell proliferation and survival.
  • KRAS gene is the most frequently mutated oncogene in human cancer. KRAS mutations are associated with poor clinical outcome and found at high frequency in pancreatic cancer (-90%), colorectal cancer ( ⁇ 44%) and non-small-cell lung cancer (NSCLC) ( ⁇ 29%) (see Cancer Discov. 2021 ; 11 : 1-16). KRAS mutations are also present in breast cancer, liver cancer, biliary tract malignancies, endometrial cancer, cervical cancer, bladder cancer and myeloid leukemia.
  • K-Ras G12C offers special opportunity, because it harbors a non-native cysteine residue, which can act as nucleophile and therefore can be targeted by covelent attachment.
  • AMG5 10 which is an approved drug for treating K-Ras G12C driven cancers
  • K- Ras G12C covelent inhibitors including MRTX849, JNJ-74699157 and LY349944631, are in clinical trials for treating cancer patients with KRAS G12C mutation (see ACS Cent. Sci. 2020;6:1753-1761).
  • K-Ras G12D mutant occupy a dynamic pocket in the switch II region of K-Ras thereby irreversibly locking K-Ras G12C in inactive GDP -bound state. Since KRAS mutations, including G12C, enrich predominantly active-state protein in cancer cells, sufficient residual GTPase activity and nucleotide cycling are required for effective inhibition of K-Ras by inactive state-selective drugs (see Cell 2020; 183(4):850-859). Compared to K-ras G12C mutant, since K- Ras G12D mutant, does not contain non-native cysteine residue and cycle through inactive state at extremely low rate, thus making K-Ras G12D mutant specific drug discovery more challenging.
  • K-Ras G12D protein As an alternative to inhibition, removal of K-Ras G12D protein would eliminate K-Ras G12D activity as well as any protein interaction or scaffolding function of K-Ras G12D. Accordingly, there is a need for bifunctional molecules that could recruit K-Ras G12D to a ubiquitin ligase and thereby causing ubiquitylation and proteasomal degradation of K-Ras G12D.
  • the present disclosure fulfills this and related needs.
  • U, V, and W are CH; or one or two of U, V, and W are N and the other of U, V, and W are CH; m is 1 or 2, and n is 1, 2, or 3;
  • R 1 is hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, hydroxy, hydroxylalkyl, alkoxy alkyl, cyano, or cyanomethyl, provided R 1 is not attached to the ring -NH-;
  • R 1b is a bond to L, hydrogen, deuterium, alkyl, alkylidenyl, alkoxy, halo, haloalkyl, hydroxy, hydroxyalkyl, alkoxyalkyl, cyano, cyanomethyl, cyanoethyl, or 2-cyanovinyl, provided R 1b is not attached to the ring -NH-;
  • R 1c is hydrogen or alkyl, provided R 1c is not attached to the ring -NH-;
  • R 2 is hydrogen, deuterium, alkyl, halo, haloalkyl, alkoxy, hydroxy, or cyano, provided that R 2 is absent when two of U, V, and W are N;
  • R 3 is a bond to L, hydrogen, deuterium, alkyl, halo, haloalkyl, alkoxy, cycloalkyl, cycloalkyloxy, hydroxy, or cyano;
  • R 4 is -Z-R 6 where Z is a bond, O, NH, N(alkyl), or S; and
  • R 6 is heterocyclylalkyl, fused heterocyclylalkyl, bicyclic heterocyclyl, bicyclic heterocyclylalkyl, fused bicyclic heterocyclyl, fused bicyclic heterocyclylalkyl, tricyclic heterocyclyl, or tricyclic heterocyclylalkyl, wherein:
  • R 7 , R 9 , and R 11 are independently a bond to L, hydrogen, deuterium, alkyl, fluoro, or haloalkyl;
  • R 8 , R 10 , and R 12 are independently hydrogen, deuterium, alkyl, halo, haloalkyl, cyano, alkyloxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, phenyl, or heteroaryl; or independently of each other, R 7 and R 8 , R 9 and R 10 , and R 11 and R 12 together with the carbon atom to which they are attached form cycloalkylene optionally substituted with alkyl, halo, alkoxy, or hydroxy;
  • R b , R c , and R b are independently a bond to L, hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, hydroxy, or -(alkylene) n i-OC(O)NR 13 R 13a , wherein nl is 0 or 1, R 13 is hydrogen, deuterium, alkyl, deuterioalkyl, haloalkyl, haloalkoxyalkyl, or alkoxyalkyl, and R 13a is hydrogen, alkyl, deuterioalkyl, cycloalkyl, alkoxy, alkoxyalkyl, haloalkyl, haloalkoxyalkyl, or heterocyclyl substituted with Ri and R k independently selected from alkyl, halo, and haloalkyl; or R 13 and R 13a together with the nitrogen atom to which they are attached form heterocyclyl, bicyclic heterocyclyl, bridged heterocyclyl, fused heterocycly
  • R c , R f , and R 1 are independently a bond to L, hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, or hydroxy, provided that one of R 1b , R 3 , R 7 , R 9 , R 11 , R b , R c , R b , R c , R f , and R 1 is a bond to L;
  • Degron is an E3 ligase ligand selected from:
  • R 15 and R 16 are hydrogen, alkyl, cycloalkyl, or alkylcarbonyloxy
  • Y a is CH or N;
  • Z a is a bond, -CH2-, -NH-, O, or -NHC(O)- where NH of -NHC(O)- is attached to Y a ;
  • ring A is a ring of formula (a), (b), or (c): where:
  • R 22 is hydrogen or alkyl
  • ring B is phenylene, cyclylaminylene, a 5- or 6-membered monocyclic heteroarylene, or a 9- or 10-membered fused bicyclic heteroarylene, wherein each heteroarylene ring contains one to three nitrogen ring atoms and further wherein the phenylene, cyclylaminylene, and heteroarylene are independently substituted with R cc and R ff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano; and
  • R 17 , R 18 , and R 19 are alkyl, hydroxyalkyl, cycloalkyl or heterocyclyl wherein cycloalkyl and heterocyclyl are substituted with R 1 TM 11 selected from hydrogen, alkyl, halo, haloalkyl, alkoxyl, haloalkoxyl, cyano, alkylcarbonyl, alkylcarbonylamino, or -COR 23 where R 23 is alkyl, hydroxalkyl, cycloalkyl or heterocyclyl, wherein cycloalkyl and heterocyclyl are substituted with R nn selected from hydrogen, alkyl, halo, haloalkyl, alkoxyl, haloalkoxyl, cyano, alkylcarbonyl, and alkylcarbonylamino; and
  • W a is bond, O, S, or alkylene
  • L is -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 - where:
  • Z 1 is a bond, alkylene, -NR”-, -O-, -(alkylene-O)-, -C(O)-, -S(O)2-, -NR’(CO)-, or -C(O)NR-;
  • Z 2 is a bond, -alkylene, -NR”-, -O-, -C(O)-, -S(O) 2 -, -NR’(CO)-, -C(O)NR-, phenylene, monocyclic heteroarylene, or heterocycylene, where each ring is substituted with R ww and R xx independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
  • Z 3 is a bond, alkylene, alkynylene, -(alkylene -NR”)-, -(NR”-alkylene)-, -O-, -C(O)- , -NR”-, -(O-alkylene)d-, -(alkylene-O)d-, cycloalkylene, spiro cyclolalkylene, phenylene, heteroarylene, heterocyclylene, fused heterocyclylene, bridged heterocyclylene, or spiro heterocyclylene, where each ring is substituted with R uu and R vv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, cyanoalkyl, hydroxy, hydroxyalkyl, alkoxyalkyl, amino, alkylamino, and dialkylamino;
  • Z 4 is a bond, alkylene, alkynylene, -C(O)NR-, -NR’(CO)-, -O-, -NR”-, -(O- alkylene) c -, -(alkylene-O)c-, cycloalkylene, spiro cyclolalkylene, phenylene, monocyclic heteroarylene, heterocyclylene, bicyclic heterocyclylene, bridged heterocyclylene, fused heterocyclylene, spiro heterocyclylene, or 11 to 13 membered spiro heterocyclylene, where each ring is substituted with R ss and R tt independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
  • Z 5 is a bond, alkylene, alkynylene, -SO2-, -SO2NR-, -NR’SO2-, -C(O)-, - C(O)N(R)-, -NR’(CO)-, -(O-alkylene)b-, -(alkylene-O)b-, - ⁇ (CH2)7-, -O(CH2)8-, cycloalkylene, or heterocyclylene, where each ring is substituted with R qq and R" independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
  • Z 6 is a bond, alkylene, -C(O)NR-, -NR’(CO)-, -S(O) 2 NR-, -NR’S(O) 2 -, -(O- alkylene) a -, -(alkylene-O)a-, phenylene, monocyclic heteroarylene, heterocyclylene, bicyclic heterocyclylene, spiro heterocyclylene, -O-heterocyclylene-, -heterocyclylene-C(O)-, where each ring is substituted with R 00 and R pp independently selected from hydrogen, deuterium, alkyl, alkoxy, cyano, hydroxy, carboxy, halo, haloalkyl, and haloalkoxy; and where each alkylene of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 - is independently one to eight carbon atoms, R, R’ and R” is independently hydrogen or
  • R yy and R zz where R yy is hydrogen or deuterium and R zz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano; provided that at least one of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 - is not a bond; wherein when several adjacent groups of Z 1 to Z 6 are a bond, the adjacent groups represent one bond; or a pharmaceutically acceptable salt thereof.
  • U, V, and W are CH; or one or two of U, V, and W are N and the other of U, V, and W are CH; m is 1 or 2, and n is 1, 2, or 3;
  • R 1 is hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, hydroxy, hydroxylalkyl, alkoxy alkyl, cyano, or cyanomethyl, provided R 1 is not attached to the ring -NH-;
  • R 1a is hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, hydroxy, hydroxylalkyl, or alkoxyalkyl, provided R 1a is not attached to the ring -NH-; or when R 1 and R 1a are attached to the carbon atoms of the ring that are opposite or diagonal to each other, then R 1 and R 1a can combine to form -(CH2) Z - where (z is 1, 2, or 3), or -C I I C I I -;
  • R 1b is a bond to L, hydrogen, deuterium, alkyl, alkylidenyl, alkoxy, halo, haloalkyl, hydroxy, hydroxyalkyl, alkoxyalkyl, cyano, cyanomethyl, cyanoethyl, or 2-cyanovinyl, provided R 1b is not attached to the ring -NH-;
  • R 1c is hydrogen or alkyl, provided R lc is not attached to the ring -NH-;
  • R 2 is hydrogen, deuterium, alkyl, halo, haloalkyl, alkoxy, hydroxy, or cyano, provided that R 2 is absent when two of U, V, and W are N;
  • R 3 is a bond to L, hydrogen, deuterium, alkyl, halo, haloalkyl, alkoxy, cycloalkyl, cycloalkyloxy, hydroxy, or cyano;
  • R 4 is -Z-R 6 where Z is a bond, O, NH, N(alkyl), or S; and R 6 is heterocyclylalkyl, fused heterocyclylalkyl, bicyclic heterocyclyl, bicyclic heterocyclylalkyl, fused bicyclic heterocyclyl, fused bicyclic heterocyclylalkyl, tricyclic heterocyclyl, or tricyclic heterocyclylalkyl, wherein:
  • R 7 , R 9 , and R 11 are independently a bond to L, hydrogen, deuterium, alkyl, fluoro, or haloalkyl;
  • R 8 , R 10 , and R 12 are independently hydrogen, deuterium, alkyl, halo, haloalkyl, cyano, alkyloxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, phenyl, or heteroaryl; or independently of each other, R 7 and R 8 , R 9 and R 10 , and R 11 and R 12 together with the carbon atom to which they are attached form cycloalkylene optionally substituted with alkyl, halo, alkoxy, or hydroxy;
  • R b , R c , and R h are independently a bond to L, hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, hydroxy, or -(alkylene)ni-OC(O)NR 13 R 13a (wherein nl is 0 or 1, R 13 is hydrogen, deuterium, alkyl, deuterioalkyl, haloalkyl, haloalkoxyalkyl, or alkoxyalkyl, and R 13a is hydrogen, alkyl, deuterioalkyl, cycloalkyl, alkoxy, alkoxyalkyl, haloalkyl, haloalkoxyalkyl, or heterocyclyl substituted with Ri and R k independently selected from alkyl, halo, and haloalkyl; or R 13 and R 13a together with the nitrogen atom to which they are attached form heterocyclyl, bicyclic heterocyclyl, bridged heterocyclyl, fused heterocyclyl, or
  • R c , R f , and R 1 are independently a bond to L, hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, or hydroxy, provided that one of R 1b , R 3 , R 7 , R 9 , R 11 , R b , R c , R b , R c , R f , and R 1 is a bond to L;
  • Degron is an E3 ligase ligand selected from:
  • R 15 and R 16 are hydrogen, alkyl, cycloalkyl, or alkylcarbonyloxy
  • Y a is CH or N
  • Z a is a bond, -CH2-, -NH-, O, or -NHC(O)- where NH of -NHC(O)- is attached to Y a ;
  • ring A is a ring of formula (a), (b), or (c): where:
  • X 1 , X 2 , X 3 ’ and X 4 are independently a bond, -alkylene-, -O-, -(O-alkylene)-
  • R gg , R hh , R jj , and R kk are independently hydrogen, alkyl, or cycloalkyl and each alkylene is optionally substituted with one or two fluoro;
  • R aa , R bb , R cc , and R dd are independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano;
  • R 22 is hydrogen or alkyl
  • ring B is phenylene, cyclylaminylene, a 5- or 6-membered monocyclic heteroarylene, or a 9- or 10-membered fused bicyclic heteroarylene, wherein each heteroarylene ring contains one to three nitrogen ring atoms and further wherein the phenylene, cyclylaminylene, and heteroarylene are independently substituted with R cc and R ff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano; and R 17 , R 18 , and R 19 are alkyl, hydroxyalkyl, cycloalkyl or heterocyclyl wherein cycloalkyl and heterocyclyl are substituted with R mm selected from hydrogen, alkyl, halo, haloalkyl, alkoxyl, haloalkoxyl, cyano, alkylcarbonyl
  • W a is bond, O, S, or alkylene
  • L is -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 - where:
  • Z 1 is a bond, alkylene, -NR”-, -O-, -(alkylene-O)-, -C(O)-, -S(O)2-, -NR’(CO)-, or -C(O)NR-;
  • Z 2 is a bond, -alkylene, -NR”-, -O-, -C(O)-, -S(O) 2 -, -NR’(CO)-, -C(O)NR-, phenylene, monocyclic heteroarylene, or heterocycylene, where each ring is substituted with R ww and R xx independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
  • Z 3 is a bond, alkylene, alkynylene, -(alkylene -NR”)-, -(NR”- alkylene)-, -O-, - C(O)-, -NR”-, -(O-alkylene)d-, -(alkylene-O)d-, cycloalkylene, spiro cyclolalkylene, phenylene, heteroarylene, heterocyclylene, fused heterocyclylene, bridged heterocyclylene, or spiro heterocyclylene, where each ring is substituted with R uu and R vv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
  • Z 4 is a bond, alkylene, alkynylene, -C(O)NR-, -NR’(CO)-, -O-, -NR”-, -(O- alkylene) c -, -(alkylene-O)c-, cycloalkylene, spiro cyclolalkylene, phenylene, monocyclic heteroarylene, heterocyclylene, bicyclic heterocyclylene, bridged heterocyclylene, fused heterocyclylene, spiro heterocyclylene, or 11 to 13 membered spiro heterocyclylene, where each ring is substituted with R ss and R tt independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
  • Z 5 is a bond, alkylene, alkynylene, -C(O)-, -C(O)N(R)-, -NR’(CO)-, -(O- alkylene)b-, -(alkylene-O)b-, -O(CH 2 )7-, -O(CH2)8-, cycloalkylene, or heterocyclylene, where each ring is substituted with R qq and R rr independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
  • Z 6 is a bond, alkylene, -C(O)NR-, -NR’(CO)-, -S(O) 2 NR-, -NR’S(O) 2 -, -(O- alkylene) a -, -(alkylene-O)a-, phenylene, monocyclic heteroarylene, or heterocyclylene,
  • R yy and R zz where R yy is hydrogen or deuterium and R zz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano; provided that at least one of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 - is not a bond; wherein when several adjacent groups of Z 1 to Z 6 are a bond, the adjacent groups represent one bond; or a pharmaceutically acceptable salt thereof.
  • a pharmaceutical composition comprising a compound of Formula (IA) or (I) (or any of the embodiments thereof described herein) or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient.
  • a method of inhibiting K-Ras in particular K-Ras G12D, in a cell, comprising contacting the cell with a compound of Formula (IA) or (I) (or any of the embodiments thereof described herein).
  • the contacting is in vitro.
  • the contacting is in vivo.
  • a method of inhibiting cell proliferation in vitro or in vivo comprising contacting a cell with a compound of Formula (IA) or (I) (or any of the embodiments thereof described herein) or a pharmaceutical composition thereof as disclosed herein.
  • the contacting is in vitro.
  • the contacting is in vivo.
  • a method of treating cancer in a patient preferably the patient is in need of such treatment, which method comprises administering to the patient, preferably a patient in need of such treatment, a therapeutically effective amount of a compound of Formula (IA) or (I) (or any of the embodiments thereof described herein) or a pharmaceutically acceptable salt thereof or a a pharmaceutical composition thereof as disclosed herein.
  • a method of treating cancer associated with K-Ras, in particular K-Ras G12D, in a patient, preferably the patient is in need of such treatment comprises administering to the patient, preferably a patient in need of such treatment, a therapeutically effective amount of a compound of Formula (IA) or (I) (or any of the embodiments thereof described herein) or a pharmaceutically acceptable salt thereof or a a pharmaceutical composition thereof as disclosed herein.
  • a compound of Formula (IA) or (I), (or any embodiments thereof described herein) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof as disclosed herein for use as a medicament is provided.
  • the medicament is useful for the treatment of cancer.
  • any of the aforementioned aspects involving the treatment of cancer are further embodiments comprising administering the compound of Formula (IA) or (I) (or any embodiments thereof disclosed herein), or a pharmaceutically acceptable salt thereof in combination with at least one additional anticancer agent.
  • the agents can be administered simultaneously or sequentially.
  • Alkyl means a linear or branched saturated monovalent hydrocarbon radical of one to six carbon atoms, e.g., methyl, ethyl, propyl, 2-propyl, butyl, pentyl, and the like.
  • Alkylene means a linear or branched saturated divalent hydrocarbon radical of one to six carbon atoms unless otherwise stated e.g., methylene, ethylene, propylene, 1 -methylpropylene, 2- methylpropylene, butylene, pentylene, and the like.
  • Alkenyl means a linear or branched unsaturated monovalent hydrocarbon radical of two to six carbon atoms containing a double bond, e.g., ethenyl, propenyl, 2-propenyl, butenyl, pentenyl, and the like.
  • Alkynyl means a linear or branched unsaturated monovalent hydrocarbon radical of two to six carbon atoms containing a triple bond, e.g., ethynyl, propynyl, and the like.
  • Alkynylene means a linear or branched unsaturated divalent hydrocarbon radical of two to six carbon atoms containing a triple bond, e.g., and the like.
  • the alkylidene group, methylidenyl is enclosed by the box which is indicated by the arrow.
  • Alkylsulfonyl means a -SO 2 R z radical where R z is alkyl as defined above, e.g., methylsulfonyl, ethylsulfonyl, and the like.
  • Alkylthio means a -SR Z radical where R z is alkyl as defined above, e.g., methylthio, ethylthio, and the like.
  • Alkylcarbonyloxy means an -OC(O)R z group, where R z is alkyl, as defined herein.
  • Alkoxy means a -OR Z radical where R z is alkyl as defined above, e.g., methoxy, ethoxy, propoxy, or 2-propoxy, n-, iso-, or tert-butoxy, and the like.
  • Alkoxyalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with at least one alkoxy group, such as one or two alkoxy groups, as defined above, e.g., 2-methoxyethyl, 1-, 2-, or 3 -methoxypropyl, 2-ethoxyethyl, and the like.
  • Alkoxycarbonyl means a -C(O)OR Z radical where R z is alkyl as defined above, e.g., methoxycarbonyl, ethoxycarbonyl, and the like.
  • Alkylcarbonylamino means a -NR Z ’C(O)R Z radical where R z is alkyl and R z ’ is H or alkyl, as defined above, e.g., methylcarbonylamino, ethylcarbonylamino, and the like.
  • Alkylcarbonyl means a -C(O)R Z radical where R z is alkyl as defined herein, e.g., methylcarbonyl, ethylcarbonyl, and the like.
  • Alkylcarbonyloxy means a -OC(O) R z radical where R z is alkyl as defined above, e.g., methylcarbonyloxy, ethylcarbonyloxy, and the like.
  • Amino means a -NH2.
  • Alkylamino means -NHR Z radical where R z is alkyl is as defined above e.g., methylamino, ethylamino, propylamino, and the like.
  • Aryl means a monovalent monocyclic or bicyclic aromatic hydrocarbon radical of 6 to 10 ring atoms e.g., phenyl or naphthyl.
  • “Arylene” means a divalent aryl (as defined above) radical e.g., phenylene or naphthylene.
  • “Aralkyl” means a -(alkylene)-R z radical where R z is aryl as defined above. Examples include, but are not limited to, benzyl, phenethyl, and the like.
  • Bicyclic heterocyclyl means a saturated monovalent fused bicyclic ring of 8 to 12 ring atoms in which one or two ring atoms are heteroatom independently selected from N, O, and S(O) n , where n is an integer from 0 to 2, the remaining ring atoms being C, unless stated otherwise. Additionally, one or two ring carbon atoms in the heterocyclyl ring can optionally be replaced by a -CO- group. More specifically the term bicyclic heterocyclyl includes, but is not limited to, hexahydro- IH-pyrrolizinyl, and the like.
  • Bicyclic heterocyclylalkyl means a -(alkylene)-R radical where R is bicyclic heterocyclyl as defined above. Examples include, but are not limited to, hexahydro- 1H- pyrrolizinylmethyl, hexahydro- IH-pyrrolizinylethyl, and the like.
  • Bicyclic heterocyclylene means a saturated or unsaturated divalent fused bicyclic group of 9 to 12 ring atoms in which one, two, or three ring atoms are heteroatom independently selected from N, O, and S(O) n , where n is an integer selected from 0 to 2 inclusive, the remaining ring atoms being carbon, unless stated otherwise. Additionally, one or two ring carbon atoms of the bicyclic heterocyclylene ring can optionally be replaced by a -CO- group. More specifically the term bicyclic heterocyclylene includes, but is not limited to, hexahydrofuro[3,2-b]furan-3,6-diyl, and the like. When the heterocyclylene ring is unsaturated it can contain one or two ring double bonds provided that the ring is not aromatic.
  • “Bridged heterocyclyl” means a saturated monovalent bicyclic ring having 5 to 8 ring carbon ring atoms in which two non-adjacent ring atoms are linked by a (CR z R z’ ) n group where n is an integer selected from 1 to 3 inclusive and R z and R z ’ are independently H or methyl (also may be referred to herein as “bridging” group) and further wherein one or two ring carbon atoms, including an atom in the bridging group, is replaced by a heteroatom selected from N, O, and S(O) n , where n is an integer selected from 0 to 2 inclusive. Examples include, but are not limited to, 3,8-diazabicyclo[3.2. l]octa-3-yl, and the like.
  • “Bridged heterocyclylene” means a saturated divalent bicyclic ring having 5 to 8 ring carbon ring atoms in which two non-adjacent ring atoms are linked by a (CR z R z’ ) n group where n is an integer selected from 1 to 3 inclusive and R z and R z’ are independently H or methyl (also may be referred to herein as “bridging” group) and further wherein one or two ring carbon atoms, including an atom in the bridging group, is replaced by a heteroatom selected from N, O, and S(O) n , where n is an integer selected from 0 to 2 inclusive.
  • Bridged heterocyclylene is optionally substituted with one or two substituents independently selected from alkyl, halo, alkoxy, hydroxy, and cyano unless stated otherwise. Examples include, but are not limited to, 3,8- diazabicyclo[3.2.1]octa-3,8-diyl, and the like.
  • Cycloalkyl means a monocyclic saturated monovalent hydrocarbon radical of three to ten carbon atoms. Examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
  • Cycloalkylalkyl means an -(alkylene)-R z radical where R z is cycloalkyl as defined above. Examples include, but are not limited to, cyclopropylmethyl cyclobutylethyl, cyclopentylmethyl, cyclohexylmethyl, and the like.
  • Cycloalkylene means a divalent saturated hydrocarbon radical of three to six carbon atoms, otherwise e.g., 1,1 -cyclopropylene, 1,1 -cyclobutylene, 1 ,4-cyclohexylene, and the like.
  • Cycloalkyloxy or “cycloalkoxy” means a -OR radical where R is cycloalkyl as defined above. Examples include, but are not limited to, cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy, and the like.
  • Cyanoalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with cyano e.g., cyanomethyl, cyanoethyl, and the like.
  • Cyanoalkynyl means an alkynyl radical as defined above where one of the hydrogen atom in the alkynyl chain is replace by a cyano. Examples include, but are not limited to, -C ⁇ C(CN), -CH 2 C ⁇ C(CN), and the like.
  • Carboxy means -COOH.
  • Cyclylaminylene means a saturated divalent monocyclic ring of 4 to 8 ring atoms in which one ring atom is nitrogen, the remaining ring atoms being C. More specifically, the term cyclylaminyl includes, but is not limited to, pyrrolidinylene, piperidinylene, homopiperidinylene, and the like.
  • Deuterioalkyl mean alkyl as defined above, which is substituted with one, two, or three deuterium.
  • Deuterohaloalkyl mean haloalkyl as defined herein, which is substituted with one, two, or three deuterium.
  • Dialkylamino means -NR z’ R z’ radical where R z’ and R z" is alkyl as defined above e.g., dimethylamino, diethylamino, methylpropylamino, and the like.
  • “Fused bicyclic heterocyclyl” means a saturated monovalent fused bicyclic ring of 8 to 10 ring atoms in which one or two ring atoms are heteroatoms independently selected from N, O, and S(O) n , where n is an integer from 0 to 2, one ring atom can be -CO-, and the remaining ring atoms being C, unless stated otherwise, and where two adjacent ring atoms of the bicyclic ring are fused to two adjacent ring atoms of phenyl or a five or six membered heteroaryl, each as defined herein, unless stated otherwise.
  • fused bicyclic heterocyclyl includes, but is not limited to, 2,3-dihydro-lH-pyrrolo[2,l-a]isoindol-9b(5H)-yl, 2,3-dihydro- lH-pyrrolo[1,2-a]indol-9a(9H)-yl, 1,3b,4,5,6,8-hexahydropyrrolo[3,2-a]pyrrolizin-3b-yl, and the like.
  • “Fused bicyclic heterocyclylalkyl” means a -(alkylene)-R radical where R is fused bicyclic heterocyclyl as defined above. Examples include, but are not limited to, hexahydro- 1H- pyrrolizinylmethyl, hexahydro- IH-pyrrolizinylethyl, 2,3-dihydro-lH-pyrrolo[2, l-a]isoindol- 9b(5H)-ylmethyl, 2,3-dihydro-lH-pyrrolo[l,2-a]indol-9a(9H)-ylmethyl, and the like.
  • fused cycloalkyl as used herein, means cycloalkyl as defined above where two adjacent ring atoms of the cycloalkyl ring are fused to two adjacent ring atoms of phenyl or a five or six membered heteroaryl, each as defined herein, unless stated otherwise.
  • the fused cycloalkyl can be attached at any atom of the ring.
  • Non limiting examples of the fused cycloalkyl include bicyclo[4. 1.0]hepta-l,3,5-triene, bicyclo[4.2.0]octa-l,3,5-triene, and the like.
  • “Fused spiro cycloalkyl” means spiro cycloalkyl as defined herein where two adjacent ring atoms of the spiro cycloalkyl are fused to two adjacent ring atoms of a phenyl or a five or six membered heteroaryl, each as defined herein.
  • “Fused heterocyclyl” as used herein means a saturated monovalent monocyclic ring of 4 to 7 ring atoms having from one to three heteroatoms independently selected from N, O, and S and the remaining ring atoms being carbon, and further wherein two adjacent ring atoms of the monocyclic ring is fused to two adjacent ring members of a phenyl or a five or six membered heteroaryl, each as defined herein, unless stated otherwise.
  • the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quatemized and one or two carbon atoms of the fused ring atoms in the saturated monocyclic ring includes the two common ring vertices shared with the fused phenyl or five or six membered heteroaryl.
  • the fused heterocyclyl can be attached at any atom of the ring.
  • Non limiting examples of the fused heterocycloalkyl include 2,3-dihydrobenzo[b][l,4]-dioxinyl, 2-oxabicyclo[3.1.0]hexanyl, indolin-2-one- 1 -yl, indolinyl, and the like.
  • “Fused heterocyclylene” as used herein refers to a divalent bicyclic ring in which two adjacent ring atoms of a saturated monocyclic ring of 4 to 7 ring atoms having one or two heteroatoms independently selected from N, O, and S(O)n (where n is 0, 1, or 2) and the remaining ring atoms being carbon, are fused to two adjacent ring members of a phenyl or a five or six membered heteroaryl, each as defined herein, unless stated otherwise.
  • the nitrogen atom is optionally oxidized or quatemized 1.
  • the fused heterocyclylene can be attached at any two atoms of the ring.
  • Representative examples include, but are not limited to, 1,2,3,4-tetrahydroquinolin- 1,4-diyl, 3,4-dihydro-2H-benzo[b][l,4]oxazin-5,8-diyl, and the like.
  • “Fused heterocyclylalkyl” as used herein, means a -(alkylene)-R radical where R is fused heterocyclyl, as defined herein.
  • “Fused heteroaryl” means fused bicyclic heteroaryl, as defined herein, where two adjacent ring atoms of the heteroaryl ring are fused to two adjacent ring atoms of phenyl. “Halo” means fluoro, chloro, bromo, or iodo, preferably fluoro or chloro.
  • Haloalkyl means alkyl radical as defined above, which is substituted with one or more halogen atoms, e.g., one to five halogen atoms, such as fluorine or chlorine, including those substituted with different halogens, e.g., -CH2CI, -CF3, -CHF2, -CH2CF3, -CF2CF3, -CF(CH 3 )2, and the like.
  • halogen atoms e.g., one to five halogen atoms, such as fluorine or chlorine, including those substituted with different halogens, e.g., -CH2CI, -CF3, -CHF2, -CH2CF3, -CF2CF3, -CF(CH 3 )2, and the like.
  • fluoroalkyl When the alkyl is substituted with only fluorine atom(s), it can be referred to in this Application as fluoroalkyl.
  • Haloalkoxy means a -OR Z radical where R z is haloalkyl as defined above e.g., -OCF3, -OCHF2, and the like.
  • R z is haloalkyl where the alkyl is substituted with only fluorine atom(s), it is referred to in this Application as fluoroalkoxy.
  • Haloalkoxyalkyl means a -(alkylene)OR z radical where R z is haloalkyl as defined above, e.g., trifluoromethoxyalkyl, and the like.
  • Halocarbonyl means a -C(O)R Z radical where R z is haloalkyl, as defined herein, e.g., trifluoromethylcarbonyl, difluoromethylcarbonyl, and the like.
  • Hydroalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with one or two hydroxy groups, provided that if two hydroxy groups are present, they are not both present on the same carbon atom.
  • Representative examples include, but are not limited to, hydroxymethyl, 2-hydroxy-ethyl, 2-hydroxypropyl, 3 -hydroxypropyl, l-(hydroxymethyl)-2-methylpropyl, 2-hydroxybutyl, 3 -hydroxybutyl, 4-hydroxybutyl, 2,3-dihydroxypropyl, 1 -(hydroxymethyl)-2- hydroxyethyl, 2,3-dihydroxybutyl, 3,4-dihydroxybutyl and 2-(hydroxymethyl)-3-hydroxypropyl, preferably 2-hydroxyethyl, 2,3-dihydroxypropyl, and l-(hydroxymethyl)-2-hydroxyethyl.
  • Heteroaryl means a monovalent monocyclic or fused bicyclic aromatic radical of 5 to 10 ring atoms, unless otherwise stated, where one or more, (in one embodiment, one, two, or three), ring atoms are heteroatom selected from N, O, and S, the remaining ring atoms being carbon.
  • Representative examples include, but are not limited to, pyrrolyl, thienyl, thiazolyl, imidazolyl, furanyl, indolyl, isoindolyl, oxazolyl, isoxazolyl, benzothiazolyl, benzoxazolyl, quinolinyl, isoquinolinyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, tetrazolyl, and the like.
  • the terms “heteroaryl” and “aryl” are mutually exclusive.
  • heteroaryl ring contains 5 or 6 ring atoms and is a monocyclic ring, it is also referred to herein as 5 -or 6-membered monocyclic heteroaryl or monocyclic heteroaryl.
  • heteroaryl ring contains 9- or 10 ring atoms and is a fused bicyclic ring, it is also referred to herein as 9-or 10- membered fused bicyclic heteroaryl.
  • Heteroaralkyl means a -(alkylene)-R radical where R is heteroaryl as defined above, e.g., pyridinylmethyl, and the like.
  • heteroaryl ring in heteroaralkyl contains 5- or 6 ring atoms it is also referred to herein as 5 -or 6-membered heteroaralkyl.
  • Heteroarylene means a divalent heteroaryl radical as defined above, unless stated otherwise. Representative examples include, but are not limited to, benzimidazoldiyl e.g., benzimidazole-l,5-diyl, and the like.
  • the heteroarylene ring contains 5 or 6 ring atoms and is a monocyclic ring and is also referred to herein as monocyclic heteroarylene or as 5 -or 6-membered monocyclic heteroarylene e.g., pyrazolyl-1.4-diyl.
  • the heteroarylene ring contains 9 or 10 ring atoms and is a fused bicyclic ring, it is also referred to herein as 9-or 10-membered fused bicyclic heteroarylene.
  • Heteroaralkyl means a -(alkylene)-R z radical where R z is heteroaryl as defined above, e.g., pyridinylmethyl, and the like.
  • R z is heteroaryl as defined above, e.g., pyridinylmethyl, and the like.
  • the heteroaryl ring in heteroaralkyl contains 5 or 6 ring atoms it is also referred to herein as 5 -or 6-membered heteroaralkyl or monocyclic heteroaralkyl.
  • Heterocyclyl means a saturated or unsaturated monovalent monocyclic group of 4 to 8 ring atoms in which one or two ring atoms are heteroatom independently selected from N, O, and S(O) n , where n is an integer selected from 0 to 2 inclusive, the remaining ring atoms being C, unless stated otherwise. Additionally, one or two ring carbon atoms in the heterocyclyl ring can optionally be replaced by a -CO- group.
  • heterocyclyl includes, but is not limited to, pyrrolidino, piperidino, homopiperidino, 2-oxopyrrolidinyl, 2-oxopiperidinyl, morpholino, piperazino, tetrahydro-pyranyl, thiomorpholino, and the like.
  • heterocyclyl ring is unsaturated it can contain one or two ring double bonds provided that the ring is not aromatic.
  • heterocyclyl group contains at least one nitrogen atom, it is also referred to herein as heterocycloamino and is a subset of the heterocyclyl group.
  • Heterocyclylalkyl or “heterocycloalkyl” means a -(alkylene)-R z radical where R z is heterocyclyl ring as defined above e.g., tetraydrofiiranylmethyl, piperazinylmethyl, morpholinylethyl, and the like.
  • Heterocyclylene means a saturated divalent monocyclic group of 4 to 6 ring atoms in which one or two ring atoms are heteroatom independently selected from N, O, and S(O) n , where n is an integer selected from 0 to 2 inclusive, the remaining ring atoms being C, unless stated otherwise. Additionally, one or two ring carbon atoms in the heterocyclylene ring can optionally be replaced by a -CO- group. More specifically, the term heterocyclylene includes, but is not limited to, , piperidin- 1 ,4-diyl, azetidin- 1,3 -diyl, and the like.
  • Heteroalkyl mean alkyl radical as defined above wherein one or two carbon atoms are replaced by O, NR (R is H or alkyl), or S, provided the heteroalkyl group is attached to the remainder of the molecule via a carbon atom, e.g., methoxymethyl, methylethylaminoethyl, and the like.
  • Phenylene refers to divalent phenyl.
  • Optionally substituted aryl means aryl as defined above, that is optionally substituted with one, two, or three substituents independently selected from alkyl, hydroxyl, cycloalkyl, carboxy, alkoxycarbonyl, hydroxy, alkoxy, alkylthio, alkylsulfonyl, amino, alkylamino, dialkylamino, halo, haloalkyl, haloalkoxy, and cyano.
  • optionally substituted aryl is optionally substituted phenyl.
  • Optionally substituted aralkyl means -(alkylene)-R z where R z is optionally substituted aryl as defined above.
  • Optionally substituted heteroaryl means heteroaryl as defined above that is optionally substituted with one, two, or three substituents independently selected from alkyl, alkylthio, alkylsulfonyl, hydroxyl, cycloalkyl, carboxy, alkoxycarbonyl, hydroxy, alkoxy, halo, haloalkyl, haloalkoxy, amino, alkylamino, dialkylamino, and cyano.
  • Optionally substituted heteroaralkyl means -(alkylene) -R z where R z is optionally substituted heteroaryl as defined above.
  • Optionally substituted heterocyclyl means heterocyclyl as defined above that is optionally substituted with one, two, or three substituents independently selected from alkyl, alkylthio, alkylsulfonyl, alkylcarbonyl, hydroxyl, cycloalkyl, cycloalkylalkyl, carboxy, alkoxycarbonyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, cyanoalkyl, halo, haloalkyl, haloalkoxy, and cyano, unless stated otherwise.
  • Optionally substituted heterocyclylalkyl means -(alkylene)-R z where R z is optionally substituted heterocyclyl as defined above.
  • heteroaryl optionally substituted with alkyl is intended to cover heteroaryl that is unsubstituted with alkyl and heteroaryl that is substituted with alkyl.
  • “Spiro cycloalkyl” means a saturated bicyclic monovalent ring having 5 to 10 ring atoms in in which the rings are connected through only one atom, the connecting atom is also called the spiroatom, most often a quaternary carbon ("spiro carbon"). Unless stated otherwise, spiro cycloalkyl is optionally substituted with one or two substituents independently selected from alkyl, halo, alkoxy, hydroxy, and cyano. Examples include, but are not limited to, Representative examples include, but are not limited to, spiro[3.3]heptane, spiro[3.4]octane, spiro [3.5] -nonane, and the like.
  • “Spiro cycloalkylene” means a saturated bicyclic divalent hydrocarbon ring having 6 to 12 ring atoms wherein the rings are connected through only one atom, the connecting atom is also called the spiroatom, most often a quaternary carbon (“spiro carbon”). Spiro cycloalkylene is optionally substituted with one or two substituents independently selected from alkyl, halo, alkoxy, hydroxy, and cyano, unless stated otherwise. Representative examples include, but are not limited to, spiro[3,5]nonandiyl e.g., spiro[3.5]nonane-2,7-diyl, and the like.
  • “Spiro heterocyclyl” means a saturated bicyclic monovalentvalent ring having 6 to 10 ring atoms in which one, two, or three ring atoms are heteroatom selected from N, O, and S(O) n , where n is an integer selected from 0 to 2 inclusive, the remaining ring atoms being C and the rings are connected through only one atom, the connecting atom is also called the spiroatom, most often a quaternary carbon (“spiro carbon”).
  • “Spiro heterocyclylene” means a saturated bicyclic divalent ring having 6 to 10 ring atoms in which one, two, or three ring atoms are heteroatom selected from N, O, and S(O) n , where n is an integer selected from 0 to 2 inclusive, the remaining ring atoms being C and the rings are connected through only one atom, the connecting atom is also called the spiroatom, most often a quaternary carbon (“spiro carbon”).
  • Tricyclic heterocyclyl means a saturated monovalent fused tricyclic ring of 9 to 14, preferably 12 to 14, ring atoms in which one, two, or three ring atoms are heteroatom independently selected from N, O, and S(O) n , where n is an integer from 0 to 2, one ring atom can be -CO-, and the remaining ring atoms being C, unless stated otherwise. Additionally, one or two ring carbon atoms in the heterocyclyl ring can optionally be replaced by a -CO- group.
  • tricyclic heterocyclyl includes, but is not limited to, , and the like.
  • Tricyclic heterocyclylalkyl means a -(alkylene)-R radical where R is tricyclic heterocyclyl as defined above. Examples include, but are not limited to,
  • the present disclosure also includes protected derivatives of compounds of Formula (IA) or (I).
  • compounds of Formula (IA) or (I) when compounds of Formula (IA) or (I) contain groups such as hydroxy, carboxy, or any group containing a nitrogen atom(s), these groups can be protected with suitable protecting groups.
  • suitable protecting groups A comprehensive list of suitable protective groups can be found in T.W. Greene, Protective Groups in Organic Synthesis, 5 th Ed., John Wiley & Sons, Inc. (2014), the disclosure of which is incorporated herein by reference in its entirety.
  • the protected derivatives of compounds of the present disclosure can be prepared by methods well known in the art.
  • the present disclosure also includes polymorphic forms and deuterated forms of the compound of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof.
  • prodrug refers to a compound that is made more active in vivo.
  • Certain compounds Formula (IA) or (I) may also exist as prodrugs, as described in Hydrolysis in Drug and Prodrug Metabolism: Chemistry, Biochemistry, and Enzymology (Testa, Bernard and Mayer, Joachim M. Wiley- VHCA, Zurich, Switzerland 2003).
  • Prodrugs of the compounds described herein are structurally modified forms of the compound that readily undergo chemical changes under physiological conditions to provide the active compound. Prodrugs are often useful because, in some situations, they may be easier to administer than the compound, or parent drug. They may, for instance, be bioavailable by oral administration whereas the parent drug is not.
  • prodrug derivatives are known in the art, such as those that rely on hydrolytic cleavage or oxidative activation of the prodrug.
  • An example, without limitation, of a prodrug would be a compound which is administered as an ester (the “prodrug”), but then is metabolically hydrolyzed to the carboxylic acid, the active entity. Additional examples include peptidyl derivatives of a compound.
  • a “pharmaceutically acceptable salt” of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound.
  • Such salts include: acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as formic acid, acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1 ,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzene
  • the compounds of Formula (IA) or (I) (and any embodiment thereof disclosed herein including specific compounds) may have asymmetric centers.
  • Compounds of Formula (IA) or (I) (and any embodiment thereof disclosed herein including specific compounds) containing an asymmetrically substituted atom may be isolated in optically active or racemic forms.
  • Individual stereoisomers of compounds can be prepared synthetically from commercially available starting materials which contain chiral centers or by preparation of mixtures of enantiomeric products followed by separation such as conversion to a mixture of diastereomers followed by separation or recrystallization, chromatographic techniques, direct separation of enantiomers on chiral chromatographic columns, or any other appropriate method known in the art.
  • Certain compounds of Formula (IA) or (I) can exist as tautomers and/or geometric isomers. All possible tautomers and cis and trans isomers, as individual forms and mixtures thereof are within the scope of this disclosure.
  • alkyl includes all the possible isomeric forms of said alkyl group albeit only a few examples are set forth.
  • the cyclic groups such as aryl is substituted, it includes all the positional isomers albeit only a few examples are set forth.
  • all hydrates of a compound of Formula (IA) or (I) (and any embodiment thereof disclosed herein including specific compounds) are within the scope of this disclosure.
  • the compounds of Formula (IA) or (I) may also contain unnatural amounts of isotopes at one or more of the atoms that constitute such compounds.
  • Unnatural amounts of an isotope may be defined as ranging from the amount found in nature to an amount 100% of the atom in question that differ only in the presence of one or more isotopically enriched atoms.
  • Exemplary isotopes that can be incorporated into compounds of the present disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, and iodine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 32 P, 33 P, 35 S, 18 F, 36 C1, 123 I, and 125 1, respectively.
  • Isotopically labeled compounds e.g., those labeled with 3 H and 14 C
  • Tritiated (i.e., 3 H) and carbon-14 (i.e., 14 C) isotopes can be useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements).
  • one or more hydrogen atoms are replaced by 2 H or 3 H, or one or more carbon atoms are replaced by 13 C- or 14 C- enriched carbon.
  • Positron emitting isotopes such as 15 O, 13 N, 11 C, and 15 F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy.
  • Isotopically labeled compounds can generally be prepared by following procedures analogous to those disclosed in the Schemes or in the Examples herein, by substituting an isotopically labeled reagent for a non- isotopically labeled reagent.
  • a “pharmaceutically acceptable carrier or excipient” means a carrier or an excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes a carrier or an excipient that is acceptable for veterinary use as well as human pharmaceutical use. “A pharmaceutically acceptable carrier/excipient” as used in the specification and claims includes both one and more than one such excipient.
  • alkylene optionally substituted with one or two fluoro in the definition of X 1 , X 2 , X 3 ’ and X 4 in Formula (IA) or (I) (and any embodiment thereof disclosed herein) (and similar phrases used to define other groups in Formula (IA) or (I)) is intended to cover alkylene that is unsubstituted and alkylene that is substituted one or two fluoro.
  • R 2 and R 3 groups are floating substituents and can replace the hydrogen atom of any one of U, V, and W of the portion of the quinazoline ring ring when U, V, and W are CH; and in the structure the R aa substituent of R aa , R bb and X 1 , and similarly the R bb and X 1 substituents, can replace hydrogen of any CH that is part of the benzo portion of the bicyclic ring that is not already substituted with R bb and X 1 , and similarly R aa and X 1 , and R aa and R bb substituents with respect to R bb and X 1 , respectively.
  • the left bond of the divalent group is attached to the group which is to its left in the remainder of the molecule
  • the right bond of the divalent group is attached to the group which is to its right in the remainder of the molecule.
  • the bond on the left of (a), (b) and (c) is attached to Z 6 of L in the following structure : and the on the right side of (a), (b), and (c) (i.e., X 1 , X 2 , and X 3 ) is attached to ring:
  • L i.e, -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -
  • Z 1 the left side in L (i.e., Z 1 ) is attached to an atom of an R 4 group as denoted in this Application and Z 6 is attached to X 1 , X 2 , X 3 , or X 4 .
  • L is a group of formula: and Degron is a group of formula (a), i.e., the left bond of L (i.e., the -NH- group) is attached to X 2 and the right hand bond of L (i.e., -SO2-) is attached to an R 4 group as denoted in this Application.
  • disease as used herein is intended to be generally synonymous, and is used interchangeably with, the terms “disorder,” “syndrome,” and “condition” (as in medical condition), in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptoms, and causes the human or animal to have a reduced duration or quality of life.
  • combination therapy means the administration of two or more therapeutic agents to treat a disease or disorder described in the present disclosure. Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each active ingredient. In addition, such administration also encompasses use of each type of therapeutic agent in a sequential manner. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the conditions or disorders described herein.
  • patient is generally synonymous with the term “subject” and includes all mammals including humans. Examples of patients include humans, livestock such as cows, goats, sheep, pigs, and rabbits, and companion animals such as dogs, cats, rabbits, and horses. Preferably, the patient is a human.
  • Treating” or “treatment” of a disease includes:
  • treating or treatment of a disease includes inhibiting the disease, i.e., delaying, arresting or reducing the development or severity of the disease or its clinical symptoms; or relieving the disease, i.e., causing regression of the disease or its clinical symptoms.
  • a “therapeutically effective amount” means the amount of a compound of the present disclosure or a pharmaceutically acceptable salt thereof that, when administered to a patient for treating a disease, is sufficient to affect such treatment for the disease.
  • the “therapeutically effective amount” will vary depending on the compound, the disease and its severity and the age, weight, etc., of the mammal to be treated.
  • the therapeutically effective amount of a K-ras inhibitor disclosed herein can be administered to the patient in a single dosage form or multiples thereof. For example, 600 mg dose of a K-ras inhibitor can be administered in a single 600 mg tablet or two 300 mg tablets.
  • K-Ras G12D includes any measurable decrease or complete inhibition to achieve a desired result. For example, there may be a decrease of about, at most about, or at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or more, or any range derivable therein, reduction of K-Ras G12D GTPase activity; a decrease of K- Ras G12D GTP binding affinity or an increase of G12D GDP binding affinity; an increase of GTP off rate or a decrease of GDP off rate; a decrease of signaling transduction molecules levels downstream in the K-Ras pathway, e.g., a decrease in pERK level; and/or a decrease of K-Ras complex binding to downstream signaling molecules compared to normal.
  • the present disclosure includes:
  • the compound of Formula (IA) or a pharmaceutically acceptable salt thereof is a compound according to structure (IA’): where R 1 , R 1a , R 1b , R 2 , R 3 , R 4 , R 5 , n, m, L, U, V, W, and Degron are as defined in the first aspect of the Summary.
  • the compound of Formula (I) or a pharmaceutically acceptable salt thereof is a compound according to structure (la): where R 1 , R 1a , R 1b , R 2 , R 3 , R 4 , R 5 , n, m, L, U, V, W, and Degron are as defined in the second aspect of the Summary. la.
  • the compound of embodiment IA or 1, a pharmaceutically acceptable salt is a compound according to structure (lb): where R 1 , R 1a , R 1b , R 2 , R 3 , R 4 , R 5 , n, m, L and Degron are as defined in the first or second aspect of the Summary, respectivley. lb.
  • the compound of embodiment 1 A or 1, or a pharmaceutically acceptable salt is a compound according to structure (Ic): where R 1 , R 1a , R 1b , R 2 , R 3 , R 4 , R 5 , n, m, L and Degron are as defined in the first or second aspect of the Summary, respectively.
  • the compound of embodiment 1A, 1, la, or lb, or a pharmaceutically acceptable salt thereof is wherein R 6 is fused heterocyclylalkyl where fused heterocyclyl of fused heterocyclylalkyl is substituted with R a , R b , and R c as defined therein.
  • the compound of embodiment 1 A, 1, la, or lb, or a pharmaceutically acceptable salt thereof is wherein the fused heterocyclyl of fused heterocyclylalkyl of R 6 is isoindolinyl substituted with R a , R b , and R c as defined therein.
  • the compound of any one of embodiments 1 A to 3, or a pharmaceutically acceptable salt thereof, is wherein the fused heterocyclylalkyl of R 6 is a ring of formula: are as defined therein.
  • the compound of embodiment 1A, 1, la, or lb, or a pharmaceutically acceptable salt thereof is wherein R 6 is heterocyclylalkyl, bicyclic heterocyclyl, or bicyclic heterocylalkylalkyl, where heterocyclyl of heterocyclylalkyl and bicyclic heterocyclyl, by itself or as part of bicyclic heterocyclylalkyl are substituted with R d , R e , and R f as defined therein.
  • the compound of embodiment 1A, 1, 1a, lb, or 5, or a pharmaceutically acceptable salt thereof is wherein R 6 is heterocyclylalkyl where heterocyclyl of heterocyclylalkyl of R 6 is substituted with R d , R e , and R f as defined therein. 7.
  • the compound of any one of embodiments 1A, 1, la, lb, 5 and 6, or a pharmaceutically acceptable salt thereof is wherein the heterocyclylalkyl of R 6 is pyrrolidin-2- ylmethyl, piperidin-2-ylmethyl, or piperi din-3 -ylmethyl, preferably pyrrolidin-2-ylmethyl substituted with R d , R e , and R f as defined therein.
  • the compound of any one of embodiments 1A, 1, 1a, lb, and 5 to 7, or a pharmaceutically acceptable salt thereof is wherein the heterocyclylalkyl of R 6 is pyrrolidin-2- ylmethyl of structure substituted with R e and R f , preferably R 6 is pyrrolidin-2-ylmethyl of structure hydrogen.
  • the compound of any one of embodiments 1A, 1, 1a, lb, and 5, or a pharmaceutically acceptable salt thereof, is wherein R 6 is bicyclic heterocyclalkylalkyl substituted with R d , R e , and R f as defined therein.
  • the compound of any one of embodiments 1A, 1, 1a, lb, 5, and 9, or a pharmaceutically acceptable salt thereof is wherein the bicyclic heterocyclylalkyl of R 6 is hexahydro- 1 H-pyrrolizin-7 a-ylalkyl-d2, preferably, hexahydro- 1 H-pyrrolizin-7a-ylmethyl-d2, where hexahydro- lH-pyrrolizin-7a-yl is substituted with R d , R e , and R f as defined therein.
  • the compound of any one of embodiments 1A, 1, 1a, lb, 5, 9, and 10, or a pharmaceutically acceptable salt thereof, is wherein the bicyclic heterocyclylalkyl is a ring of formula: substituted with R e , and R f as defined therein. 12.
  • R 11 is wherein the bicyclic heterocyclylalkyl of R 6 is a ring of formula: where R d , R e , R f , are as defined therein.
  • the compound of any one of embodiments 1A, 1, la, lb, 5, and 9 to 12, or a pharmaceutically acceptable salt thereof is wherein the bicyclic heterocyclylalkyl of R 6 is a ring of formula:
  • R d and R f are as defined therein.
  • the compound of embodiment 1A, 1, la, or lb, or a pharmaceutically acceptable salt thereof is wherein R 6 is fused bicyclic heterocyclyl, fused bicyclic heterocyclylalkyl, heterocyclyl fused bicyclic heterocyclyl, heterocyclyl fused bicyclic heterocyclylalkyl, tricyclic heterocyclyl, or tricyclic heterocyclylalkyl, wherein fused bicyclic heterocyclyl, by itself or as part of fused bicyclic heterocyclylalkyl, heterocyclyl fused bicyclic heterocyclyl, by itself or as part of heterocyclyl fused bicyclic heterocyclylalkyl, or tricyclic heterocyclyl, by itself or as part of tricyclic heterocyclylalkyl are independently substituted with R g , R h , and R 1 as defined therein.
  • the compound of embodiment 1A, 1, la, lb, or 14, or a pharmaceutically acceptable salt thereof is wherein R 6 is fused bicyclic heterocyclyl substituted with R g , R h , , and R 1 as defined therein.
  • the compound of embodiment 1A, 1, la, lb, or 15, or a pharmaceutically acceptable salt thereof is wherein the fused bicyclic heterocyclyl of R 6 is a ring of formula: where ring Al is phenyl or 5- or 6-membered heteroaryl and the fused bicyclic heterocyclyl is additionally substituted with R h and R 1 where R g , R h and R 1 are as defined as defined therein, preferably ring Al is phenyl or 5- or 6-membered heteroaryl substituted with R h and R 1 as defined therein.
  • the compound of embodiment 1A, 1, la, lb, 15, or 16, or a pharmaceutically acceptable salt thereof is wherein the fused bicyclic heterocyclyl of R 6 is a ring of formula: where ring Al is phenyl, pyrazolyl, pyridinyl, or pyrimidinyl, each ring substituted with R h and R 1 as defined therein.
  • the compound of any one of embodiments 1A, 1, la, lb, and 14, or a pharmaceutically acceptable salt thereof is wherein R 6 is fused bicyclic heterocylalkylalkyl where fused bicyclic heterocyclyl of fused bicyclic heterocyclylalkyl is substituted with R g , R h , and R 1 as defined therein.
  • the compound of embodiment 1A, 1, la, lb, or 14, or a pharmaceutically acceptable salt thereof is wherein fused bicyclic heterocyclyl of fused bicyclic heterocyclylalkyl of R 6 is a ring of formula: where ring Al is phenyl or 5- or 6-membered heteroaryl and the fused bicyclic heterocyclyl is additionally substituted with R h and R 1 where R g , R h and R 1 are as defined as defined therein.
  • the compound of embodiment 1A, 1, la, lb, 14, or 19, or a pharmaceutically acceptable salt thereof is wherein the fused bicyclic heterocyclyl of fused bicyclic heterocyclylalkyl of R 6 is a ring of formula: where ring Al is phenyl, pyrazolyl, pyridinyl, or pyrimidinyl, each ring substituted with R b and R 1 as defined therein.
  • the compound of any one of embodiments 1A, 1, la, lb, and 14 to 20, or a pharmaceutically acceptable salt thereof is wherein the fused bicyclic heterocyclyl and fused bicyclic heterocyclyl of fused bicyclic heterocyclylalkyl of R 6 is 2,3 -dihydro- lH-pyrrolo[2,l- a]isoindol-9b(5H)-yl, 2,3-dihydro-lH-pyrrolo[l,2-a]indol-9a(9H)-yl, 1,3b, 4,5,6, 8-hexahydropyrrolo- [3,2-a]pyrrolizin-3b-yl, l-methyl-l,3b,4,5,6,8-hexahydropyrrolo[4,3-a]pyrrolizin-3b-yl, 4b, 6,7,9- tetrahydro-5H-pyrido[3,2-a]-pyrrolizin-4b
  • the compound of any one of embodiments 1A, 1, la, lb, and 14, or a pharmaceutically acceptable salt thereof is wherein R 6 is tricyclic heterocyclyl substituted with R g , R b , and R 1 as defined therein.
  • the compound of any one of embodiments 1A, 1, la, lb, and 14, or a pharmaceutically acceptable salt thereof is wherein R 6 is tricyclic heterocyclylalkyl where tricyclic heterocyclyl of tricyclic heterocyclylalkyl is substituted with R g , R b , and R 1 as defined therein.
  • the compound of any one of embodiments 1 A to 23, or a pharmaceutically acceptable salt thereof is wherein R 7 , R 9 , and R 11 are independently hydrogen, deuterium, alkyl, fluoro, or haloalkyl, R b , R e , and R b are a bond to L, and R c , R f , and R 1 are independently hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, or hydroxy.
  • the compound of any one of embodiments 1 A to 23, or a pharmaceutically acceptable salt thereof is wherein R 7 , R 9 , and R 11 are independently hydrogen, deuterium, alkyl, fluoro, or haloalkyl, R b , R e , and R b are independently hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, hydroxy, -(alkylene) n1 -OC(O)NR 13 R 13a and R c , R f , and R 1 are independently a bond to L.
  • the compound of any one of embodiments 1A to 23, or a pharmaceutically acceptable salt thereof is wherein R 7 , R 9 , and R 11 are independently a bond to L, R b , R e , and R b are independently hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, hydroxy, or -(alkyl ene) n i- OC(O)NR 13 R 13a and R c , R f , and R 1 are independently hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, or hydroxy.
  • the compound of any one of embodiments 1A to 25, or a pharmaceutically acceptable salt thereof is wherein R 7 , R 9 , and R 11 are independently hydrogen, deuterium, methyl, ethyl, propyl, fluoro, difluoromethyl, or trifluoromethyl.
  • the compound of any one of embodiments 1A to 25, and 27, or a pharmaceutically acceptable salt thereof, is wherein R 7 , R 9 , and R 11 are independently hydrogen.
  • the compound of any one of embodiments 1A to 25, and 27, or a pharmaceutically acceptable salt thereof, is wherein R 7 , R 9 , and R 11 are independently deuterium.
  • the compound of any one of embodiments 1A to 25, or a pharmaceutically acceptable salt thereof is wherein R 7 , R 9 , and R 11 are independently alkyl, preferably methyl, ethyl, or propyl, preferably methyl.
  • the compound of any one of embodiments 1 A to 25, and 27, or a pharmaceutically acceptable salt thereof, is wherein R 7 , R 9 , and R 11 are independently fluoro.
  • the compound of any one of embodiments 1A to 25, or a pharmaceutically acceptable salt thereof is wherein R 7 , R 9 , and R 11 are independently haloalkyl, preferably fluoromethyl, difluoromethyl, or trifluoromethyl, preferably trifluoromethyl.
  • the compound of any one of embodiments 1A to 32, or a pharmaceutically acceptable salt thereof is wherein R 8 , R 10 , and R 12 are independently hydrogen, deuterium, alkyl, halo, haloalkyl, cyano, alkyloxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, phenyl, or heteroaryl.
  • the compound of any one of embodiments 1A to 33, or a pharmaceutically acceptable salt thereof is wherein R 8 , R 10 , and R 12 are independently hydrogen, deuterium, fluoro, methyl, cyano, methoxymethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclopropylethyl, cyclobutylethyl, cyclopentylethyl, phenyl, pyrrolidinyl, furanyl, pyranyl, piperidinyl, morpholinyl, or 5- or 6- membereing heteroaryl (such as pyridyl, pyrimidinyl, or pyrazolyl).
  • R 8 , R 10 , and R 12 are independently hydrogen, deuterium, fluoro, methyl, cyano, methoxymethyl, cyclopropyl, cyclobutyl,
  • the compound of any one of embodiments 1A to 34, or a pharmaceutically acceptable salt thereof is wherein R 8 , R 10 , and R 12 are independently hydrogen.
  • the compound of any one of embodiments 1A to 34, or a pharmaceutically acceptable salt thereof is wherein R 8 , R 10 , and R 12 are independently fluoro ormethyl.
  • the compound of any one of embodiments 1A to 34, or a pharmaceutically acceptable salt thereof is wherein R 8 , R 10 , and R 12 are independently hydrogen or cyano.
  • the compound of any one of embodiments 1A to 34, or a pharmaceutically acceptable salt thereof is wherein R 8 , R 10 , and R 12 are methoxymethyl.
  • the compound of any one of embodiments 1A to 34, or a pharmaceutically acceptable salt thereof is wherein R 8 , R 10 , and R 12 are independently cyclopropyl, cyclobutyl, cyclopentyl, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclopropylethyl, cyclobutylethyl, or cyclopentylethyl.
  • the compound of any one of embodiments 1A to 34, or a pharmaceutically acceptable salt thereof is wherein R 8 , R 10 , and R 12 are phenyl, pyrrolidinyl, furanyl, pyranyl, piperidinyl, morpholinyl, or 5- or 6-membereing heteroaryl (such as pyridyl, pyrimidinyl, or pyrazolyl).
  • the compound of any one of embodiments 1 A to 23, or a pharmaceutically acceptable salt thereof is wherein, independently of each other, R 7 andR 8 , R 9 and R 10 , and R 11 and R 12 together with the carbon atom to which they are attached form cycloalkylene optionally substituted with alkyl, halo, alkoxy, or hydroxy, R b , R e , and R b are a bond to L, and R c , R f , and R 1 are independently hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, or hydroxy.
  • the compound of any one of embodiments 1A to 23, or a pharmaceutically acceptable salt thereof is wherein, independently of each other, R 7 andR 8 , R 9 and R 10 , and R 11 and R 12 together with the carbon atom to which they are attached form cycloalkylene optionally substituted with alkyl, halo, alkoxy, or hydroxy, R b , R e , and R b are independently hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, hydroxy, -(alkylene) n1 -OC(O)NR 13 R 13a and R c , R f , and R 1 are independently a bond to L.
  • the compound of any one of embodiments 1A to 23, 41, and 42, or a pharmaceutically acceptable salt thereof is wherein R 7 and R 8 , R 9 and R 10 , and R 11 and R 12 together with the carbon atom to which they are attached form cyclopropyl, cyclobutylene, or cyclopentylene, each ring optionally substituted with methyl, fluoro, or methoxy.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, and 43, or a pharmaceutically acceptable salt thereof is wherein R b , R e , and R b are independently hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, or hydroxy.
  • the compound of any one of embodiments 1A to 23, 25 to 40, and 42 to 44, or a pharmaceutically acceptable salt thereof is wherein R b , R e , and R b are independently hydrogen, methyl, fluoro, methoxy, ethoxy, or methoxymethyl.
  • the compound of any one of embodiments 1A to 23, 25 to 40, and 42 to 45, or a pharmaceutically acceptable salt thereof, is wherein R b , R e , and R b are hydrogen.
  • the compound of any one of embodiments 1A to 23 and 25 to 45, or a pharmaceutically acceptable salt thereof is wherein R b , R e , and R b are alkyl, preferably methyl.
  • the compound of any one of embodiments 1 A to 23 and 25 to 45, or a pharmaceutically acceptable salt thereof is wherein R b , R e , and R b are halo, preferably fluoro.
  • the compound of any one of embodiments 1A to 23 and 25 to 45, or a pharmaceutically acceptable salt thereof is wherein R b , R e , and R b are alkoxy, preferably methoxy or ethoxy.
  • the compound of any one of embodiments 1A to 23, 25 to 40, and 42 to 45, or a pharmaceutically acceptable salt thereof, is wherein R b , R e , and R b are alkoxyalkyl, preferably methoxymethyl.
  • the compound of any one of embodiments 1 A to 23, 25 to 40, 42 and 43, or a pharmaceutically acceptable salt thereof is wherein R b , R e , and R b are independently - ( alkyl ene)ni-OC(O)NR 13 R 13a .
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51, or a pharmaceutically acceptable salt thereof, is wherein nl is 0.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51, or a pharmaceutically acceptable salt thereof, is wherein nl is 1.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51, and 53, or a pharmaceutically acceptable salt thereof is wherein alkylene is methylene, ethylene, -CH(CH3)-, or -C(CH3)2-, preferably methylene.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 54, or a pharmaceutically acceptable salt thereof is wherein R 13 is hydrogen, deuterium, alkyl, deuterioalkyl, haloalkyl, haloalkoxyalkyl, or alkoxyalkyl and R 13a is deuterioalkyl, cycloalkyl, alkoxy, alkoxyalkyl, haloalkyl, haloalkoxyalkyl, or heterocyclyl optionally substituted with one or two substituents independently selected from alkyl, halo, alkoxy, haloalkoxy, and haloalkyl.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 55, or a pharmaceutically acceptable salt thereof, is wherein R 13 is hydrogen or deuterium.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 55, or a pharmaceutically acceptable salt thereof, is wherein R 13 is alkyl.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 55, or a pharmaceutically acceptable salt thereof, is wherein R 13 is haloalkyl.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 55, or a pharmaceutically acceptable salt thereof, is wherein R 13 is haloalkoxyalkyl.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 55, or a pharmaceutically acceptable salt thereof, is wherein R 13 is alkoxyalkyl.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 55, or a pharmaceutically acceptable salt thereof, is wherein R 13 is deuterioalkyl.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 61, or a pharmaceutically acceptable salt thereof, is wherein R 13a is alkoxy.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 61, or a pharmaceutically acceptable salt thereof, is wherein R 13a is alkoxyalkyl.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 61, or a pharmaceutically acceptable salt thereof, is wherein R 13a is haloalkyl.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 61, or a pharmaceutically acceptable salt thereof, is wherein R 13a is haloalkoxyalkyl.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 61, or a pharmaceutically acceptable salt thereof, is wherein R 13a is cycloalkyl.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 61, or a pharmaceutically acceptable salt thereof, is wherein R 13a is deuterioalkyl.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 61, or a pharmaceutically acceptable salt thereof is wherein R 13a is heterocyclyl substituted with Ri and R k independently selected from alkyl, halo, alkoxy, haloalkoxy, and haloalkyl. 69.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 68, or a pharmaceutically acceptable salt thereof is wherein R 13 is hydrogen, methyl, methyl-d3, methoxyethyl, ethoxyethyl, or propoxyethyl; and R 13a is methyl-d3, cyclopropyl, 2,2- difluoroethyl, 2,2,2-trifluoroethyl, 2,2-difluoroethoxy, 2,2,2-trifluoroethoxy, methoxyethyl, ethoxyethyl, oxetan-3-yl, tetrahydrofuranyl, or tetrahydropyranyl.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 54, or a pharmaceutically acceptable salt thereof is wherein R 13 and R 13a together with the nitrogen atom to which they are attached form heterocyclyl, bicyclic heterocyclyl, bridged heterocyclyl, fused heterocyclyl, or spiroheterocyclyl wherein (a) heterocyclyl is substituted with R m , R n , R°, and R p and (b) bicyclic heterocyclyl, bridged heterocyclyl, fused heterocyclyl, and spiroheterocyclyl are independently substituted with R q , R r , and R s .
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54 and 70, or a pharmaceutically acceptable salt thereof is wherein R 13 and R 13a together with the nitrogen atom to which they are attached form heterocyclyl substituted with R m , R n , R°, and RP.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54 and 70, or a pharmaceutically acceptable salt thereof is wherein R 13 and R 14 together with the nitrogen atom to which they are attached form bicyclic heterocyclyl, bridged heterocyclyl, fused heterocyclyl, or spiroheterocyclyl, each ring independently substituted with R r , R s , and R t .
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, 70, and 71, or a pharmaceutically acceptable salt thereof is wherein R 13 and R 13a together with the nitrogen atom to which they are attached form azetidin-l-yl, pyrrolidin- 1 -yl, piperidin- 1 -yl, piperazin-l-yl, morpholin-l-yl, or homomorpholin- 1 -yl, each ring substituted with R m , R n , R°, and R p .
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, 70, 71, and 73, or a pharmaceutically acceptable salt thereof is wherein R 13 and R 13a together with the nitrogen atom to which they are attached form azetidin-l-yl, pyrrolidin- 1 -yl, piperidin- 1 -yl, piperazin-l-yl, morpholin-l-yl, or homomorpholin- 1 -yl, each ring substituted with R m , R n , R°, and R p where R m and R n are independently selected from hydrogen, deuterium, methyl, fluoro, difluoromethyl, trifluoromethyl, difluoromethoxy, trifluoromethoxy, cyano, or methoxy, R° is hydrogen, deuterium, methoxymethyl, or fluoro, and R p is hydrogen, deuterium, or fluoro.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, 70, 71, 73, and 74, or a pharmaceutically acceptable salt thereof is wherein R m , R n , R°, and R p are hydrogen.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, 70, 71, 73, and 74, or a pharmaceutically acceptable salt thereof is wherein R 13 and R 13a together with the nitrogen atom to which they are attached form 3-methoxymethyl-azetidin-l-yl, 2- methoxymethyl-piperidin-l-yl, 3,3,4,4-tetrafluoropyrrolidin-l-yl, morpholin-l-yl.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, 70, 71, and 73 to 76, or a pharmaceutically acceptable salt thereof is wherein R 13 and R 13a together with the nitrogen atom to which they are attached form morpholin-l-yl.
  • R 13 and R 13a together with the nitrogen atom to which they are attached form morpholin-l-yl.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, and 70, or a pharmaceutically acceptable salt thereof is wherein R 13 and R 13a together with the nitrogen atom to which they are attached form bicyclic heterocyclyl substituted with R q , R r , and R s .
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, and 70, or a pharmaceutically acceptable salt thereof is wherein R 13 and R 13a together with the nitrogen atom to which they are attached form bridged heterocyclyl substituted with R q , R r , and R s .
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, and 70, or a pharmaceutically acceptable salt thereof is wherein R 13 and R 13a together with the nitrogen atom to which they are attached form fused heterocyclyl substituted with R q , R r , and R s .
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, and 70, or a pharmaceutically acceptable salt thereof is wherein R 13 and R 13a together with the nitrogen atom to which they are attached form spiroheterocyclyl substituted with R q , R r , and
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, and 70, or a pharmaceutically acceptable salt thereof is wherein R 13 and R 13a together with the nitrogen atom to which they are attached form a ring selected from: each ring substituted with R q , R r , and R s .
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 70 and 78 to 82, or a pharmaceutically acceptable salt thereof, is wherein R s is hydrogen.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 70 and 78 to 83, or a pharmaceutically acceptable salt thereof, is wherein R r and R s are hydrogen.
  • the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 70 and 78 to 84, or a pharmaceutically acceptable salt thereof is wherein R q , R r , and R s (where applicable) are independently selected from hydrogen, methyl, methoxy, or fluoro.
  • the compound of any one of embodiments 1A to 24 and 26 to 85, or a pharmaceutically acceptable salt thereof, is wherein R c , R f , and R 1 are hydrogen.
  • the compound of any one of embodiments 1A to 24 and 26 to 85, or a pharmaceutically acceptable salt thereof, is wherein R c , R f , and R 1 are deuterium.
  • the compound of any one of embodiments 1A to 24 and 26 to 85, or a pharmaceutically acceptable salt thereof is wherein R c , R f , and R 1 are independently methyl, methoxy, methyloxy, chloro, or fluoro.
  • R c , R f , and R 1 are independently chloro or fluoro, preferably, R c , R f , and R 1 are fluoro.
  • the compound of any one of embodiments 1A to 89, or a pharmaceutically acceptable salt thereof, is wherein Z is O.
  • the compound of any one of embodiments 1A to 90, or a pharmaceutically acceptable salt thereof is wherein m and n are each 1 , or one of m and n is i and the other of m and n is 2.
  • the compound of any one of embodiments 1A to 90, or a pharmaceutically acceptable salt thereof is wherein m and n are each 1 , or one of m and n is i.
  • the compound of any one of embodiments 1A to 90, or a pharmaceutically acceptable salt thereof is wherein one of m and n is 1 and the other of m and n is 2.
  • the compound of any one of embodiments 1A to 90, or a pharmaceutically acceptable salt thereof is wherein m and n are each 1.
  • the compound of any one of embodiments 1A to 90, or a pharmaceutically acceptable salt thereof is wherein m is 1 and n is 3.
  • the compound of any one of embodiments 1A to 95, or a pharmaceutically acceptable salt thereof is wherein R 1 and R 1a are independently selected from hydrogen, methyl, and ethyl, and R 1b and R lc are hydrogen.
  • the compound of any one of embodiments 1A to 95, or a pharmaceutically acceptable salt thereof is wherein R 1 is cyanomethyl and R 1a is hydrogen, methyl, or ethyl, preferably R 1b is hydrogen, and R 1b and R lc are hydrogen.
  • the compound of any one of embodiments 1A to 95, or a pharmaceutically acceptable salt thereof is wherein R 1 and R 1a are attached to the carbon atoms of the ring that are opposite or diagonal to each other and combine to form -(CH2) Z - where z is 1, 2, or 3, preferably z is 2, and R 1b and R lc are hydrogen.
  • the compound of any one of embodiments 1A to 92, 94, and 98, or a pharmaceutically acceptable salt thereof is wherein is: .
  • the compound of any one of embodiments 1A to 100, or a pharmaceutically acceptable salt thereof is wherein R 5 is -Q-R 14 where Q is bond and R 14 is cycloalkyl, fused cycloalkyl, fused spirocycloalkyl, aryl, heteroaryl, or fused heteroaryl, wherein aryl, heteroaryl, and fused heteroaryl are substituted with R t R u , R v , and R w wherein R t and R u are independently selected from hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, hydroxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, and cyano, R v is hydrogen, alkenyl, alkynyl, cyanoalkenyl, cyanoalkynyl, or halo, and R w is hydrogen, alkyl, cycloalkyl,
  • the compound of any one of embodiments 1A to 100, or a pharmaceutically acceptable salt thereof is wherein R 5 is -Q-R 14 where Q is alkylene and R 14 is cycloalkyl, aryl, or fused heteroaryl, wherein aryl, fused heteroaryl, and heteroaryl are substituted with R t R u , R v , and R w wherein R t and R u are independently selected from hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, hydroxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, and cyano, R v is hydrogen, alkynyl, or halo, and R w is hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, cyano, optionally substituted
  • the compound of any one of embodiments 1A to 100, or a pharmaceutically acceptable salt thereof is wherein R 5 is -Q-R 14 where Q is -C(O)- and R 14 is cycloalkyl, aryl, fused heteroaryl, or heteroaryl, wherein aryl, fused heteroaryl and heteroaryl are substituted with R t R u , R v , and R w wherein R t and R u are independently selected from hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, hydroxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, and cyano, R v is hydrogen, alkynyl, or halo, and R w is hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, cyano, R v is hydrogen,
  • the compound of any one of embodiments 1 A to 101, or a pharmaceutically acceptable salt thereof is wherein R 14 is cycloalkyl, fused cycloalkyl, aryl, or heteroaryl wherein aryl, and heteroaryl are substituted with R t R u , R v , and R w wherein R t and R u are independently selected from hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, hydroxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, and cyano, R v is hydrogen, alkenyl, alkynyl, cyanoalkenyl, cyanoalkynyl, or halo, and R w is hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, cyano, R v is hydrogen, al
  • the compound of any one of embodiments 1 A to 101, or a pharmaceutically acceptable salt thereof is wherein R 5 is -Q-R 14 where Q is bond and R 14 is phenyl or naphthyl substituted with R t R u , R v , and R w .
  • the compound of embodiment 105, or a pharmaceutically acceptable salt thereof is wherein R t and R u are independently selected from hydrogen, halo, and hydroxyl, R w is independently selected from hydrogen or halo, and R v is hydrogen or alkynyl.
  • the compound of embodiment 105, or a pharmaceutically acceptable salt thereof is wherein R 14 is: , substituted with R t R u , and R w where R v is alkynyl.
  • the compound of embodiment 105, or a pharmaceutically acceptable salt thereof is wherein R 14 is: , substituted with R t R u , and R w wherein R v is ethenyl.
  • the compound of any one of embodiments 1A to 101 and 105, or a pharmaceutically acceptable salt thereof is wherein R 5 is -Q-R 14 where Q is bond and R 14 is phenyl or naphthyl substituted with R t R u , R v , and R w where R t and R u are independently selected from hydrogen, alkyl, halo, haloalkyl, hydroxy, alkoxy, haloalkoxy, cycloalkyl, amino, cyano, and hydroxyalkyl, R v is hydrogen, fluoro, alkynyl, and R w is hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, cyano, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, or optionally substituted heterocyclylalky
  • the compound of any one of embodiments 1A to 105 and 106, or a pharmaceutically acceptable salt thereof is wherein R t and R u independently selected from hydrogen, methyl, ethyl, fluoro, chloro, trifluoromethyl, difluoromethyl, trifluoromethoxy, hydroxy, methyl, ethoxy, cyclopropyl, amino, cyano, and hydroxymethyl, R v is hydrogen, ethynyl, 2- cyano vinyl, 2-cyanoethyn-l-yl, or fluoro, and R w is hydrogen, methyl, fluoro, amino, or cyclopropyl.
  • the compound of any one of embodiments 1A to 101, or a pharmaceutically acceptable salt thereof is wherein R 5 is -Q-R 14 where Q is bond and R 14 is heteroaryl or fused heteroaryl substituted with R t R u , R v , and R w .
  • the compound of any one of embodiments 1A to 101 and 108, or a pharmaceutically acceptable salt thereof is wherein R 5 is -Q-R 14 where Q is bond and R 14 is a monocyclic heteroaryl (e.g., pyridyl, pyrimidinyl, benzothiazolyl) substituted with R t R u , R v , and R w .
  • R 5 is -Q-R 14 where Q is bond and R 14 is a monocyclic heteroaryl (e.g., pyridyl, pyrimidinyl, benzothiazolyl) substituted with R t R u , R v , and R w .
  • the compound of any one of embodiments 1A to 101 and 108, or a pharmaceutically acceptable salt thereof is wherein R 5 is -Q-R 14 where Q is bond and R 14 is bicyclic heteroaryl (e,g, quinolinyl, isoquinolinyl, or indazolyl), substituted with R t R u , R v , and R w .
  • R 5 is -Q-R 14 where Q is bond and R 14 is bicyclic heteroaryl (e,g, quinolinyl, isoquinolinyl, or indazolyl), substituted with R t R u , R v , and R w .
  • R t and R u are independently selected from hydrogen, alkyl, halo, haloalkyl, hydroxy, alkoxy, haloalkoxy, cycloalkyl, amino, cyano, and hydroxyalkyl and R w is hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, cyano, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, or optionally substituted heterocyclylalkyl and R v is hydrogen.
  • R t and R u are independently selected from hydrogen, methyl, ethyl, fluoro, chloro, trifluoromethyl, difluoromethyl, trifluoromethoxy, hydroxy, methyl, ethoxy, cyclopropyl, amino, cyano, and hydroxymethyl
  • R v is hydrogen or fluoro
  • R w is hydrogen, methyl, fluoro, amino, or cyclopropyl.
  • R 5 is -Q-R 14 where Q is bond and R 14 is: 113.
  • the compound of any one of embodiments 1A to 101, 103-105, and 112, or a pharmaceutically acceptable salt thereof, is wherein R 5 is -Q-R 14 where Q is bond and R 14 is:
  • R 5 is -Q-R 14 where Q is bond and R 14 is:
  • the compound of any one of embodiments 1 A to 114, or a pharmaceutically acceptable salt thereof is wherein R 2 is hydrogen, halo, or alkyl, and R 3 is hydrogen, halo, cycloalkyl, cycloalkyloxy, or alkyl.
  • the compound of any one of embodiments 1A to 115, or a pharmaceutically acceptable salt thereof, is wherein R 2 and R 3 are each hydrogen.
  • the compound of any one of embodiments 1A to 115, or a pharmaceutically acceptable salt thereof is wherein R 2 is hydrogen or chloro and R 3 is hydrogen, fluoro, cyclopropyl, or cyclopropyloxy.
  • the compound of any one of embodiments 1 A to 115, or a pharmaceutically acceptable salt thereof is wherein R 2 is hydrogen and R 3 is fluoro or cyclopropyl.
  • the compound of any one of embodiments 1 A to 115 and 118, or a pharmaceutically acceptable salt thereof is wherein R 2 is hydrogen and R 3 is fluoro or cyclopropyl and R 3 is attached to C-8, the carbon substituted with R 5 being C-7.
  • the compound of any one of embodiments 1A to 119, or a pharmaceutically acceptable salt thereof, is wherein the Degron is an E3 ligase ligand of formula (i): (i).
  • the compound of any one of embodiments 1A to 120, or a pharmaceutically acceptable salt thereof is wherein ring A of the E3 ligase ligand of formula (i) is a group of formula (a):
  • the compound of any one of embodiments 1 A to 121, or a pharmaceutically acceptable salt thereof, is wherein R 20 and R 21 are independently hydrogen or alkyl.
  • the compound of any one of embodiments 1A to 122, or a pharmaceutically acceptable salt thereof, is wherein R 20 and R 21 are hydrogen.
  • the compound of any one of embodiments 1A to 122, or a pharmaceutically acceptable salt thereof is wherein R 20 is hydrogen and R 21 is methyl.
  • the compound of any one of embodiments 1A to 120, or a pharmaceutically acceptable salt thereof is wherein the ring A of the E3 ligase ligand of formula (i) is a group of formula (b):
  • the compound of any one of embodiments 1A to 120 and 126, or a pharmaceutically acceptable salt thereof, is wherein R 22 is hydrogen.
  • the compound of any one of embodiments 1A to 120, or a pharmaceutically acceptable salt thereof, is wherein ring A of the E3 ligase ligand of formula (i) is a group of formula (c): 130.
  • the compound of any one of embodiments 1A to 128, or a pharmaceutically acceptable salt thereof is wherein ring A of the E3 ligase ligand of formula (i) is:
  • the compound of any one of embodiments 1A to 128 and 130, or a pharmaceutically acceptable salt thereof is wherein ring A of the E3 ligase ligand of formula (i) is: 132.
  • ring A of the E3 ligase ligand of formula (i) is:
  • embodiment 132a the compound of any one of embodiments 1A to 128, 130, and
  • ring A of the E3 ligase ligand of formula (i) is:
  • embodiment 133 the compound of any one of embodiments 1A to 128, 130, and
  • ring A of the E3 ligase ligand of formula (i) is:
  • embodiment 134 the compound of any one of embodiments 1A to 128, 130, and
  • ring A of the E3 ligase ligand of formula (i) is: 135.
  • ring A of the E3 ligase ligand of formula (i) is:
  • embodiment 136 the compound of any one of embodiments 1A to 128, 130, and
  • ring A of the E3 ligase ligand of formula (i) is:
  • ring A of the E3 ligase ligand of formula (i) is:
  • the compound of any one of embodiments 1A to 128, and 130 to 134, or a pharmaceutically acceptable salt thereof is wherein R aa , R bb , R cc , and R dd are independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy.
  • the compound of any one of embodiments 1 to 128, and 130 to 134, or a pharmaceutically acceptable salt thereof is wherein R aa , R bb , R cc , and R dd are independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, and cyano.
  • the compound of any one of embodiments 1A to 128, 130 to 134, and 138, or a pharmaceutically acceptable salt thereof is wherein R aa , R bb , R cc , and R dd are independently selected from hydrogen, methyl, methoxy, ethoxy, fluoro, trifluoromethyl, difluoromethyl, and trifluoromethoxy.
  • R aa , R bb , R cc , and R dd are independently selected from hydrogen and methyl.
  • the compound of any one of embodiments 1A to 128, 130 to 134, and 138, or a pharmaceutically acceptable salt thereof is wherein R aa , R bb , R cc , and R dd are independently selected from hydrogen and methoxy.
  • the compound of any one of embodiments 1A to 128, 130 to 134, and 138, or a pharmaceutically acceptable salt thereof is wherein R aa , R bb , R cc , and R dd are independently selected from hydrogen and fluoro.
  • the compound of any one of embodiments 1A to 128, 130 to 134, and 138, or a pharmaceutically acceptable salt thereof is wherein R aa , R bb , R cc , and R dd are hydrogen.
  • the compound of any one of embodiments 1A to 128, 130 to 134, and 138, or a pharmaceutically acceptable salt thereof is wherein R aa , R bb , R cc , and R dd are independently selected from hydrogen, trifluoromethyl, and difluoromethyl.
  • the compound of any one of embodiments 1A to 128, 130 to 134, and 138, or a pharmaceutically acceptable salt thereof is wherein R aa , R bb , R cc , and R dd are independently selected from hydrogen and trifluoromethoxy.
  • the compound of any one of embodiments 1A to 128, 130 to 134, and 138, or a pharmaceutically acceptable salt thereof is wherein R aa , R bb , R cc , and R dd are independently selected from hydrogen, fluoro, and trifluoromethyl.
  • the compound of any one of embodiments 1A to 146, or a pharmaceutically acceptable salt thereof, is wherein R 15 is hydrogen.
  • the compound of any one of embodiments 1A to 119, or a pharmaceutically acceptable salt thereof is wherein the Degron is an E3 ligase ligand of formula (ii):
  • the compound of any one of embodiments 1A to 119 and 147, or a pharmaceutically acceptable salt thereof is wherein Y a is CH. 149. In embodiment 149, the compound of any one of embodiments 1A to 119 and 147, or a pharmaceutically acceptable salt thereof, is wherein Y a is N.
  • embodiment 150 the compound of any one of embodiments 1A to 119, and 147-
  • Z a is a bond, -NH-, O, or -NHC(O)-.
  • Z a is a bond, -NH-, or -NHC(O)-.
  • the compound of any one of embodiments 1A to 119, and 147- 150, or a pharmaceutically acceptable salt thereof, is wherein Z a is a bond.
  • the compound of any one of embodiments 1A to 119, and 147- 150, or a pharmaceutically acceptable salt thereof is wherein Z a is -NH-, or -NHC(O)-.
  • the compound of any one of embodiments 1A to 119, 147, 148, and 150, or a pharmaceutically acceptable salt thereof, is wherein Z a is -NH-.
  • the compound of any one of embodiments 1A to 119, and 147- 150, or a pharmaceutically acceptable salt thereof, is wherein Z a is -NHC(O)-.
  • the compound of any one of embodiments 1A to 119, and 147- 154a, or a pharmaceutically acceptable salt thereof is wherein ring B is phenylene substituted with R ee and R ff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • the compound of any one of embodiments 1A to 119, and 147- 154a, or a pharmaceutically acceptable salt thereof is wherein ring B is cyclylaminylene substituted with R ee and R ff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • the compound of any one of embodiments 1A to 119, and 147- 154a, or a pharmaceutically acceptable salt thereof is wherein ring B is 5- or 6-membered monocyclic heteroarylene or a 9- or 10-membered fused bicyclic heteroarylene, wherein each heteroarylene ring contains one to three nitrogen ring atoms and each ring is substituted with R ee and R ff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • the compound of any one of embodiments 1A to 119, 147-154a and 157, or a pharmaceutically acceptable salt thereof is wherein ring B is 5- or 6-membered monocyclic heteroarylene containing one or two nitrogen ring atoms substituted with R ee and R ff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • the compound of any one of embodiments 1A to 119, 147-150 and 157, or a pharmaceutically acceptable salt thereof is wherein ring B is a 9- or 10-membered fused bicyclic heteroarylene containing one to three nitrogen ring atoms and substituted with R ee and R ff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • the compound of any one of embodiments 1A to 119, 147-150 and 157, or a pharmaceutically acceptable salt thereof is wherein ring B is a 9- or 10-membered fused bicyclic heteroarylene containing two nitrogen ring atoms and substituted with R ee and R ff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • the compound of any one of embodiments 1A to 119 and 147 to 160, or a pharmaceutically acceptable salt thereof, is wherein the E3 ligase ligand of formula (ii) is:
  • E3 ligase ligand of formula (ii) is:
  • E3 ligase ligand of formula (ii) is:
  • the compound of any one of embodiments 1A to 119, and 147 to 163, or a pharmaceutically acceptable salt thereof is wherein each R ee and R ff are independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, and haloalkoxy.
  • the compound of any one of embodiments 1A to 119, and 147 to 163, or a pharmaceutically acceptable salt thereof is wherein R ee and R ff are independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, and cyano.
  • the compound of any one of embodiments 1A to 119, and 147 to 165, or a pharmaceutically acceptable salt thereof is wherein R ee and R ff are independently selected from hydrogen, methyl, ethyl, isopropyl, cyclopropyl, methoxy, ethoxy, fluoro, chloro, trifluoromethyl, 2,2,2-trifluoroethyl, difluoromethyl, difluoromethoxy, trifluoromethoxy, or cyano.
  • the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein R ee and R ff are independently selected from hydrogen and methyl, ethyl, or isopropyl.
  • the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein R ee and R ff are independently selected from hydrogen and methoxy.
  • the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein R ee and R ff are independently selected from hydrogen, methyl, ethyl, isopropyl, chloro, and fluoro.
  • the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof is wherein one of R ee and R ff is hydrogen or fluoro and the other of R ee and R ff is selected from hydrogen, trifluoromethyl, 2,2,2-trifluoroethyl, and difluoromethyl.
  • R ee and R ff are independently selected from hydrogen, difluoromethoxy, and trifluoromethoxy.
  • the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein R ee and R ff are independently selected from hydrogen, chloro, fluoro, and trifluoromethyl.
  • the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein R ee and R ff are independently hydrogen.
  • the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein R ee and R ff are independently chloro.
  • the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein R ee and R ff are independently fluoro.
  • the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof is wherein R ee and R ff are independently trifluoromethyl or 2,2,2-trifluoroethyl.
  • the compound of any one of embodiments 1 A to 119, or a pharmaceutically acceptable salt thereof is wherein the Degron is an E3 ligase ligand of formula (iii), (iv), (v), (vi), or (vii).
  • the compound of any one of embodiments 1A to 119 and 177, or a pharmaceutically acceptable salt thereof, is wherein the Degron is an E3 ligase ligand of formula
  • the compound of any one of embodiments 1A to 119 and 177, or a pharmaceutically acceptable salt thereof, is wherein the Degron is an E3 ligase ligand of formula
  • the compound of any one of embodiments 1A to 119 and 177, or a pharmaceutically acceptable salt thereof, is wherein the Degron is an E3 ligase ligand of formula
  • the compound of any one of embodiments 1A to 119 and 177, or a pharmaceutically acceptable salt thereof is wherein the Degron is an E3 ligase ligand of formula
  • the compound of any one of embodiments 1A to 119, or a pharmaceutically acceptable salt thereof is wherein the Degron is an E3 ligase ligand of formula
  • the compound of any one of embodiments 1A to 119, or a pharmaceutically acceptable salt thereof is wherein the Degron is an E3 ligase ligand of formula (ix).
  • the compound of any one of embodiments 1A to 119, 177 to 180, and 181 c, or a pharmaceutically acceptable salt thereof is wherein the Degron is an E3 ligase ligand of formula (iii), (iv), (v), or (ix), where R 17 , R 18 , and R 19 are 1 -fluorocycloprop- 1 -yl and W a is bond, S, or methylene.
  • the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof is wherein X 1 , X 2 , X 3 , and X 4 are independently a bond.
  • the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof is wherein X 1 , X 2 , X 3 , and X 4 are independently -alkylene-, preferably methylene.
  • the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof is wherein X 1 , X 2 , X 3 , and X 4 are independently -O-.
  • the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof is wherein X 1 , X 2 , X 3 , and X 4 are independently -(O- alkylene)-.
  • the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof is wherein X 1 , X 2 , X 3 , and X 4 are independently -(alkylene-O)-.
  • the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof is wherein X 1 , X 2 , X 3 , and X 4 are independently -(NR gg - alkylene)-.
  • X 1 , X 2 , X 3 , and X 4 are independently -(alkylene - NR hh )-.
  • the compound of any one of embodiments 1 to 176, or a pharmaceutically acceptable salt thereof is wherein X 1 , X 2 , X 3 , and X 4 are
  • the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof is wherein X 1 , X 2 , X 3 , and X 4 are -NH-.
  • the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof is wherein X 1 , X 2 , X 3 , and X 4 are independently -N(alkyl)-.
  • the compound of any one of embodiments 1A and 2 to 176, or a pharmaceutically acceptable salt thereof is wherein X 1 , X 2 , X 3 , and X 4 are independently alkynylene.
  • the compound of any one of embodiments 1A and 2 to 176, or a pharmaceutically acceptable salt thereof is wherein X 1 , X 2 , X 3 , and X 4 are independently -(alkylene)- heterocyclylene- or cycloalkylene,.
  • the compound of any one of embodiments 1A to 176, 188, 189, 194, and 195, or a pharmaceutically acceptable salt thereof is wherein R gg , R hh , R jj , and R kk are independently hydrogen or alkyl.
  • the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein Z 6 is a bond or alkylene.
  • the compound of any one of embodiments 1A to 197, or a pharmaceutically acceptable salt thereof, is wherein Z 1 is -alkylene-O-.
  • the compound of any one of embodiments 1A to 197, or a pharmaceutically acceptable salt thereof is wherein Z 1 is -methylene-O-. 197a2. In embodiment 197a2, the compound of any one of embodiments 1A to 197, or a pharmaceutically acceptable salt thereof, is wherein Z 1 is -O-.
  • the compound of any one of embodiments 1A to 197a, or a pharmaceutically acceptable salt thereof, is wherein Z 2 is -C(O)-.
  • the compound of any one of embodiments 1A to 197b, or a pharmaceutically acceptable salt thereof is wherein Z 3 is -NR”-, or heterocyclene substituted with R ww and R xx .
  • the compound of any one of embodiments 1A to 197c, or a pharmaceutically acceptable salt thereof is wherein R'TM and R xx are hydrogen.
  • the compound of any one of embodiments 1A to 197d, or a pharmaceutically acceptable salt thereof is wherein R” is hydrogen.
  • the compound of any one of embodiments 1A to 197e, or a pharmaceutically acceptable salt thereof is wherein Z 4 is a bond, alkylene, -(O-alkylene)c-, or -(alkylene-O)c-.
  • the compound of any one of embodiments 1A to 197f, or a pharmaceutically acceptable salt thereof is wherein Z 5 is a bond, alkylene, -(O-alkylene)b-, or -(alkylene-O)b-.
  • the compound of any one of embodiments 1A to 197g, or a pharmaceutically acceptable salt thereof, is wherein Z 4 -Z 5 is a bond.
  • the compound of any one of embodiments 1A to 197h, or a pharmaceutically acceptable salt thereof is wherein Z 6 is a bond, alkylene, -(O-alkylene)a-, or -(alkylene-O)a-.
  • the compound of any one of embodiments 1A to 197i, or a pharmaceutically acceptable salt thereof is wherein a, b, c, and d is independently an integer selected from 1 to 3.
  • Z 1 is a bond, alkylene, or -alkylene-O-;
  • Z 2 is a bond, -alkylene, -C(O)-, -C(O)NR-, phenylene, monocyclic heteroarylene, or heterocycylene, where each ring is substituted with R'TM and R xx independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
  • Z 3 is a bond, alkylene, -NR”-, -(O-alkylene)d-, -(alkylene-O)d-, phenylene, heteroarylene, heterocyclylene, bridged heterocyclylene, or spiro heterocyclylene, where each ring is substituted with R uu and R vv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
  • Z 4 is a bond, alkylene, -C(O)NR-, -(O-alkylene)c-, or -(alkylene-O)c-;
  • Z 5 is a bond, alkylene, -C(O)-, -C(O)N(R)-, -NR’(CO)-, -(O-alkylene)b-, -(alkylene-O)b-, - 0(CH2) 7 -, -O(CH2) 8 -, or heterocyclylene, where each ring is substituted with R qq and R" independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
  • Z 6 is a bond, alkylene, -C(O)NR-, -NR’(CO)-, -S(O)2NR-, -NR’S(O)2-, -(O-alkylene)a-, - (alkylene-O)a-, phenylene, monocyclic heteroarylene, or heterocyclylene, where each ring is substituted with R 00 and R pp independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy; and where each alkylene of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 - is independently one to eight carbon atoms, R, R’ and R” is independently hydrogen or alkyl, each a, b, c, and d is independently an integer selected from 1 to 6, and each alkylene of -Z 1 -, -Z 2 -, -Z 3 -, -Z 4
  • R yy and R zz where R yy is hydrogen or deuterium and R zz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano
  • Z 1 is a bond, alkylene, or -alkylene-O-;
  • Z 2 is a bond, -alkylene, -C(O)-, or -C(O)NR-;
  • Z 3 is a bond, alkylene, -NR”-, -(alkylene-O)d-, phenylene, heteroarylene, or heterocyclylene, where each ring is substituted with R uu and R vv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
  • Z 4 is a bond, alkylene, -C(O)NR-, -(O-alkylene)c-, or -(alkylene-O)c-;
  • Z 5 is a bond, alkylene, -C(O)-, or heterocyclylene, where the ring is substituted with R qq and R" independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
  • Z 6 is a bond or alkylene; and where each alkylene of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 - is independently one to eight carbon atoms, R, R’ and R” is independently hydrogen or alkyl, each a, b, c, and d is independently an integer selected from 1 to 6, and each alkylene of -Z 1 -, -Z 2 -, -Z 3 -, -Z 4 -, -Z 5 - and
  • R yy and R zz where R yy is hydrogen or deuterium and R zz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • Z 1 is -alkylene-O-
  • Z 2 is -C(O)NR-
  • Z 3 , Z 4 and Z 5 is a bond
  • Z 6 is alkylene; and where each alkylene of -Z 1 and -Z 6 - is independently one to eight carbon atoms, R is hydrogen or alkyl, and each alkylene of -Z 1 - and -Z 6 - is substituted with R yy and R zz where R yy is hydrogen or deuterium and R zz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • Z 1 is -alkylene-O-
  • Z 2 is -C(O)NR-
  • Z 3 is -(alkylene-O)d-
  • Z 4 and Z 5 are bond and
  • Z 6 is alkylene; and where each alkylene of -Z 1 -, -Z 3 -, and -Z 6 - is independently one to eight carbon atoms, R is hydrogen or alkyl, d is an integer selected from 1 to 6, and each alkylene of -Z 1 -, -Z 3 -, and -Z 6 - is substituted with R yy and R zz where R yy is hydrogen or deuterium and R zz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • Z 1 is -alkylene-O-
  • Z 2 is -C(O)-
  • Z 3 is heterocyclylene substituted with R uu and R vv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
  • Z 6 is alkylene; and where each alkylene of -Z 1 - and -Z 2 - is independently one to eight carbon atoms, and each alkylene of -Z 1 - and -Z 2 - is substituted with R yy and R zz where R yy is hydrogen or deuterium and R zz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
  • Z 1 is -alkylene-O-
  • Z 2 , Z 3 , Z 4 , and Z 5 are a bond
  • Z 6 is alkylene; and where each alkylene of -Z 1 - and -Z 6 - is independently one to eight carbon atoms, and each alkylene of -Z 1 - and -Z 6 - is substituted with R yy and R zz where R yy is hydrogen or deuterium and R zz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • Z 1 is -alkylene-O-;
  • Z 2 is a bond;
  • Z 3 is -(alkylene-O)d-
  • Z 4 and Z 5 is a bond
  • Z 6 is alkylene; and where each alkylene of -Z 1 -, -Z 2 -, and -Z 6 - is independently one to eight carbon atoms.
  • the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
  • Z 1 , Z 2 , Z 3 , Z 4 and Z 5 are a bond
  • Z 6 is alkylene; and where alkylene of -Z 6 - is one to eight carbon atoms.
  • the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
  • Z 1 is alkylene
  • Z 2 is -C(O)NR-
  • Z 3 , Z 4 , and Z 5 are a bond
  • Z 6 is alkylene; and where each alkylene of -Z 1 - and -Z 6 - is independently one to eight carbon atoms, R is independently hydrogen or alkyl, and each alkylene of -Z 1 - and -Z 6 - is substituted with R yy and R zz where R yy is hydrogen or deuterium and R zz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • Z 1 is alkylene
  • Z 2 is a bond
  • Z 3 is phenylene, heterocyclylene or heteroarylene, each ring substituted with R uu and R" independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
  • Z 4 and Z 5 is a bond
  • Z 6 is alkylene; and where each alkylene of -Z 1 - and -Z 6 - is independently one to eight carbon atoms, and each alkylene of -Z 1 - and -Z 6 - is substituted with R yy and R zz where R yy is hydrogen or deuterium and R zz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • Z 1 is -alkylene-O-
  • Z 2 is a bond
  • Z 3 is heterocyclylene substituted with R uu and R vv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
  • Z 4 and Z 5 is a bond
  • each alkylene of -Z 1 - and -Z 6 - is independently one to eight carbon atoms, and each alkylene of -Z 1 - and -Z 6 - is substituted with R yy and R zz where R yy is hydrogen or deuterium and R zz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • Z 1 , Z 2 , Z 3 , Z 4 and Z 5 is a bond
  • Z 6 is alkylene of one to eight carbon atom and the alkylene of -Z 6 - is substituted with R yy and R zz where R yy is hydrogen or deuterium and R zz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof is wherein: Z 1 is -alkylene-O-;
  • Z 2 is -C(0)-
  • Z 3 is heterocyclylene substituted with R uu and R vv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
  • Z 4 is or -(O-alkylene)c- or -(alkylene-O)c-;
  • Z 5 is a bond
  • Z 6 is a bond or alkylene; and where each alkylene of -Z 1 -, -Z 4 -, and -Z 6 - is independently one to eight carbon atoms, each c is independently an integer selected from 1 to 6, and each alkylene of -Z 1 -, -Z 4 -, and -Z 6 - is substituted with R yy and R zz where R yy is hydrogen or deuterium and R zz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
  • Z 1 is -alkylene-O-
  • Z 2 is -C(O)-
  • Z 3 is heterocyclylene substituted with R uu and R vv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
  • Z 4 is alkylene
  • Z 5 is heterocyclylene substituted with R qq and R rr independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
  • Z 6 is a bond or alkylene; and where each alkylene of -Z 1 -, -Z 4 -, and -Z 6 - is independently one to eight carbon atoms, and each alkylene of -Z 1 -, -Z 4 -, and -Z 6 - is substituted with R yy and R zz where R yy is hydrogen or deuterium and R zz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
  • Z' is O
  • Z 2 is alkylene
  • Z 3 is phenylene (preferred p-phenylene)
  • Z 4 is monocyclic heteroarylene (preferred 1 ,4-triazolyl, 1,4-imidazolyl, 1,4-pyrazolyl, 1,4- pyrrolyl)
  • Z 5 and Z 6 are bond; and where each alkylene of -Z 3 - is independently one to eight carbon atoms, Ris independently hydrogen or alkyl, and alkylene of -Z 2 - is substituted with R yy and R zz where R yy is hydrogen or deuterium and R zz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
  • the compound of any one of embodiments 1 A to 210, or a pharmaceutically acceptable salt thereof is wherein each alkylene of -Z 1 -, -Z 2 -, -Z 3 -, -Z 4 -, -Z 5 -, and - Z 6 - is independently selected from methylene, ethylene, propylene butylene, pentylene, or hexylene, preferably methylene, ethylene, or propylene.
  • the compound of any one of embodiments 1 A to 211, or a pharmaceutically acceptable salt thereof is wherein each heterocyclylene of -Z 2 -, -Z 3 -, -Z 4 -, -Z 5 -, and -Z 6 - is independently selected from azetidindiyl, pyrrolidindiyl, piperidindiyl, or piperazindiyl.
  • the compound of any one of embodiments 1 to 212, or a pharmaceutically acceptable salt thereof is wherein each heterocyclylene of -Z 2 -, -Z 3 -, -Z 4 -, -Z 5 -, and -Z 6 - is independently selected from 1,3- azetidindiyl, 1,3-pyrrolidindiyl, 1,4-piperidinyl, or 1,4- piperazindiyl.
  • the compound of any one of embodiments 1 A to 213, or a pharmaceutically acceptable salt thereof is wherein each heteroarylene of -Z 2 -, -Z 3 -, -Z 4 -, -Z 5 -, and - Z 6 - is independently selected from 2,4, or 3,5-pyridindiyl, 2,4, or 2,5-pyrimidindiyl, or 1,3, 1,4- pyrazoldiyl. 215.
  • the compound of any one of embodiments 1 A to 214, or a pharmaceutically acceptable salt thereof is wherein each alkylene of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, as part of another group (e.g, -(O-alkylene)a, -(alkylene-O)a-, -(alkylene -NR”)-) and when present, is ethylene or propylene.
  • each alkylene of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 - as part of another group (e.g, -(O-alkylene)a, -(alkylene-O)a-, -(alkylene -NR”)-) and when present, is ethylene or propylene.
  • the compound of any one of embodiments 1A to 215, or a pharmaceutically acceptable salt thereof is wherein each alkylene of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, as part of another group (e.g, -(O-alkylene)a, -(alkylene-O)a-, -(alkylene -NR”)-) and when present, is ethylene.
  • the compound of any one of embodiments 1 to 216, or a pharmaceutically acceptable salt thereof is wherein each R, R’ and R” of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, when present, is independently hydrogen or methyl.
  • the compound of any one of embodiments 1A to 217, or a pharmaceutically acceptable salt thereof is wherein each R, R’ and R” of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, when present, is hydrogen.
  • the compound of any one of embodiments 1 A to 217, or a pharmaceutically acceptable salt thereof is wherein each R, R’ and R” of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, when present, is methyl.
  • the compound of any one of embodiments 1 A to 219, or a pharmaceutically acceptable salt thereof is wherein phenylene of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, when present, is independently selected from 1,3-phenylene and 1 ,4-phenylene unless stated otherwise in any of the embodiment above.
  • the compound of any one of embodiments 1A to 220, or a pharmaceutically acceptable salt thereof is wherein heterocyclylene, bridged heterocyclylene, and spiro heterocyclylene, of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, when present, are independently selected from
  • the compound of any one of embodiments 1 A to 221, or a pharmaceutically acceptable salt thereof is wherein L (when the Degron is a group of formula (iii) to (vi)), -X'-L-, -X 2 -L-, -X 3 -L- and -X 4 -L- are independently selected from: 223.
  • the compound of any one of embodiments 1A to 222, or a pharmaceutically acceptable salt thereof is wherein R 15 and R 16 are hydrogen.
  • the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein L is where:
  • Z 1 is -alkylene-O
  • Z 2 is -C(O)- or -C(O)NR-.
  • the compound of any one of embodiments 1A to 196 and 224, or a pharmaceutically acceptable salt thereof, is wherein Z 2 is -C(O)- .
  • the compound of any one of embodiments 1A to 196 and 224, or a pharmaceutically acceptable salt thereof is wherein Z 2 is -C(O)NR-, preferably -CONH-.
  • the compound of any one of embodiments 1A to 182 and 225, or a pharmaceutically acceptable salt thereof is wherein Z 3 is heterocyclylene, bridged heterocyclylene, or spiro heterocyclylene, where each ring is substituted with R uu and R vv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, cyanoalkyl, hydroxy, hydroxyalkyl, alkoxyalkyl, amino, alkylamino, and dialkylamino.
  • the compound of any one of embodiments 1 to 182 and 225, or a pharmaceutically acceptable salt thereof is wherein Z 3 is heterocyclylene, bridged heterocyclylene, or spiro heterocyclylene, where each ring is substituted with R uu and R vv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy amino, alkylamino, and dialkylamino.
  • the compound of any one of embodiments 1A to 182 and 226, or a pharmaceutically acceptable salt thereof is wherein Z 3 is alkylene, alkynylene, -(alkylene -NR”)-, - (alkylene-O)a-, cycloalkylene, or spiro cyclolalkylene.
  • Z 4 is a bond, alkylene, alkenylene, alkynylene, -NR’(CO)-, -O-, -(O-alkylene)c-, -(alkylene-O)c-, phenylene, or monocyclic heteroarylene, where each ring is substituted with R ss and R t independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy.
  • 230 or a pharmaceutically acceptable salt thereof, is wherein Z 4 is a bond.
  • 231a the compound of any one of embodiments 1A to 182 and 224 to 230, or a pharmaceutically acceptable salt thereof, is wherein Z 4 is alkylene.
  • the compound of any one of embodiments 1 A to 182 and 224 to 230, or a pharmaceutically acceptable salt thereof, is wherein Z 4 is alkynylene.
  • the compound of any one of embodiments 1A to 182 and 224 to 230, or a pharmaceutically acceptable salt thereof is wherein Z 4 is -NR’(CO)-, -O-, -(O-alkylene)c-, - (alkylene-O)c-, phenylene, or monocyclic heteroarylene, where each ring is substituted with R ss and R t independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy.
  • the compound of any one of embodiments 1A to 182 and 224 to 230, or a pharmaceutically acceptable salt thereof, is wherein Z 4 is -O-.
  • the compound of any one of embodiments 1A to 182 and 224 to 230, or a pharmaceutically acceptable salt thereof, is wherein Z 4 is -(O-alkylene)c.
  • the compound of any one of embodiments 1A to 182 and 224 to 230, or a pharmaceutically acceptable salt thereof, is wherein Z 4 is -(alkylene-O)c.
  • the compound of any one of embodiments 1A to 182 and 224 to 230, or a pharmaceutically acceptable salt thereof is wherein Z 4 is phenylene substituted with R ss and R t independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy.
  • the compound of any one of embodiments 1A to 182 and 224 to 230, or a pharmaceutically acceptable salt thereof is wherein Z 4 is monocyclic heteroarylene substituted with R ss and R t independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy.
  • Z 5 is a bond, alkylene, -SO2-, -NR’SO2-, -C(O)-, -NR’(CO)-, -(O-alkylene)b-, or -(alkylene-O)b-.
  • the compound of any one of embodiments 1A to 182 and 224 to 238, or a pharmaceutically acceptable salt thereof, is wherein Z 5 is a alkylene.
  • the compound of any one of embodiments 1A to 182 and 224 to 238, or a pharmaceutically acceptable salt thereof is wherein Z 5 is a -SO2-.
  • the compound of any one of embodiments 1A to 182 and 224 to 238, or a pharmaceutically acceptable salt thereof is wherein Z 5 is a -NR’SO 2 -.
  • the compound of any one of embodiments 1A to 182 and 224 to 238, or a pharmaceutically acceptable salt thereof, is wherein Z 5 is -C(O)-.
  • the compound of any one of embodiments 1A to 182 and 224 to 238, or a pharmaceutically acceptable salt thereof, is wherein Z 5 is a -NR’(CO)-.
  • the compound of any one of embodiments 1A to 182 and 224 to 238, or a pharmaceutically acceptable salt thereof is wherein Z 5 is a -(O-alkylene)b-.
  • the compound of any one of embodiments 1A to 182 and 224 to 238, or a pharmaceutically acceptable salt thereof, is wherein Z 5 is a -(alkylene-O)b.
  • Z 6 is a bond, alkylene, -C(O)NR-, - NR’(CO)-, -(alkylene-O)a-, phenylene, heterocyclylene, bicyclic heterocyclylene, spiro heterocyclylene, -O-heterocyclylene-, or -heterocyclylene-C(O)-, where each ring is substituted with R 00 and R pp independently selected from hydrogen, deuterium, alkyl, alkoxy, cyano, hydroxy, carboxy, halo, haloalkyl, and haloalkoxy.
  • the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof, is wherein Z 6 is alkylene.
  • the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof, is wherein Z 6 is a -C(O)NR-.
  • the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof, is wherein Z 6 is -NR’(CO)-.
  • the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof, is wherein Z 6 is a -(alkylene-O)a-.
  • the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof is wherein Z 6 is phenylene substituted with R 00 and R pp independently selected from hydrogen, deuterium, alkyl, alkoxy, cyano, hydroxy, carboxy, halo, haloalkyl, and haloalkoxy. 254.
  • the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof is wherein Z 6 is heterocyclylene substituted with R 00 and R pp independently selected from hydrogen, deuterium, alkyl, alkoxy, cyano, hydroxy, carboxy, halo, haloalkyl, and haloalkoxy.
  • the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof is wherein Z 6 is bicyclic heterocyclylene substituted with R 00 and R pp independently selected from hydrogen, deuterium, alkyl, alkoxy, cyano, hydroxy, carboxy, halo, haloalkyl, and haloalkoxy.
  • the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof is wherein Z 6 is spiro heterocyclylene substituted with R 00 and R pp independently selected from hydrogen, deuterium, alkyl, alkoxy, cyano, hydroxy, carboxy, halo, haloalkyl, and haloalkoxy.
  • the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof is wherein Z 6 is -O-heterocyclylene- or - heterocyclylene-C(O)-, where each ring is substituted with R 00 and R pp independently selected from hydrogen, deuterium, alkyl, alkoxy, cyano, hydroxy, carboxy, halo, haloalkyl, and haloalkoxy.
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)NR
  • Z 3 is alkylene, alkynylene, -(alkylene -NR”)-, or -(alkylene-O)a; and Z 4 , Z 5 , Z 6 , X 1 , X 2 , X 3 , and X 4 are a bond.
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)-
  • Z 3 is a heterocyclylene
  • Z 4 , Z 5 , Z 6 , X 1 , X 2 , X 3 , and X 4 are a bond.
  • Z 1 is -(alkylene-O)-;
  • Z 2 is -C(0)-;
  • Z 3 is a heterocyclylene
  • Z 4 is alkylene
  • Z 5 , Z 6 , X 1 , X 2 , X 3 , and X 4 are a bond.
  • the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)NR-
  • Z 3 is alkylene
  • Z 6 is -C(O)NR- or heterocyclylene
  • Z 4 , Z 5 , X 1 , X 2 , X 3 , and X 4 are a bond.
  • the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)NR-
  • Z 3 is alkylene
  • Z 4 , Z 5 , Z 6 , X 1 , X 2 , X 3 , and X 4 are a bond.
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)-
  • Z 3 is heterocyclylene or spiro heterocyclylene
  • Z 6 is alkylene, -C(O)NR-, -(alkylene-O)a-, or phenylene;
  • Z 4 , Z 5 , X 1 , X 2 , X 3 , and X 4 are a bond.
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)-
  • Z 3 is heterocyclylene
  • Z 4 is alkynylene, phenylene, or -(alkylene-O) c ;
  • Z 5 , Z 6 , X 1 , X 2 , X 3 , and X 4 are a bond. 263.
  • the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
  • Z 1 is -(alkylene-O)-
  • Z 2 is a -C(O)NR-
  • Z 3 is a -(alkylene-O)a- or cycloalkylene
  • Z 4 is alkylene or -O-; preferably -O-;
  • Z 6 is -C(O)NR- or - heterocyclylene
  • Z 5 , X 1 , X 2 , X 3 , and X 4 are a bond.
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O);
  • Z 3 is a heterocyclylene
  • Z 4 is alkylene, -(O-alkylene)c, -O-, or heteroarylene;
  • Z 6 is -C(O)NR-, heterocyclylene, phenylene, -O-heterocyclylene, or -heterocyclylene-CO-;
  • Z 5 , X 1 , X 2 , X 3 , and X 4 are a bond.
  • Z 1 is -(alkylene-O)-
  • Z 2 is a -C(O)NR-
  • Z 3 is alkylene, -(alkylene-O)i-3, cycloalkylene, or spiro cycloalkylene;
  • Z 5 is alkylene, NHCO, or -CO-;
  • Z 6 is -C(O)NR- or heterocyclylene
  • Z 4 , X 1 , X 2 , X 3 , and X 4 are a bond.
  • Z 1 is -(alkylene-O)-
  • Z 2 is a -C(O);
  • Z 3 is a heterocyclylene, spiro heterocyclylene, bridged heterocycylene, or heterocyclylene substituted with cyano, methoxy, methoxymethyl, hydroxymethyl, cyanomethyl, or 1 or 2 methyl;
  • Z 5 is CO or -(alkylene-O)1-3-;
  • Z 6 is heterocyclylene, bicyclic heterocycylene, spiro heterocycylene, -O- heterocyclylene, or heterocyclylene substituted with carboxy, cyano, hydroxy, methyl, methoxy, or fluoro;
  • Z 4 , X 1 , X 2 , X 3 , and X 4 are a bond.
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)-
  • Z 3 is heterocyclylene
  • Z 4 is alkylene or -O-alkylene-
  • Z 6 is heterocyclylene
  • X 1 , X 2 , X 3 , and X 4 are a bond.
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)NR-
  • Z 3 is -(alkylene-O) 1-3
  • X 1 , X 2 , X 3 , and X 4 are -alkyl ene-NH-;
  • Z 4 , Z 5 , and Z 6 are bond.
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)-
  • Z 3 is a heterocyclylene or spiro heterocyclylene
  • X 1 , X 2 , X 3 , and X 4 are -alkyl ene-NH-;
  • Z 4 , Z 5 , and Z 6 are a bond.
  • the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)NR-
  • Z 3 is heterocyclylene
  • Z 5 is -C(O)-
  • X 1 , X 2 , X 3 , and X 4 are cycloalkylene
  • Z 4 and Z 6 are a bond.
  • Z 1 is -(alkylene-O)-
  • Z 2 is a -C(O)-
  • Z 3 is a heterocyclylene
  • Z 5 is NHCO, -NHSO2-, alkylene, -(O-alkylene)-, or C(O)-;
  • X 1 , X 2 , X 3 , and X 4 are -NH- or cycloalkylene
  • Z 4 and Z 6 are a bond.
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)NR-
  • Z 3 is a heterocyclylene
  • Z 5 is CO or SO2
  • Z 6 is phenylene
  • X 1 , X 2 , X 3 , and X 4 are -alkyl ene-heterocycylene-;
  • Z 4 is a bond
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O);
  • Z 3 is heterocyclylene
  • Z 5 is CO; 7 6 is heterocyclylene;
  • X 1 , X 2 , X 3 , and X 4 are -NH- or O and Z 4 is a bond.
  • Z 1 is -(alkylene-O)-
  • Z 2 is a -C(O);
  • Z 3 is heterocyclylene
  • Z 6 is heterocyclylene
  • X 1 , X 2 , X 3 , and X 4 are alkynylene -; and Z 5 is a bond.
  • the compound of any one of embodiments 1A to 119 and 177 to 181b, or a pharmaceutically acceptable salt thereof, is wherein:
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)- or -CONH
  • Z 3 is cycloalkylene, heterocyclylene, spiro heterocycylene, or alkylene; and Z 3 , Z 4 , Z 5 , X 1 , X 2 , X 3 , and X 4 are bond.
  • the compound of any one of embodiments 1A to 119 and 177 to 181b, or a pharmaceutically acceptable salt thereof, is wherein:
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)- or -CONH
  • Z 3 is (alkylene-O)i-3, heterocyclylene, spiro heterocycylene, bridged heterocyclylene, or alkylene;
  • Z 4 is heteroarylene, alkylene, phenylene, or NHCO
  • Z 5 , Z 6 , X 1 , X 2 , X 3 , and X 4 are a bond.
  • the compound of any one of embodiments 1A to 119 and 177 to 181b, or a pharmaceutically acceptable salt thereof, is wherein:
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)- or -CONH
  • Z 3 is heterocyclylene or alkylene;
  • Z 6 is -heterocyclylene-CO-;
  • Z 5 , Z 5 , X 1 , X 2 , X 3 , and X 4 are a bond.
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)-
  • Z 3 is heterocyclylene
  • Z 5 is alkylene or CO
  • Z 6 is NHCO, alkylene, phenylene, or -heterocyclylene-CO-;
  • Z 4 , X 1 , X 2 , X 3 , and X 4 are a bond.
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)NH-
  • Z 3 is alkylene
  • Z 4 is phenylene
  • Z 5 is alkylene; -;
  • Z 6 , X 1 , X 2 , X 3 , and X 4 are a bond.
  • the compound of any one of embodiments 1A to 119 and 177 to 181b, or a pharmaceutically acceptable salt thereof, is wherein:
  • Z 1 is -(alkylene-O)-
  • Z 2 is -C(O)- or -CONH-;
  • Z 3 is heterocyclylene or alkylene
  • Z 4 is alkylene
  • Z 6 is heterocyclylene-CO or -NHCO—
  • Z 5 , X 1 , X 2 , X 3 , and X 4 are a bond.
  • the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof is wherein each alkylene of -Z 1 -, -Z 3 -, -Z 4 -, -Z 5 -, and -Z 6 -, when present, is independently selected from methylene, ethylene, propylene butylene, pentylene, or hexylene, preferably methylene, ethylene, or propylene. 282.
  • the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof is wherein each heterocyclylene of -Z 3 - and -Z 6 - is independently selected from azetidindiyl, pyrrolidindiyl, piperidindiyl, or piperazindiyl.
  • each heterocyclylene of -Z 3 - and -Z 6 - is independently selected from 1,3- azetidindiyl, 1,3 -pyrrolidindiyl, 1 ,4-piperidinyl, or 1,4- piperazindiyl.
  • the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof is wherein each alkylene of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, as part of another group (e.g, -(O-alkylene)a, -(alkylene-O)a-, -(alkylene -NR”)-) and when present, is ethylene or propylene.
  • the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof is wherein each alkylene of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, as part of another group (e.g, -(O-alkylene)a, -(alkylene-O)a-, -(alkylene -NR”)-) and when present, is ethylene.
  • the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof is wherein each R, R’ and R” of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, when present, is independently hydrogen or methyl.
  • the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof is wherein each R, R’ and R” of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, when present, is hydrogen.
  • the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof is wherein each R, R’ and R” of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, when present, is methyl.
  • the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof is wherein phenylene of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, when present, is independently selected from 1,3-phenylene and 1 ,4-phenylene unless stated otherwise in any of the embodiments above.
  • the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof is wherein heterocyclylene, bridged heterocyclylene, and spiro heterocyclylene, of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, when present, are independently selected from
  • the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof is wherein alkenylene of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, when present, are independently selected from ethenylene, propenylene butenylene, pentyenylene, or hexenylene.
  • the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof is wherein alkynylene of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, when present, are independently selected from ethynylene, propynylene butynylene, pentynylene, or hexynylene.
  • the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof is wherein cycloalkylene of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, when present, are independently selected from cycloprolylene, cyclobutylene, cyclopentylene, cyclohexylene, and cycloheptylene.
  • the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof is wherein cycloalkylene of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, when present are cyclohexylene.
  • the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof is wherein spiro cycloalkylene of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, when present, are independently selected from spiro[5.5]undecanylene, spiro[4.5]decanylene, spiro[3.5]nonanylene, spiro[4.4]nonanylene, and spiro[3.4]octanylene. 296.
  • the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof is wherein spiro cycloalkylene of -Z 1 -Z 2 -Z 3 -Z 4 -Z 5 -Z 6 -, when present, are spiro[5.5]undecanylene.
  • a pharmaceutical composition comprising a compound of any one of embodiments 1A to 299, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  • the starting materials and reagents used in preparing these compounds are either available from commercial suppliers such as Aldrich Chemical Co., (Milwaukee, Wis.), Bachem (Torrance, Calif.), or Sigma (St. Louis, Mo.) or are prepared by methods known to those skilled in the art following procedures set forth in references such as Fieser and Fieser’s Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd’s Chemistry of Carbon Compounds, Volumes 1-5 and Suppiementals (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1- 40 (John Wiley and Sons, 1991), March’s Advanced Organic Chemistry, (John Wiley and Sons, 4th Edition) and Larock’s Comprehensive Organic Transformations (VCH Publishers Inc., 1989).
  • Chlorination of a compound of formula 1-a where X a is a halogen, and other groups as defined in the Summary with a suitable chlorination reagent such as POCh optionally in presence of a base such as DIPEA provides a 2,4-dichloro compound of formula 1-b.
  • a suitable chlorination reagent such as POCh optionally in presence of a base such as DIPEA
  • a base such as DIPEA
  • Compounds of Formula 1-a where X a is halogen, U, V and W are CH, R 2 and R 3 are as defined in the Summary (or any embodiments thereof) can be by reacting a compound of formula urea at elevated temperature.
  • Compounds of formula 5 are either commercially available or can be made by methods known in the art. For example, 2-amino-4-bromo-5 -chi oro-3 - fluorobenzoic acid, 2-amino-4-bromo-3 -fluorobenzoic acid and 2-amino-4-bromobenzoic acid are commercially available.
  • Amines of formula (a”) are either commercially available or can be made by methods known in the art.
  • R 5 -M where R 5 is cycloalkyl, aryl or heteroaryl as defined in the Summary or a precursor group thereof and M is boronic acid, boronic ester, or stannane, under Suzuki, Negeshi, and Stille reaction conditions respectively, to provide a compound of formula 2-a.
  • Chlorination of a compound of formula 2-a with a suitable chlorination reagent such as POCl 3 optionally in presence of a base such as DIPEA provides compound of formula 2-b.
  • Compound 2-b is converted to a compound of Formula (IA) or (I) as described in Scheme 1 above.
  • NR" or heterocyclylene can be prepared by a precursor group of formula as illustrated and described in Scheme 3 below.
  • Treatment of a compound of formula 3-a (prepared by example 1 or 2) with a coulping agent such as CDI, and the like, treatment of the resulting intermediate with an amine of Z 3 -Z 4 -Z 5 -Z 6 - Degron precursor, provides a compound of formula 3-b which can the converted to compound of Formula (IA) or (I) as described above.
  • the present disclosure provides treatment of cancer mediated by K-ras, in particular with K- ras G12D mutants.
  • the cancer is pancreatic cancer, colorectal cancer, lung cancer, gall bladder cancer, thyroid cancer, and bile duct cancer.
  • the lung cancer is a non- small cell lung carcinoma (NSCLC), for example adenocarcinoma, squamous-cell lung carcinoma or large-cell lung carcinoma.
  • the lung cancer is a small cell lung carcinoma.
  • Other lung cancers treatable with the disclosed compounds include, but are not limited to, glandular tumors, carcinoid tumors and undifferentiated carcinomas.
  • K-ras G12D mutations are observed in hematological malignancies that affect blood, bone marrow, and/or lymph nodes.
  • the compounds of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof can be used for the treatment of acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), chronic myelogenous leukemia (CML), acute monocytic leukemia (AMoL) and/ or other leukemias, lymphomas such as all subtypes of Hodgkins lymphoma or non-Hodgkins lymphoma, plasma cell malignancies such as multiple myeloma, mantle cell lymphoma, and Waldenstrom’s macroglubunemia.
  • ALL acute lymphoblastic leukemia
  • AML acute myelogenous leukemia
  • CLL chronic lymphocytic leukemia
  • the compounds of Formula (IA) or (I), or a pharmaceutically acceptable salt thereof can be used for the treatment of a hyperproliferative disorder or metastasis in human who suffers from a cancer such as acute myeloid leukemia, cancer in adolescents, adrenocortical carcinoma childhood, AIDS related cancers (e.g.
  • Lymphoma and Kaposi's Sarcoma anal cancer, appendix cancer, astrocytomas, atypical teratoid, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumors, Burkitt lymphoma, carcinoid tumor, atypical teratoid, embryonal tumors, germ cell tumor, primary lymphoma, cervical cancer, childhood cancers, chordoma, cardiac tumors, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myleoproliferative disorders, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, extrahepatic ductal carcinoma in situ (DCIS), embryonal tumors, CNS cancer, endometrial cancer, ependymoma, esophageal cancer, esthesioneuroblastoma,
  • the compounds of Formula (IA) or (I), or a pharmaceutically acceptable salt thereof can also be used for the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
  • a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
  • K-Ras G12D activity of the compounds of Formula (IA) or (I), or a pharmaceutically acceptable salt thereof can be tested using the in vitro assay described in Biological Examples 1 below.
  • the compounds Formula (IA) or (I) (unless stated otherwise, reference to compound/compounds of Formula (IA) or (I) herein includes any embodiments thereof described herein or a pharmaceutically acceptable salt thereof) will be administered in a therapeutically effective amount by any of the accepted modes of administration for agents that serve similar utilities.
  • Therapeutically effective amounts of compounds Formula (IA) or (I) may range from about 0.01 to about 500 mg per kg patient body weight per day, which can be administered in single or multiple doses.
  • a suitable dosage level may be from about 0.1 to about 250 mg/kg per day; about 0.5 to about 100 mg/kg per day.
  • a suitable dosage level may be about 0.01 to about 250 mg/kg per day, about 0.05 to about 100 mg/kg per day, or about 0.1 to about 50 mg/kg per day. Within this range the dosage can be about 0.05 to about 0.5, about 0.5 to about 5 or about 5 to about 50 mg/kg per day.
  • the compositions can be provided in the form of tablets containing about 1.0 to about 1000 milligrams of the active ingredient, particularly about 1, 5, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, 500, 600, 750, 800, 900, and 1000 milligrams of the active ingredient.
  • the actual amount of the compound Formula (IA) or (I), i.e., the active ingredient, will depend upon numerous factors such as the severity of the disease to be treated, the age and relative health of the patient, the potency of the compound being utilized, the route and form of administration, and other factors.
  • compositions will be administered as pharmaceutical compositions by any one of the following routes: oral, systemic (e.g., transdermal, intranasal or by suppository), or parenteral (e.g., intramuscular, intravenous or subcutaneous) administration.
  • routes oral, systemic (e.g., transdermal, intranasal or by suppository), or parenteral (e.g., intramuscular, intravenous or subcutaneous) administration.
  • parenteral e.g., intramuscular, intravenous or subcutaneous
  • compositions can take the form of tablets, pills, capsules, semisolids, powders, sustained release formulations, solutions, suspensions, elixirs, aerosols, or any other appropriate compositions.
  • formulations in the form of tablets, pills or capsules, including enteric coated or delayed release tablets, pills or capsules are preferred.
  • compositions are comprised of in general, a compound of Formula (IA) or (I) in combination with at least one pharmaceutically acceptable excipient.
  • Acceptable excipients are generally non-toxic, aid administration, and do not adversely affect the therapeutic benefit of the compound of Formula (IA) or (I).
  • excipient may be any solid, liquid, semi-solid or, in the case of an aerosol composition, gaseous excipient that is generally available to one of skill in the art.
  • Solid pharmaceutical excipients include starch, cellulose, talc, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, magnesium stearate, sodium stearate, glycerol monostearate, sodium chloride, dried skim milk and the like.
  • Liquid and semisolid excipients may be selected from glycerol, propylene glycol, water, ethanol and various oils, including those of petroleum, animal, vegetable or synthetic origin, e.g., peanut oil, soybean oil, mineral oil, sesame oil, etc.
  • Preferred liquid carriers, particularly for injectable solutions include water, saline, aqueous dextrose, and glycols.
  • the compounds of Formula (IA) or (I) may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in powder form or in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, saline or sterile pyrogen-free water, immediately prior to use.
  • sterile liquid carrier for example, saline or sterile pyrogen-free water
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • Formulations for parenteral administration include aqueous and non-aqueous (oily) sterile injection solutions of the active compounds which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the compounds of Formula (IA) or (I) may also be formulated as a depot preparation.
  • Such long -acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • compositions may take the form of tablets, lozenges, pastilles, or gels formulated in conventional manner.
  • Such compositions may comprise the active ingredient in a flavored basis such as sucrose and acacia or tragacanth.
  • the compounds of Formula (IA) or (I) may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter, polyethylene glycol, or other glycerides.
  • Certain compounds of Formula (IA) or (I) may be administered topically, that is by non- systemic administration. This includes the application of a compound of Formula (IA) or (I) externally to the epidermis or the buccal cavity and the instillation of such a compound into the ear, eye and nose, such that the compound does not significantly enter the blood stream.
  • systemic administration refers to oral, intravenous, intraperitoneal and intramuscular administration.
  • Formulations suitable for topical administration include liquid or semi-liquid preparations suitable for penetration through the skin to the site of inflammation such as gels, liniments, lotions, creams, ointments or pastes, and drops suitable for administration to the eye, ear or nose.
  • the active ingredient for topical administration may comprise, for example, from 0.001% to 10% w/w (by weight) of the formulation. In certain embodiments, the active ingredient may comprise as much as 10% w/w. In other embodiments, it may comprise less than 5% w/w. In certain embodiments, the active ingredient may comprise from 2% w/w to 5% w/w. In other embodiments, it may comprise from 0.1% to 1% w/w of the formulation.
  • compounds of Formula (IA) or (I) may be conveniently delivered from an insufflator, nebulizer pressurized packs or other convenient means of delivering an aerosol spray.
  • Pressurized packs may comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the compounds of Formula (IA) or (I) may take the form of a dry powder composition, for example a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the powder composition may be presented in unit dosage form, in for example, capsules, cartridges, gelatin or blister packs from which the powder may be administered with the aid of an inhalator or insufflator.
  • suitable pharmaceutical excipients and their formulations are described in Remington’s Pharmaceutical Sciences, edited by E. W. Martin (Mack Publishing Company, 20th ed., 2000).
  • the level of the compound of Formula (IA) or (I) in a formulation can vary within the full range employed by those skilled in the art.
  • the formulation will contain, on a weight percent (wt. %) basis, from about 0.01-99.99 wt. % of a compound of Formula (IA) or (I) based on the total formulation, with the balance being one or more suitable pharmaceutical excipients.
  • the compound is present at a level of about 1-80 wt. %.
  • the compounds of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof may be used in combination with one or more other drugs in the treatment of diseases or conditions for which compounds of Formula (IA) or (I) or the other drugs may have utility.
  • Such other drug(s) may be administered, by a route and in an amount commonly used therefore, contemporaneously or sequentially with a compound of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof.
  • a pharmaceutical composition in unit dosage form containing such other drugs and the compound of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof can be used.
  • the pharmaceutical compositions of the present disclosure also include those that contain one or more other drugs, in addition to a compound of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof.
  • the combination therapy may also include therapies in which the compound of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof and one or more other drugs are administered on different overlapping schedules.
  • the compounds of Formula (IA) or (I) and the other active ingredients may be used in lower doses than when each is used singly.
  • the weight ratio of the compound of this disclosure to the second active ingredient may be varied and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used.
  • the patient can be treated with a compound of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof in any combination with one or more other anti-cancer agents including but not limited to:
  • MAP kinase pathway (RAS/RAF/MEK/ERK) inhibitors including but not limited to: Vemurafanib (PLX4032, CAS No. 918504-65-1), Dabrafenib (CAS No. 1195765-45-7), Encorafenib (LGX818 CAS No. 1269440-17-6), TQ-B3233, XL-518 (Cas No. 1029872- 29-4, available from ACC Corp); trametinib (CAS No. 871700-17-3), selumetinib (AZD6244 CAS No. 606143-52-6), TQ- B3234, PD184352 (CAS No.
  • SHP2 inhibitors including but not limited to: SHP099 (CAS No. 2200214-93-1), TNO155 (CAS No. 1801765-04-7), RMC4630, JAB-3312, JAB-3068 and ERAS-601;
  • S0S1 inhibitors including but not limited to BI 1701963 and BAY-293;
  • CSF1R inhibitors PLX3397, LY3022855,
  • CSF1R antibodies IMC-054, RG7155
  • TGF beta receptor kinase inhibitor such as LY2157299
  • BTK inhibitor such as ibrutinib; BCR-ABL inhibitors: Imatinib (CAS No. 152459-95-5); Inilotinib hydrochloride; Nilotinib (CAS No. 923288-95-3); Dasatinib (BMS-345825 CAS No. 302962-49-8); Bosutinib (SKI-606 CAS No. 380843-75-4); Ponatinib (AP24534 CAS No. 943319- 70-8); Bafetinib (INNO406 CAS No. 859212-16-1); Danusertib (PHA-739358 CAS No. 827318-97- 8), AT9283 (CAS No. 896466-04-9); Saracatinib (AZD0530 CAS No. 379231-04-6); and PF- 03814735 (CAS 942487-16-3);
  • ALK inhibitors PF-2341066 (XALKOPJ® ; crizotinib); 5-chloro-N4-(2- (isopropyl- sulfonyl)phenyl)-N2-(2-methoxy-4-(4-(4-methylpiperazin-l-yl)piperidin-l-yl)phenyl)pyrimidine-2,4- diamine; GSK1838705A (CAS No. 1116235-97-2); CH5424802 (CAS No. 1256580-46-7); Ceritinib (ZYKADIA CAS No. 1032900-25-6); TQ-B3139, and TQ-B3101;
  • PI3 K inhibitors 4- [2-( 1 H-indazol-4-yl)-6- [ [4-(methylsulfonyl)-piperazin-l- yl]methyl]thieno[3,2-d]pyrimidin-4-yl]morpholine (also known as GDC 0941 and described in PCT Publication Nos. WO 09/036082 and WO 09/055730), BEZ235 or NVP-BEZ235 (CAS No. 915019- 65-7), disclosed in PCT Publication No. WO 06/122806);
  • VEGF receptor inhibitors Bevacizumab (sold under the trademark Avastin® by Genentech/Roche), axitinib, (N-methyl-2-[[3-[(E)-2- pyridin-2- ylethenyl]- lH-indazol-6-yl]sulfanyl]benzamide, also known as AG013736, and described in PCT Publication No.
  • Brivanib Alaninate ((S)-((R)-l-(4-(4-fluoro-2-methyl-4H-indol- 5-yloxy)-5- methylpyrrolo[2,l-f][l,2,4]triazin-6-yloxy)propan-2-yl)2-aminopropanoate, also known as BMS- 582664), motesanib (N-(2,3-dihydro-3,3- dimethyl-lH-indol-6-yl)-2-[(4- pyridinylmethyl)amino]-3- pyridinecarboxamide, and described in PCT Publication No.
  • pasireotide also known as SOM230, and described in PCT Publication No. WO 02/010192
  • sorafenib sold under the tradename Nexavar®, CAS No. 284461-73-0
  • AL-2846 AL-2846
  • MET inhibitor such as foretinib (CAS No. 849217-64-7), cabozantinib (CAS No. 1140909- 48-3), capmatinib (CAS No. 1029712-80-8), tepotinib (CAS No. 1100598-32-0), savolitinib (CAS No. 1313725-88-0, or crizotinib (CAS No. 877399-52-5);
  • FLT3 inhibitors - sunitinib malate (CAS No. 341031-54-7, sold under the tradename Sutent® by Pfizer); PKC412 (CAS No. 120685-11-2, midostaurin); tandutinib (CAS No. 387867-13-2), sorafenib (CAS No. 284461-73-0), lestaurtinib (CAS No.: 111358-88-4), KW-2449 (CAS No. 1000669-72-6), quizartinib (AC220, CAS No. 950769-58-1), or crenolanib (CAS No. 670220-88-9);
  • Epidermal growth factor receptor (EGFR) inhibitors Gefitnib (sold under the tradename Iressa®), N- [4- [(3 -chloro-4-fluorophenyl)amino] -7- [ [(3 S)-tetrahydro-3 - furanyl] oxy] -6- quinazolinyl]-4(dimethylamino)-2-butenamide, sold under the tradename Tovok® by Boehringer Ingelheim), cetuximab (sold under the tradename Erbitux® by Bristol-Myers Squibb), or panitumumab (sold under the tradename Vectibix® by Amgen);
  • HER2 receptor inhibitors Trastuzumab (sold under the trademark Herceptin® by Genentech/Roche), neratinib (also known as HKI-272, (2E)-N-[4-[[3-chloro-4-[(pyridin-2- yl)methoxy]phenyl]amino]-3-cyano-7-ethoxyquinolin-6-yl]-4-(d imethylamino)but-2- enamide, and described PCT Publication No. WO 05/028443), lapatinib (CAS No.
  • lapatinib ditosylate (CAS No: 388082-77-7) (sold under the trademark Tykerb® by GlaxoSmithKline); or Trastuzumab emtansine (in the United States, ado-trastuzumab emtansine, trade name Kadcyla) - an antibody-drug conjugate consisting of the monoclonal antibody trastuzumab (Herceptin) linked to the cytotoxic agent mertansine (DM1);
  • HER dimerization inhibitors Pertuzumab (sold under the trademark Omnitarg®, by Genentech);
  • FGFR inhibitors Erdafitinib (CAS No. 1346242-81-6), Pemigatinib (CAS No. 1513857-77-6) or Infigratinib (CAS No. 872511-34-7)
  • Aurora kinase inhibitors TAS-119 (CAS No. 1453099-83-6), LY3295668 (CAS No. 1919888-06-4), or alisertib (CAS No. 1028486-01-2); CD20 antibodies: Rituximab (sold under the trademarks Riuxan® and MabThera® by Genentech/Roche), tositumomab (sold under the trademarks Bexxar® by GlaxoSmithKline), or ofatumumab (sold under the trademark Arzerra® by GlaxoSmithKline);
  • Tyrosine kinase inhibitors Erlotinib hydrochloride (CAS No. 183319-69-9, sold under the trademark Tarceva® by Genentech/Roche), Linifanib (N-[4-(3-amino-lH-indazol-4-yl)phenyl]-N'-(2- fluoro-5- methylphenyl)urea, also known as ABT 869, available from Genentech), sunitinib malate (CAS No.
  • DNA Synthesis inhibitors Capecitabine (CAS No. 154361-50-9) (sold under the trademark Xeloda® by Roche), gemcitabine hydrochloride (CAS No. 122111-03-9) (sold under the trademark Gemzar® by Eli Lilly and Company), or nelarabine ((2R3S,4R,5R)-2-(2-amino-6-methoxypurin-9- yl)-5-(hydroxymethyl)oxolane-3,4- diol, sold under the tradenames Arranon® and Atriance® by G laxo SmithKline) ;
  • Antineoplastic agents oxaliplatin (CAS No. 61825-94-3) (sold under the tradename Eloxatin® ay Sanofi- Aventis and described in US Patent No. 4,169,846);
  • G-CSF modulators Filgrastim (sold under the tradename Neupogen® by Amgen);
  • Immunomodulators Afutuzumab (available from Roche®), pegfilgrastim (sold under the tradename Neulasta® by Amgen), lenalidomide (CAS No. 191732-72-6, also known as CC-5013, sold under the tradename Revlimid®), or thalidomide (CAS No. 50-35-1, sold under the tradename Thalomid®);
  • CD40 inhibitors Dacetuzumab (also known as SGN-40 or huS2C6, available from Seattle Genetics, Inc);
  • Pro-apoptotic receptor agonists Dulanermin (also known as AMG-951, available from Amgen/Genentech); Hedgehog antagonists: 2-chloro-N-[4-chloro-3-(2-pyridinyl)phenyl]-4-(methylsulfony 1)- benzamide (also known as GDC-0449, and described in PCT Publication No. WO 06/028958);
  • Phospholipase A2 inhibitors Anagrelide (CAS No. 58579-51-4, sold under the tradename Agrylin®);
  • BCL-2 inhibitors 4-[4-[[2-(4-chlorophenyl)-5,5-dimethyl-l-cyclohexen-l-yl]met hyl]- 1- piperazinyl]-N-[[4-[[(lR)-3-(4-morpholinyl)-l-[(phenylthio)m ethyl]propyl]amino]-3- [(trifluoromethyl)sulfonyl]phenyl]sulfonyl]benzamide (also known as ABT-263 and described in PCT Publication No. WO 09/155386);
  • MCL-1 inhibitors MIK665 (CAS No. 1799631-75-6, S64315), AMG 397, and AZD5991 (CAS No. 2143010-83-5);
  • Aromatase inhibitors Exemestane (CAS No. 107868-30-4, sold under the trademark Aromasin® by Pfizer), letrozole (CAS No. 112809-51-5, sold under the tradename Femara® by Novartis), or anastrozole (CAS No. 120511-73-1, sold under the tradename Arimidex®);
  • Topoisomerase I inhibitors Irinotecan (CAS No. 97682-44-5, sold under the trademark Camptosar® by Pfizer), topotecan hydrochloride (CAS No. 119413-54-6, sold under the tradename Hycamtin® by GlaxoSmithKline);
  • Topoisomerase II inhibitors etoposide (CAS No. 33419-42-0, also known as VP-
  • mTOR inhibitors Temsirolimus (CAS No. 162635-04-3, sold under the tradename Torisel® by Pfizer), ridaforolimus (CAS No. 572924-54-0, formally known as deferolimus, AP23573 and MK8669, and described in PCT Publication No. WO 03/064383), or everolimus (CAS No. 159351- 69-6, sold under the tradename Afmitor® by Novartis);
  • Proteasome inhibitor such as carfilzomib (CAS No. 868540-17-4), MLN9708 (CAS No. 1201902-80-8), delanzomib (CAS No. 847499-27-8), or bortezomib (CAS No. 179324-69-7);
  • BET inhibitors such as INCB054329 (CAS No. 1628607-64-6), OTX015 (CAS No. 202590- 98-5), or CPI-0610 (CAS No. 1380087-89-7);
  • LSD1 inhibitors such as GSK2979552, or INCB059872;
  • HIF-2a inhibitors such as PT2977 (1672668-24-4), NKT2152, or PT2385 (CAS No. 1672665- 49-4); Osteoclastic bone resorption inhibitors: l-hydroxy-2-imidazol-l-yl-phosphonoethyl) phosphonic acid monohydrate (sold under the tradename Zometa® by Novartis);
  • CD33 Antibody Drug Conjugates Gemtuzumab ozogamicin (sold under the tradename Mylotarg® by Pfizer/Wyeth);
  • CD22 Antibody Drug Conjugates Inotuzumab ozogamicin (also referred to as CMC-544 and WAY-207294, available from Hangzhou Sage Chemical Co., Ltd.);
  • CD20 Antibody Drug Conjugates Ibritumomab tiuxetan (sold under the tradename Zevalin®);
  • octreotide also known as octreotide acetate, sold under the tradenames Sandostatin® and Sandostatin LAR®
  • Sandostatin® also known as octreotide acetate, sold under the tradenames Sandostatin® and Sandostatin LAR®
  • IL-1 1 Synthetic Interleukin- 11 (IL-1 1): oprelvekin (sold under the tradename Neumega® by Pfizer/Wyeth);
  • Radioactive Activator for Nuclear Factor k B (RANK) inhibitors Denosumab (sold under the tradename Prolia® by Amgen);
  • Thrombopoietin mimetic peptibodies Romiplostim (sold under the tradename
  • IGF-1R Anti-insulin-like Growth Factor-1 receptor antibodies: Figitumumab (also known as CP-751,871, available from ACC Corp), robatumumab (CAS No. 934235-44-6);
  • Anti-CSl antibodies Elotuzumab (HuLuc63, CAS No. 915296-00-3);
  • CD52 antibodies Alemtuzumab (sold under the tradename Campath®);
  • Histone deacetylase inhibitors Voninostat (sold under the tradename Zolinza® by Merck);
  • Alkylating agents Temozolomide (sold under the tradenames Temodar® and Temodal® by Schering-Plough/Merck), dactinomycin (also known as actinomycin-D and sold under the tradename Cosmegen®), melphalan (also known as L-PAM, L-sarcolysin, and phenylalanine mustard, sold under the tradename Alkeran®), altretamine (also known as hexamethylmelamine (HMM), sold under the tradename Hexalen®), carmustine (sold under the tradename BiCNU®), bendamustine (sold under the tradename Treanda®), busulfan (sold under the tradenames Busulfex® and Myleran®), carboplatin (sold under the tradename Paraplatin®), lomustine (also known as CCNU, sold under the tradename CeeNU®), cisplatin (also known as CDDP, sold under the tradenames Platinol® and Platinol
  • Biologic response modifiers bacillus calmette-guerin (sold under the tradenames theraCys® and TICE® BCG), or Denileukin diftitox (sold under the tradename Ontak®);
  • Anti-tumor antibiotics doxorubicin (sold under the tradenames Adriamycin® and Rubex®), bleomycin (sold under the tradename lenoxane®), daunorubicin (also known as dauorubicin hydrochloride, daunomycin, and rubidomycin hydrochloride, sold under the tradename Cerubidine®), daunorubicin liposomal (daunorubicin citrate liposome, sold under the tradename DaunoXome®), mitoxantrone (also known as DHAD, sold under the tradename Novantrone®), epirubicin (sold under the tradename EllenceTM), idarubicin (sold under the tradenames Idamycin®, Idamycin PFS®), or mitomycin C (sold under the tradename Mutamycin®);
  • Anti -microtubule agents Estramustine (CAS No. 52205-73-9, sold under the tradename Emcyl®);
  • Cathepsin K inhibitors Odanacatib (CAS No. 603139-19-1, also know as MK-0822 available from Lanzhou Chon Chemicals, ACC Corp., and Chemi eTek, and described in PCT Publication no. WO 03/075836);
  • Epothilone B analogs Ixabepilone (CAS No. 219989-84-1, sold under the tradename Lxempra® by Bristol- Myers Squibb);
  • HSP Heat Shock Protein
  • Tanespimycin (17-allylamino-17- demethoxygeldanamycin, also known as KOS-953 and 17-AAG, available from SIGMA, and described in US Patent No. 4,261,989), NVP-HSP990 (CAS No. 934343-74-5), AUY922 (CAS No. 747412-49-3), AT13387 (CAS No. 912999-49-6), STA-9090 (CAS No. 888216-25-9), Debio 0932, KW-2478 (CAS No. 819812-04-9), XL888 (CAS No. 1149705-71-4), CNF2024 (CAS No. 848695- 25-0), and TAS-116 (CAS No. 1260533-36-5); TpoR agonists: Eltrombopag (sold under the tradenames Promacta® and Revolade® by G laxo SmithKline) ;
  • Anti-mitotic agents Docetaxel (CAS No. 114977-28-5, sold under the tradename Taxotere® by Sanofi- Aventis); Adrenal steroid inhibitors: aminoglutethimide (CAS No. 125-84-8, sold under the tradename Cytadren®);
  • Nilutamide (CAS No. 63612-50-0, sold under the tradenames Nilandron® and Anandron®), bicalutamide (CAS No. 90357-06-5, sold under tradename Casodex®), or flutamide (CAS No. 13311-84-7, sold under the tradename FulexinTM);
  • CDK inhibitors including but not limited to: Alvocidib (CAS No. 146426-40-6, pan-CDK inhibitor, also known as flovopirdol or HMR-1275, 2-(2-chlorophenyl)-5,7-dihydroxy-8-[(3S,4R)-3-hydroxy-l-methyl -4-piperidinyl]-4- chromenone, and described in US Patent No. 5,621,002);
  • CDK2 inhibitor PF-07104091 CDK2 inhibitor PF-07104091
  • CDK4/6 inhibitors pabociclib (CAS No. 827022-33-3), ribociclib (CAS No. 1211441-98-3), abemaciclib (CAS No. 1231929-97-7), PF-06873600 (CAS No. 2185857-97-8), NUV-422 and Trilaciclib (CAS No. 1374743-00-6);
  • CDK7 inhibitors CT7001 (CAS No. 1805789-54-1) and SY- 1365 (CAS No. 1816989-16-8);
  • CDK9 inhibtiors AZD 4573 (CAS No. 2057509-72-3), P276-00 (CAS No. 920113-03-7), AT7519 (CAS No. 844442-38-2), CYC065 (CAS No. 1070790-89-4) or TP-1287;
  • GnRH Gonadotropin-releasing hormone receptor agonists: Leuprolide or leuprolide acetate (sold under the tradenames Viadure® by Bayer AG, Eligard® by Sanofi-Aventis and Lupron® by Abbott Lab);
  • Taxane anti-neoplastic agents Cabazitaxel (l-hydroxy-7,10 -dimethoxy-9-oxo-5,20- epoxytax-1 l-ene-2a,4,13a-triyl-4-acetate-2-benzoate-13-[(2R,3S)-3- ⁇ [(tert- butoxy)carbonyl]- amino ⁇ -2-hydroxy-3-phenylpropanoate), or larotaxel ((2a,3x,4a,5b,7a,10b,13a)- 4,10-bis(acetyloxy)- 13-( ⁇ (2R,3S)-3-[(tert-butoxycarbonyl)amino]-2-hydroxy-3- phenylpropanoyl ⁇ oxy)-l-hydroxy-9-oxo- 5,20-epoxy-7,19-cyclotax-l l-en-2-ylbenzoate);
  • 5HTla receptor agonists Xaliproden (also known as SR57746, l-[2-(2- naphthyl)ethyl]-4-[3- (trifluoromethyl)phenyl]-l,2,3,6-tetrahydropyridine, and described in US Patent No. 5,266,573);
  • HPC vaccines Cervarix® sold by GlaxoSmithKline, Gardasil® sold by Merck; Iron Chelating agents: Deferasinox (CAS No. 201530-41-8, sold under the tradename Exjade® by Novartis);
  • Anti -metabolites Claribine (2-chlorodeoxyadenosine, sold under the tradename leustatin®), 5 -fluorouracil (sold under the tradename Adrucil®), 6-thioguanine (sold under the tradename Purinethol®), pemetrexed (sold under the tradename Alimta®), cytarabine (also known as arabinosylcytosine (Ara-C), sold under the tradename Cytosar-U®), cytarabine liposomal (also known as Liposomal Ara-C, sold under the tradename DepoCytTM), decitabine (sold under the tradename Dacogen®), hydroxyurea (sold under the tradenames Hydrea®, DroxiaTM and MylocelTM), fludarabine (sold under the tradename Fludara®), floxuridine (sold under the tradename FUDR®), cladribine (also known as 2-chlorodeoxya
  • Bisphosphonates Pamidronate (CAS No. 57248-88-1, sold under the tradename Aredia®), zoledronic acid CAS No. 118072-93-8 (sold under the tradename Zometa®);
  • Demethylating agents 5-azacitidine (CAS No. 320-67-2, sold under the tradename Vidaza®), decitabine (CAS No. 2353-33-5, sold under the tradename Dacogen®);
  • Paclitaxel protein-bound (sold under the tradename Abraxane®), vinblastine (also known as vinblastine sulfate, vincaleukoblastine and VLB, sold under the tradenames Alkaban- AQ® and Velban®), vincristine (also known as vincristine sulfate, LCR, and VCR, sold under the tradenames Oncovin® and Vincasar Pfs®), vinorelbine (sold under the tradename Navelbine®), or paclitaxel (sold under the tradenames Taxol and OnxalTM);
  • Retinoids Ali tretinoin (sold under the tradename Panretin®), tretinoin (all-trans retinoic acid, also known as ATRA, sold under the tradename Vesanoid®), Isotretinoin (13- cis-retinoic acid, sold under the tradenames Accutane®, Amnesteem®, Claravis®, Clarus®, Decutan®, Isotane®, Izotech®, Oratane®, Isotret®, and Sotret®), or bexarotene (sold under the tradename Targretin®);
  • Glucocorticosteroids Hydrocortisone (also known as cortisone, hydrocortisone sodium succinate, hydrocortisone sodium phosphate, and sold under the tradenames Ala- Cort®, Hydrocortisone Phosphate, Solu-Cortef®, Hydrocort Acetate® and Lanacort®), dexamethazone ((8S, 9R, 10S, 1 IS, 13S,14S,16R, 17R)-9-fluoro-l 1,17-dihydroxy-17-(2- hydroxyacetyl)- 10, 13,16- trimethyl-6,7,8,9, 10,11,12, 13, 14, 15,16, 17-dodecahydro-3H- cyclopenta[a]phenanthren- 3-one), prednisolone (sold under the tradenames Delta-Cortel®, Orapred®, Pediapred® and Prelone®), prednisone (sold under the tradenames Deltasone®, Liquid Red®
  • Cytokines interleukin-2 (also known as aldesleukin and IL-2, sold under the tradename Proleukin®), interleukin- 11 (also known as oprevelkin, sold under the tradename Neumega®), alpha interferon alfa (also known as IFN-alpha, sold under the tradenames Intron® A, and Roferon-A®);
  • Estrogen receptor downregulators Fulvestrant (CAS No. 129453-61-8, sold under the tradename Faslodex®);
  • Anti-estrogens tamoxifen (CAS No. 10540-29-1, sold under the tradename Novaldex®); or Toremifene (CAS No. 89778-27-8, sold under the tradename Fareston®);
  • SERMs selective estrogen receptor modulators: Raloxifene (CAS No. 84449-90-1, sold under the tradename Evista®);
  • LfRH Leutinizing hormone releasing hormone
  • Goserelin CAS No. 145781-92-6, sold under the tradename Zoladex®
  • Progesterones megestrol (also known as megestrol acetate, CAS No. 595-33-5, sold under the tradename Megace®);
  • Miscellaneous cytotoxic agents Arsenic trioxide (sold under the tradename Trisenox®), or asparaginase (also known as L-asparaginase, Erwinia L-asparaginase, sold under the tradenames Elspar® and Kidrolase®);
  • immune checkpoint inhibitors include inhibitors (smack molecules or biologies) against immune checkpoint molecules such as CD27, CD28, CD40, CD 122, CD96, CD73, CD39, CD47, 0X40, GITR, CSF1R, JAK, PI3K delta, PI3K gamma, TAM kinase, arginase, CD 137 (also known as 4-1BB), ICOS, A2AR, A2BR, HIF-2a, B7-H3, B7- H4, BTLA, CTLA-4, LAG3, TIM3, VISTA, CD96, TIGIT, PD-1, PD-L1 and PD-L2.
  • inhibitors smack molecules or biologies against immune checkpoint molecules
  • immune checkpoint molecules such as CD27, CD28, CD40, CD 122, CD96, CD73, CD39, CD47, 0X40, GITR, CSF1R, JAK, PI3K delta, PI3K gamm
  • the immune checkpoint molecule is a stimulatory checkpoint molecule selected from CD27, CD28, CD40, ICOS, 0X40, GITR, CD 137 and STING.
  • the immune checkpoint molecule is an inhibitory checkpoint molecule selected from B7-H3, B7-H4, BTLA, CTLA-4, IDO, TDO, Arginase, KIR, LAG3, PD-1, TIM3, CD96, TIGIT and VISTA.
  • the compounds provided herein can be used in combination with one or more agents selected from KIR inhibitors, TIGIT inhibitors, LAIR1 inhibitors, CD 160 inhibitors, 2B4 inhibitors and TGFR beta inhibitors.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of PD-1, e.g., an anti-PD-1 monoclonal antibody.
  • the anti-PD-1 monoclonal antibody is nivolumab, pembrolizumab (also known as MK-3475), pidilizumab, SHR-1210, PDR001, or AMP - 224.
  • the anti-PD-1 monoclonal antibody is nivolumab, or pembrolizumab or PDR001.
  • the anti -PD 1 antibody is pembrolizumab.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of PD- Ll, e.g., an anti-PD-Ll monoclonal antibody.
  • the anti-PD-Ll monoclonal antibody is BMS-935559, MEDI4736, MPDL3280A (also known as RG7446), or MSB0010718C.
  • the anti-PD-Ll monoclonal antibody is MPDL3280A (atezolizumab) or MEDI4736 (durvalumab).
  • the anti-PD-Ll small molecule inhibitor is INCB86550.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of CTLA-4, e.g., an anti-CTLA-4 antibody.
  • the anti-CTLA-4 antibody is ipilimumab or tremelimumab.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of LAG3, e.g., an anti-LAG3 antibody.
  • the anti-LAG3 antibody is BMS-986016 or LAG525.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of GITR, e.g., an anti-GITR antibody.
  • the anti-GITR antibody is TRX518 or, MK-4166, INCAGN01876 or MK-1248.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of 0X40, e.g., an anti-OX40 antibody or OX40L fusion protein.
  • the anti-OX40 antibody is MED 10562 or, INCAGNO 1949, GSK2831781, GSK-3174998, MOXR-0916, PF-04518600 or LAG525.
  • the OX40L fusion protein is MEDI6383.
  • Compounds of the invention can also be used to increase or enhance an immune response, including increasing the immune response to an antigen; to improve immunization, including increasing vaccine efficacy; and to increase inflammation.
  • the compounds of the invention can be sued to enhance the immune response to vaccines including, but not limited, Listeria vaccines, oncolytic viral vaccines, and cancer vaccines such as GV AX® (granulocytemacrophage colony-stimulating factor (GM-CF) gene- transfected tumor cell vaccine).
  • Anti-cancer vaccines include dendritic cells, synthetic peptides, DNA vaccines and recombinant viruses.
  • immune-modulatory agents also include those that block immune cell migration such as antagonists to chemokine receptors, including CCR2 and CCR4; Sting agonists and Toll receptor agonists.
  • Other anti-cancer agents also include those that augment the immune system such as adjuvants or adoptive T cell transfer. Compounds of this application may be effective in combination with CAR (Chimeric antigen receptor) T cell treatment as a booster for T cell activation.
  • CAR Chimeric antigen receptor
  • a compound of the invention can also be used in combination with the following adjunct therapies: Anti-nausea drugs: NK-1 receptor antagonists: Casopitant (sold under the tradenames Rezonic® and Zunrisa® by GlaxoSmithKline); and Cytoprotective agents: Amifostine (sold under the tradename Ethyol®), leucovorin (also known as calcium leucovorin, citrovorum factor and folinic acid).
  • NK-1 receptor antagonists Casopitant (sold under the tradenames Rezonic® and Zunrisa® by GlaxoSmithKline)
  • Cytoprotective agents Amifostine (sold under the tradename Ethyol®), leucovorin (also known as calcium leucovorin, citrovorum factor and folinic acid).
  • Step 1 (2S)-tert-butyl 2-(hydroxymethyl)-5-methoxypyrrolidine- 1 -carboxylate
  • DCM 900 mL
  • DIBAL-H 1 M, 2.47 L
  • methyl alcohol 3000 mL
  • 2 M HC1 5000 mL
  • Step 4 (5 S)-l -tert-butyl 2-methyl 5-(((tert-butyldiphenylsilyl)oxy)methyl)pyrrolidine-l,2- dicarboxylate
  • Step 5 1 -(tert-butyl) 2-methyl (2S,5S)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2-(2- (chloromethyl)allyl)pyrrolidine- 1 ,2-dicarboxylate
  • Step 6 methyl (5S,7aS)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2-methylenetetrahydro-lH- pyrrolizine-7a(5H)-carboxylate
  • Step 7 ((5S,7aS)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2-methylenetetrahydro-lH-pyrrolizin-
  • Step 8 (lR,5S)-tert-butyl 3 -(2-(((5S,7aS)-5 -(((tert-butyl diphenylsilyl)oxy) methyl)-2-methylene- hexahydro-lH-pyrrolizin-7a-yl)methoxy)-7-chloro-8-fluoropyrido[4,3-d] pyrimidin-4-yl)-3,8- diazabicyclo[3.2.1]octane-8-carboxylate
  • Step 9 (lR,5S)-tert-butyl 3-(2- (((5S,7aS)-5-(((tert-butyldiphenylsilyl)oxy) methyl)-2-methylenehexahydro-lH-pyrrolizin-7a- yl)methoxy)-8-fluoro-7-(7-fluoro-3-(methoxy methoxy)-8-((triisopropylsilyl)ethynyl)naphthalen-l- yl)pyrido [4,3-d]pyrimidin-4-yl)-3,8-diazabicyclo-[3.2.1]octane-8-carboxylate
  • Step 10 (lR,5S)-tert-butyl 3-(7-(8-ethynyl-7-fluoro-3-(methoxymethoxy) naphthal en-l-yl)-8-fluoro- 2-(((5S,7aS)-5-(hydroxymethyl)-2-methylene hexahydro- lH-pyrrolizin-7a-yl)methoxy)pyrido[4, 3- d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1] octane-8-carboxylate
  • Step 10 tert-butyl (lR,5S)-3-(7-(8-ethynyl-7-fluoro-3 -(methoxymethoxy) naphthal en-l-yl)-8-fluoro-
  • Step 11 tert-butyl (lR,5S)-3-(2-(((5S,7aS)-5-(((4-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3- dioxoisoindolin-4-yl)amino)ethyl)piperazine- 1 -carbonyl)oxy)methyl)-2-methylenetetrahydro- 1 H- pyrrolizin-7a(5H)-yl)methoxy)-7-(8-ethynyl-7-fluoro-3-(methoxymethoxy)naphthalen-l-yl)-8- fluoropyrido[4,3-d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate
  • Step 12 ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl) -6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(2-((2-(2,6-dioxopiperi din-3 -yl)- 1,3 -di oxoisoindolin-4- yl)amino)ethyl)piperazine- 1 -carboxylate
  • Step 1 tert-butyl (8-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-8- oxooctyl)carbamate

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present disclosure provides certain bifunctional compounds that cause degradation of K- ras G12D via ubiquitin proteasome pathway and are therefore useful for the treatment of diseases mediated by K-ras G12D. Also provided are pharmaceutical compositions containing such compounds and processes for preparing such compounds.

Description

BIFUNCTIONAL COMPOUNDS FOR DEGRADING KRAS G12D VIA UBIQUITIN PROTE ASOME PATHWAY
Related Applications
This application claims priority to, and the benefit of, International Application No. PCT/CN2022/117697, filed on September 8, 2022, and International Application No. PCT/CN2023/072897, filed on January 18, 2023, the contents of each of which are incorporated herein by reference in their entireties.
Field of the disclosure
The present disclosure provides certain bifunctional compounds that cause degradation of K-ras G12D via ubiquitin proteasome pathway and are therefore useful for the treatment of diseases mediated by K-ras G12D. Also provided are pharmaceutical compositions containing such compounds and processes for preparing such compounds.
Background
Kirsten Rat Sarcoma 2 Viral Oncogene Homolog (KRAS) gene is a prevalent oncogene that encodes a small GTPase transductor protein called K-Ras. K-Ras can serve as a molecular switch by cycling between active GTP-bound and inactive GDP-bound forms (see Science 2001;294: 1299-304.). K-Ras signaling is activated by RAS guanine nucleotide exchange factors (GEFs), e.g., Son of Sevenless homologue (SOS) protein, that facilitate the GDP to GTP exchange of K-Ras (see Curr Biol 2005;15:563-74.). The interaction between K-Ras and GTPase-activating proteins (GAPs) such as pl20GAP and neurofibromin, potentiates K-Ras intrinsic GTPase activity and accelerates GTP hydrolysis and diminishing K-Ras signaling (see Curr. Biol. 2005;15:563- 74.).
K-Ras plays a crucial role in the regulation of cell proliferation, differentiation and survival by signaling through several major downstream pathways, including the MAPK, the PI3K and the Ral-GEFs pathways (see Lung Cancer 2018; 124: 53-64), among them the MAPK pathway is the best characterized (see Mol. Cell Biol.l995;15:6443-6453.). K-Ras-GTP binds to and activates RAF kinases, which phosphorylates MEK and subsequently phosphorylates ERK. Phospho-ERK can further activate downstream cytosolic proteins and which then translocate to the nucleus to drive the expression of diverse genes, propagating the growth signal. PI3K pathway is also involved in RAS-mediated tumorigenesis (see Cell 2007; 129:957- 968.). Upon activation by K-Ras-GTP, PI3K phosphorylates PIP2 to form PIP3, activates PDK1 and then phosphorylates AKT. pAKT yields phosphorylation of several physiological substrates, e.g., mTOR, FOXO and NF-KB that promote metabolism, cell-cycle progression, resistance to apoptosis, cell survival and migration. The Ral-GEFs signaling pathway plays a key role in RAS- mediated oncogenesis as well (see Proc. Natl. Acad. Sci. U. S. A. 1994; 91:11089-11093.). The K-Ras effector, RALGDS, stimulates the RAS family RAL-A/B small GTPases for the subsequent signaling cascades. RALGDS can also promote the JNK pathway to stimulate transcription of pro-survival and cell-cycle progression genes for cell proliferation and survival.
KRAS gene is the most frequently mutated oncogene in human cancer. KRAS mutations are associated with poor clinical outcome and found at high frequency in pancreatic cancer (-90%), colorectal cancer (~44%) and non-small-cell lung cancer (NSCLC) (~29%) (see Cancer Discov. 2021 ; 11 : 1-16). KRAS mutations are also present in breast cancer, liver cancer, biliary tract malignancies, endometrial cancer, cervical cancer, bladder cancer and myeloid leukemia. The most common KRAS mutations are observed at residues G12 (77%), G13 (10%), and Q61 (6%), and the most predominant KRAS variant in human malignancies is G12D (35%), followed by G12V (29%), G12C (21%), G12A (7%), G12R (5%), and G12S (3%) (see Cancer Discov. 2021 ; 11 :1-16). These mutations perturbate GTP hydrolysis of K-Ras by interfering with GAP binding/stimulation and/or reducing K-Ras intrinsic GTPase activity, resulting in constitutive activation of the protein and K-Ras signaling.
Targeting KRAS signaling has been a long pursuit in drug discovery. Among KRAS mutants, K-Ras G12C offers special opportunity, because it harbors a non-native cysteine residue, which can act as nucleophile and therefore can be targeted by covelent attachment. Besides AMG5 10, which is an approved drug for treating K-Ras G12C driven cancers, several other K- Ras G12C covelent inhibitors, including MRTX849, JNJ-74699157 and LY349944631, are in clinical trials for treating cancer patients with KRAS G12C mutation (see ACS Cent. Sci. 2020;6:1753-1761). These compounds occupy a dynamic pocket in the switch II region of K-Ras thereby irreversibly locking K-Ras G12C in inactive GDP -bound state. Since KRAS mutations, including G12C, enrich predominantly active-state protein in cancer cells, sufficient residual GTPase activity and nucleotide cycling are required for effective inhibition of K-Ras by inactive state-selective drugs (see Cell 2020; 183(4):850-859). Compared to K-ras G12C mutant, since K- Ras G12D mutant, does not contain non-native cysteine residue and cycle through inactive state at extremely low rate, thus making K-Ras G12D mutant specific drug discovery more challenging. As an alternative to inhibition, removal of K-Ras G12D protein would eliminate K-Ras G12D activity as well as any protein interaction or scaffolding function of K-Ras G12D. Accordingly, there is a need for bifunctional molecules that could recruit K-Ras G12D to a ubiquitin ligase and thereby causing ubiquitylation and proteasomal degradation of K-Ras G12D. The present disclosure fulfills this and related needs.
Summary
In a first aspect, provided is a compound of Formula (IA):
Figure imgf000004_0001
wherein:
U, V, and W are CH; or one or two of U, V, and W are N and the other of U, V, and W are CH; m is 1 or 2, and n is 1, 2, or 3;
R1 is hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, hydroxy, hydroxylalkyl, alkoxy alkyl, cyano, or cyanomethyl, provided R1 is not attached to the ring -NH-;
R1a is hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, hydroxy, hydroxylalkyl, or alkoxyalkyl, provided R1a is not attached to the ring -NH-; or when R1 and R1a are attached to the carbon atoms of the ring that are opposite or diagonal to each other, then R1 and R1a can combine to form -(CH2)Z- where (z is 1, 2, or 3), or -CH=CH -;
R1b is a bond to L, hydrogen, deuterium, alkyl, alkylidenyl, alkoxy, halo, haloalkyl, hydroxy, hydroxyalkyl, alkoxyalkyl, cyano, cyanomethyl, cyanoethyl, or 2-cyanovinyl, provided R1b is not attached to the ring -NH-;
R1c is hydrogen or alkyl, provided R1c is not attached to the ring -NH-;
R2 is hydrogen, deuterium, alkyl, halo, haloalkyl, alkoxy, hydroxy, or cyano, provided that R2 is absent when two of U, V, and W are N;
R3 is a bond to L, hydrogen, deuterium, alkyl, halo, haloalkyl, alkoxy, cycloalkyl, cycloalkyloxy, hydroxy, or cyano; R4 is -Z-R6 where Z is a bond, O, NH, N(alkyl), or S; and R6 is heterocyclylalkyl, fused heterocyclylalkyl, bicyclic heterocyclyl, bicyclic heterocyclylalkyl, fused bicyclic heterocyclyl, fused bicyclic heterocyclylalkyl, tricyclic heterocyclyl, or tricyclic heterocyclylalkyl, wherein:
(1) fused heterocyclyl of fused heterocyclylalkyl is substituted with Ra, Rb, and Rc where Ra is =CR7R8;
(2) heterocyclyl of heterocyclylalkyl and bicyclic heterocyclyl, by itself or as part of bicyclic heterocyclylalkyl, are substituted with Rd, Rc, and Rf where Rd is =CR9R10; and
(3) fused bicyclic heterocyclyl, by itself or as part of fused bicyclic heterocyclylalkyl, and tricyclic heterocyclyl, by itself or as part of tricyclic heterocyclylalkyl are independently substituted with Rg, Rh, and R1 where Rg is =CRnR12; where:
R7, R9, and R11 are independently a bond to L, hydrogen, deuterium, alkyl, fluoro, or haloalkyl;
R8, R10, and R12 are independently hydrogen, deuterium, alkyl, halo, haloalkyl, cyano, alkyloxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, phenyl, or heteroaryl; or independently of each other, R7 and R8, R9 and R10, and R11 and R12 together with the carbon atom to which they are attached form cycloalkylene optionally substituted with alkyl, halo, alkoxy, or hydroxy;
Rb, Rc, and Rb are independently a bond to L, hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, hydroxy, or -(alkylene)ni-OC(O)NR13R13a, wherein nl is 0 or 1, R13 is hydrogen, deuterium, alkyl, deuterioalkyl, haloalkyl, haloalkoxyalkyl, or alkoxyalkyl, and R13a is hydrogen, alkyl, deuterioalkyl, cycloalkyl, alkoxy, alkoxyalkyl, haloalkyl, haloalkoxyalkyl, or heterocyclyl substituted with Ri and Rk independently selected from alkyl, halo, and haloalkyl; or R13 and R13a together with the nitrogen atom to which they are attached form heterocyclyl, bicyclic heterocyclyl, bridged heterocyclyl, fused heterocyclyl, or spiro heterocyclyl wherein (a) heterocyclyl is substituted with Rm, Rn, R°, and Rp where Rm and Rn are independently selected from hydrogen, deuterium, alkyl, halo, haloalkyl, haloalkoxy, cyano, alkoxy and hydroxy, R° is hydrogen, deuterium, alkylidene, deuterioalkylidene, alkenyl, alkynyl, fluoro, alkoxyalkyl, alkylcarbonyl, haloalkylcarbonyl, or alkylsulfonyl, and Rp is hydrogen, deuterium, or fluoro and (b) bicyclic heterocyclyl, bridged heterocyclyl, fused heterocyclyl, and spiro heterocyclyl are independently substituted with Rq, Rr, and Rs independently selected from hydrogen, deuterium, alkyl, alkylthio, halo, haloalkyl, haloalkoxy, cyano, alkoxy, and hydroxy;
Rc, Rf, and R1 are independently a bond to L, hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, or hydroxy, provided that one of R1b, R3, R7, R9, R11, Rb, Rc, Rb, Rc, Rf, and R1 is a bond to L;
R5 is -Q-R14 where Q is bond, alkylene, or -C(=O)-; and R14 is cycloalkyl, fused cycloalkyl, fused spiro cycloalkyl, aryl, aralkyl, heteroaryl, fused heteroaryl, or heteroaralkyl wherein aryl, aryl in aralkyl, heteroaryl, fused heteroaryl, and heteroaryl in heteroaralkyl are independently substituted with Rl, Ru, Rv, and Rw wherein Rl and Ru are independently selected from hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, hydroxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, and cyano, Rv is hydrogen, alkenyl, alkynyl, cyanoalkenyl, cyanoalkynyl, or halo, and Rw is hydrogen, alkyl, alkylthio, cycloalkyl, halo, haloalkyl, haloalkoxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, cyano, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, or optionally substituted heterocyclylalkyl;
Degron is an E3 ligase ligand selected from:
(a) a group of formula (i);
Figure imgf000006_0001
(b) a group of formula (ii);
Figure imgf000006_0002
Figure imgf000007_0001
(d) a group of formula (iv):
Figure imgf000007_0002
(f) a group of formula (vi):
Figure imgf000008_0001
(vi)
(g) a group of formula (vii):
Figure imgf000008_0002
(h) a group of formula (viii):
Figure imgf000008_0003
where:
R15 and R16 are hydrogen, alkyl, cycloalkyl, or alkylcarbonyloxy;
Ya is CH or N; Za is a bond, -CH2-, -NH-, O, or -NHC(O)- where NH of -NHC(O)- is attached to Ya; ring A is a ring of formula (a), (b), or (c):
Figure imgf000009_0001
where:
X1, X2, X3’ and X4 are independently a bond, -alkylene-, alkynylene, -O-, - (O-alkylene)-, -(alkylene-O)-, -(NRgg-alkylene)-, -(alkylene-NRhh)-, -NH-, -N(alkyl)-, -C(=O)-, -(alkylene)-heterocyclylene-, cycloalkylene, -NRjjC(=O)-, or -C(=O)NRkk-, where Rgg, Rhh, Rjj, and Rkk are independently hydrogen, alkyl, or cycloalkyl and each alkylene is optionally substituted with one or two fluoro; Raa, Rbb, Rcc, and Rdd are independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano;
R20 and R21 are independently hydrogen or alkyl; or R20 and R21 together with the carbon to which they are attached form >C=O; and
R22 is hydrogen or alkyl; ring B is phenylene, cyclylaminylene, a 5- or 6-membered monocyclic heteroarylene, or a 9- or 10-membered fused bicyclic heteroarylene, wherein each heteroarylene ring contains one to three nitrogen ring atoms and further wherein the phenylene, cyclylaminylene, and heteroarylene are independently substituted with Rcc and Rff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano; and
R17, R18, and R19 are alkyl, hydroxyalkyl, cycloalkyl or heterocyclyl wherein cycloalkyl and heterocyclyl are substituted with R111 selected from hydrogen, alkyl, halo, haloalkyl, alkoxyl, haloalkoxyl, cyano, alkylcarbonyl, alkylcarbonylamino, or -COR23 where R23 is alkyl, hydroxalkyl, cycloalkyl or heterocyclyl, wherein cycloalkyl and heterocyclyl are substituted with Rnn selected from hydrogen, alkyl, halo, haloalkyl, alkoxyl, haloalkoxyl, cyano, alkylcarbonyl, and alkylcarbonylamino; and
Wa is bond, O, S, or alkylene; and
L is -Z1-Z2-Z3-Z4-Z5-Z6- where:
Z1 is a bond, alkylene, -NR”-, -O-, -(alkylene-O)-, -C(O)-, -S(O)2-, -NR’(CO)-, or -C(O)NR-;
Z2 is a bond, -alkylene, -NR”-, -O-, -C(O)-, -S(O)2-, -NR’(CO)-, -C(O)NR-, phenylene, monocyclic heteroarylene, or heterocycylene, where each ring is substituted with Rww and Rxx independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
Z3 is a bond, alkylene, alkynylene, -(alkylene -NR”)-, -(NR”-alkylene)-, -O-, -C(O)- , -NR”-, -(O-alkylene)d-, -(alkylene-O)d-, cycloalkylene, spiro cyclolalkylene, phenylene, heteroarylene, heterocyclylene, fused heterocyclylene, bridged heterocyclylene, or spiro heterocyclylene, where each ring is substituted with Ruu and Rvv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, cyanoalkyl, hydroxy, hydroxyalkyl, alkoxyalkyl, amino, alkylamino, and dialkylamino;
Z4 is a bond, alkylene, alkynylene, -C(O)NR-, -NR’(CO)-, -O-, -NR”-, -(O- alkylene)c-, -(alkylene-O)c-, cycloalkylene, spiro cyclolalkylene, phenylene, monocyclic heteroarylene, heterocyclylene, bicyclic heterocyclylene, bridged heterocyclylene, fused heterocyclylene, spiro heterocyclylene, or 11 to 13 membered spiro heterocyclylene, where each ring is substituted with Rss and Rtt independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
Z5 is a bond, alkylene, alkynylene, -SO2-, -SO2NR-, -NR’SO2-, -C(O)-, - C(O)N(R)-, -NR’(CO)-, -(O-alkylene)b-, -(alkylene-O)b-, -©(CH2)7-, -O(CH2)8-, cycloalkylene, or heterocyclylene, where each ring is substituted with Rqq and R" independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
Z6 is a bond, alkylene, -C(O)NR-, -NR’(CO)-, -S(O)2NR-, -NR’S(O)2-, -(O- alkylene)a-, -(alkylene-O)a-, phenylene, monocyclic heteroarylene, heterocyclylene, bicyclic heterocyclylene, spiro heterocyclylene, -O-heterocyclylene-, -heterocyclylene-C(O)-, where each ring is substituted with R00 and Rpp independently selected from hydrogen, deuterium, alkyl, alkoxy, cyano, hydroxy, carboxy, halo, haloalkyl, and haloalkoxy; and where each alkylene of -Z1-Z2-Z3-Z4-Z5-Z6- is independently one to eight carbon atoms, R, R’ and R” is independently hydrogen or alkyl, each a, b, c, and d is independently an integer selected from 1 to 6, and each alkylene of -Z1-, -Z2-, -Z3-, -Z4-, -Z5- and
-Z6- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano; provided that at least one of -Z1-Z2-Z3-Z4-Z5-Z6- is not a bond; wherein when several adjacent groups of Z1 to Z6 are a bond, the adjacent groups represent one bond; or a pharmaceutically acceptable salt thereof.
In a second aspect, provided is a compound of Formula (I):
Figure imgf000011_0001
wherein:
U, V, and W are CH; or one or two of U, V, and W are N and the other of U, V, and W are CH; m is 1 or 2, and n is 1, 2, or 3;
R1 is hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, hydroxy, hydroxylalkyl, alkoxy alkyl, cyano, or cyanomethyl, provided R1 is not attached to the ring -NH-;
R1a is hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, hydroxy, hydroxylalkyl, or alkoxyalkyl, provided R1a is not attached to the ring -NH-; or when R1 and R1a are attached to the carbon atoms of the ring that are opposite or diagonal to each other, then R1 and R1a can combine to form -(CH2)Z- where (z is 1, 2, or 3), or -C I I C I I -;
R1b is a bond to L, hydrogen, deuterium, alkyl, alkylidenyl, alkoxy, halo, haloalkyl, hydroxy, hydroxyalkyl, alkoxyalkyl, cyano, cyanomethyl, cyanoethyl, or 2-cyanovinyl, provided R1b is not attached to the ring -NH-;
R1c is hydrogen or alkyl, provided Rlc is not attached to the ring -NH-;
R2 is hydrogen, deuterium, alkyl, halo, haloalkyl, alkoxy, hydroxy, or cyano, provided that R2 is absent when two of U, V, and W are N;
R3 is a bond to L, hydrogen, deuterium, alkyl, halo, haloalkyl, alkoxy, cycloalkyl, cycloalkyloxy, hydroxy, or cyano;
R4 is -Z-R6 where Z is a bond, O, NH, N(alkyl), or S; and R6 is heterocyclylalkyl, fused heterocyclylalkyl, bicyclic heterocyclyl, bicyclic heterocyclylalkyl, fused bicyclic heterocyclyl, fused bicyclic heterocyclylalkyl, tricyclic heterocyclyl, or tricyclic heterocyclylalkyl, wherein:
(1) fused heterocyclyl of fused heterocyclylalkyl is substituted with Ra, Rb, and Rc where Ra is =CR7R8;
(2) heterocyclyl of heterocyclylalkyl and bicyclic heterocyclyl, by itself or as part of bicyclic heterocyclylalkyl, are substituted with Rd, Rc, and Rf where Rd is Rd is =CR9R10; and (3) fused bicyclic heterocyclyl, by itself or as part of fused bicyclic heterocyclylalkyl, and tricyclic heterocyclyl, by itself or as part of tricyclic heterocyclylalkyl are independently substituted with Rg, Rh, and R1 where Rg is =CRnR12; where:
R7, R9, and R11 are independently a bond to L, hydrogen, deuterium, alkyl, fluoro, or haloalkyl;
R8, R10, and R12 are independently hydrogen, deuterium, alkyl, halo, haloalkyl, cyano, alkyloxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, phenyl, or heteroaryl; or independently of each other, R7 and R8, R9 and R10, and R11 and R12 together with the carbon atom to which they are attached form cycloalkylene optionally substituted with alkyl, halo, alkoxy, or hydroxy;
Rb, Rc, and Rh are independently a bond to L, hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, hydroxy, or -(alkylene)ni-OC(O)NR13R13a (wherein nl is 0 or 1, R13 is hydrogen, deuterium, alkyl, deuterioalkyl, haloalkyl, haloalkoxyalkyl, or alkoxyalkyl, and R13a is hydrogen, alkyl, deuterioalkyl, cycloalkyl, alkoxy, alkoxyalkyl, haloalkyl, haloalkoxyalkyl, or heterocyclyl substituted with Ri and Rk independently selected from alkyl, halo, and haloalkyl; or R13 and R13a together with the nitrogen atom to which they are attached form heterocyclyl, bicyclic heterocyclyl, bridged heterocyclyl, fused heterocyclyl, or spiro heterocyclyl wherein (a) heterocyclyl is substituted with Rm, Rn, R°, and Rp where Rm and Rn are independently selected from hydrogen, deuterium, alkyl, halo, haloalkyl, haloalkoxy, cyano, alkoxy and hydroxy, R° is hydrogen, deuterium, alkylidene, deuterioalkylidene, alkenyl, alkynyl, fluoro, alkoxyalkyl, alkylcarbonyl, haloalkylcarbonyl, or alkylsulfonyl, and Rp is hydrogen, deuterium, or fluoro and (b) bicyclic heterocyclyl, bridged heterocyclyl, fused heterocyclyl, and spiro heterocyclyl are independently substituted with Rq, Rr, and Rs independently selected from hydrogen, deuterium, alkyl, alkylthio, halo, haloalkyl, haloalkoxy, cyano, alkoxy, and hydroxy;
Rc, Rf, and R1 are independently a bond to L, hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, or hydroxy, provided that one of R1b, R3, R7, R9, R11, Rb, Rc, Rb, Rc, Rf, and R1 is a bond to L;
R5 is -Q-R14 where Q is bond, alkylene, or -C(=O)-; and R14 is cycloalkyl, fused cycloalkyl, fused spiro cycloalkyl, aryl, aralkyl, heteroaryl, fused heteroaryl, or heteroaralkyl wherein aryl, aryl in aralkyl, heteroaryl, fused heteroaryl, and heteroaryl in heteroaralkyl are independently substituted with Rl, Ru, Rv, and Rw wherein Rl and Ru are independently selected from hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, hydroxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, and cyano, Rv is hydrogen, alkenyl, alkynyl, cyanoalkenyl, cyanoalkynyl, or halo, and Rw is hydrogen, alkyl, alkylthio, cycloalkyl, halo, haloalkyl, haloalkoxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, cyano, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, or optionally substituted heterocyclylalkyl;
Degron is an E3 ligase ligand selected from:
(a) a group of formula (i);
Figure imgf000013_0001
(b)
Figure imgf000013_0002
(c) a group of formula (iii):
Figure imgf000013_0003
(d) a group of formula (iv):
Figure imgf000014_0001
(e) a group of formula (v):
Figure imgf000014_0002
(f) a group of formula (vi):
Figure imgf000014_0003
Figure imgf000015_0001
where:
R15 and R16 are hydrogen, alkyl, cycloalkyl, or alkylcarbonyloxy;
Ya is CH or N;
Za is a bond, -CH2-, -NH-, O, or -NHC(O)- where NH of -NHC(O)- is attached to Ya; ring A is a ring of formula (a), (b), or (c):
Figure imgf000015_0002
where:
X1, X2, X3’ and X4 are independently a bond, -alkylene-, -O-, -(O-alkylene)-
-(alkylene-O)-, -(NRgg-alkylene)-, -(alkylene -NRhh)-,
Figure imgf000015_0003
-NH-, -N(alkyl)-, -C(=O)-,
-NRjjC(=O)-, or -C(=O)NRkk-, where Rgg, Rhh, Rjj, and Rkk are independently hydrogen, alkyl, or cycloalkyl and each alkylene is optionally substituted with one or two fluoro; Raa, Rbb, Rcc, and Rdd are independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano;
R20 and R21 are independently hydrogen or alkyl; or R20 and R21 together with the carbon to which they are attached form >C=O; and
R22 is hydrogen or alkyl; ring B is phenylene, cyclylaminylene, a 5- or 6-membered monocyclic heteroarylene, or a 9- or 10-membered fused bicyclic heteroarylene, wherein each heteroarylene ring contains one to three nitrogen ring atoms and further wherein the phenylene, cyclylaminylene, and heteroarylene are independently substituted with Rcc and Rff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano; and R17, R18, and R19 are alkyl, hydroxyalkyl, cycloalkyl or heterocyclyl wherein cycloalkyl and heterocyclyl are substituted with Rmm selected from hydrogen, alkyl, halo, haloalkyl, alkoxyl, haloalkoxyl, cyano, alkylcarbonyl, alkylcarbonylamino, or -COR23 where R23 is alkyl, hydroxalkyl, cycloalkyl or heterocyclyl, wherein cycloalkyl and heterocyclyl are substituted with Rnn selected from hydrogen, alkyl, halo, haloalkyl, alkoxyl, haloalkoxyl, cyano, alkylcarbonyl, and alkylcarbonylamino; and
Wa is bond, O, S, or alkylene; and
L is -Z1-Z2-Z3-Z4-Z5-Z6- where:
Z1 is a bond, alkylene, -NR”-, -O-, -(alkylene-O)-, -C(O)-, -S(O)2-, -NR’(CO)-, or -C(O)NR-;
Z2 is a bond, -alkylene, -NR”-, -O-, -C(O)-, -S(O)2-, -NR’(CO)-, -C(O)NR-, phenylene, monocyclic heteroarylene, or heterocycylene, where each ring is substituted with Rww and Rxx independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
Z3 is a bond, alkylene, alkynylene, -(alkylene -NR”)-, -(NR”- alkylene)-, -O-, - C(O)-, -NR”-, -(O-alkylene)d-, -(alkylene-O)d-, cycloalkylene, spiro cyclolalkylene, phenylene, heteroarylene, heterocyclylene, fused heterocyclylene, bridged heterocyclylene, or spiro heterocyclylene, where each ring is substituted with Ruu and Rvv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
Z4 is a bond, alkylene, alkynylene, -C(O)NR-, -NR’(CO)-, -O-, -NR”-, -(O- alkylene)c-, -(alkylene-O)c-, cycloalkylene, spiro cyclolalkylene, phenylene, monocyclic heteroarylene, heterocyclylene, bicyclic heterocyclylene, bridged heterocyclylene, fused heterocyclylene, spiro heterocyclylene, or 11 to 13 membered spiro heterocyclylene, where each ring is substituted with Rss and Rtt independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
Z5 is a bond, alkylene, alkynylene, -C(O)-, -C(O)N(R)-, -NR’(CO)-, -(O- alkylene)b-, -(alkylene-O)b-, -O(CH2)7-, -O(CH2)8-, cycloalkylene, or heterocyclylene, where each ring is substituted with Rqq and Rrr independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy; Z6 is a bond, alkylene, -C(O)NR-, -NR’(CO)-, -S(O)2NR-, -NR’S(O)2-, -(O- alkylene)a-, -(alkylene-O)a-, phenylene, monocyclic heteroarylene, or heterocyclylene, where each ring is substituted with R00 and Rpp independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy; and where each alkylene of -Z1-Z2-Z3-Z4-Z5-Z6- is independently one to eight carbon atoms, R, R’ and R” is independently hydrogen or alkyl, each a, b, c, and d is independently an integer selected from 1 to 6, and each alkylene of -Z1-, -Z2-, -Z3-, -Z4-, -Z5- and
-Z6- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano; provided that at least one of -Z1-Z2-Z3-Z4-Z5-Z6- is not a bond; wherein when several adjacent groups of Z1 to Z6 are a bond, the adjacent groups represent one bond; or a pharmaceutically acceptable salt thereof.
In a third aspect, provided is a pharmaceutical composition comprising a compound of Formula (IA) or (I) (or any of the embodiments thereof described herein) or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient.
In a fourth aspect, provided is a method of inhibiting K-Ras, in particular K-Ras G12D, in a cell, comprising contacting the cell with a compound of Formula (IA) or (I) (or any of the embodiments thereof described herein). In one embodiment of the third aspect, the contacting is in vitro. In another embodiment of the third aspect, the contacting is in vivo.
In a fifth aspect, provided is a method of inhibiting cell proliferation in vitro or in vivo, comprising contacting a cell with a compound of Formula (IA) or (I) (or any of the embodiments thereof described herein) or a pharmaceutical composition thereof as disclosed herein. In one embodiment of the fourth aspect, the contacting is in vitro. In another embodiment of the fourth aspect, the contacting is in vivo.
In a sixth aspect, provided is a method of treating cancer in a patient, preferably the patient is in need of such treatment, which method comprises administering to the patient, preferably a patient in need of such treatment, a therapeutically effective amount of a compound of Formula (IA) or (I) (or any of the embodiments thereof described herein) or a pharmaceutically acceptable salt thereof or a a pharmaceutical composition thereof as disclosed herein. In a seventh aspect, provided is a method of treating cancer associated with K-Ras, in particular K-Ras G12D, in a patient, preferably the patient is in need of such treatment, which method comprises administering to the patient, preferably a patient in need of such treatment, a therapeutically effective amount of a compound of Formula (IA) or (I) (or any of the embodiments thereof described herein) or a pharmaceutically acceptable salt thereof or a a pharmaceutical composition thereof as disclosed herein.
In an eighth aspect, provided is a compound of Formula (IA) or (I), (or any embodiments thereof described herein) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof as disclosed herein for use as a medicament. In one embodiment, the medicament is useful for the treatment of cancer.
In a ninth aspect, provided is a compound of Formula (IA) or (I), (or any embodiments thereof described herein) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof as disclosed herein for use as a therapy.
In a tenth aspect, provided is a compound of Formula (IA) or (I), (or any embodiments thereof described herein) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof as disclosed herein for use in the treatment of cancer.
In an eleventh aspect, provided is a compound of Formula (IA) or (I), (or any embodiments thereof described herein) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof as disclosed herein for use in the treatment of cancers associated with KRas, in particular cancers associated with K-Ras G12D.
In a twelfth eleventh aspect, provided is a compound of Formula (IA) or (I), (or any embodiments thereof described herein) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof as disclosed herein for use in inhibiting K-Ras, in particular K-Ras G12D.
In any of the aforementioned aspects involving the treatment of cancer, are further embodiments comprising administering the compound of Formula (IA) or (I) (or any embodiments thereof disclosed herein), or a pharmaceutically acceptable salt thereof in combination with at least one additional anticancer agent. When combination therapy is used, the agents can be administered simultaneously or sequentially.
Detailed Description
Unless otherwise stated, the following terms used in the specification and claims are defined for the purposes of this Application and have the following meaning:
“Alkyl” means a linear or branched saturated monovalent hydrocarbon radical of one to six carbon atoms, e.g., methyl, ethyl, propyl, 2-propyl, butyl, pentyl, and the like.
“Alkylene” means a linear or branched saturated divalent hydrocarbon radical of one to six carbon atoms unless otherwise stated e.g., methylene, ethylene, propylene, 1 -methylpropylene, 2- methylpropylene, butylene, pentylene, and the like.
“Alkenyl” means a linear or branched unsaturated monovalent hydrocarbon radical of two to six carbon atoms containing a double bond, e.g., ethenyl, propenyl, 2-propenyl, butenyl, pentenyl, and the like.
“Alkynyl” means a linear or branched unsaturated monovalent hydrocarbon radical of two to six carbon atoms containing a triple bond, e.g., ethynyl, propynyl, and the like.
“Alkynylene” means a linear or branched unsaturated divalent hydrocarbon radical of two to six carbon atoms containing a triple bond, e.g., and the
Figure imgf000019_0001
like.
“Alkylidenyl” means refers to a group of formula R= where R is alkyl as defined above. Examples include, but are not limited to, methylidenyl (H2C=), ethylidenyl (CH3CH =), hexylidenyl (CH3(CH3)4CH=), 2-propylidenyl (=C(CH3)2), and the like. For example, in the compound below:
Figure imgf000019_0002
the alkylidene group, methylidenyl, is enclosed by the box which is indicated by the arrow.
“Alkylsulfonyl” means a -SO2Rz radical where Rz is alkyl as defined above, e.g., methylsulfonyl, ethylsulfonyl, and the like.
“Alkylthio” means a -SRZ radical where Rz is alkyl as defined above, e.g., methylthio, ethylthio, and the like.
“Alkylcarbonyloxy” means an -OC(O)Rz group, where Rz is alkyl, as defined herein.
“Alkoxy” means a -ORZ radical where Rz is alkyl as defined above, e.g., methoxy, ethoxy, propoxy, or 2-propoxy, n-, iso-, or tert-butoxy, and the like.
“Alkoxyalkyl” means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with at least one alkoxy group, such as one or two alkoxy groups, as defined above, e.g., 2-methoxyethyl, 1-, 2-, or 3 -methoxypropyl, 2-ethoxyethyl, and the like. “Alkoxycarbonyl” means a -C(O)ORZ radical where Rz is alkyl as defined above, e.g., methoxycarbonyl, ethoxycarbonyl, and the like.
“Alkylcarbonylamino” means a -NRZ’C(O)RZ radical where Rz is alkyl and Rz’ is H or alkyl, as defined above, e.g., methylcarbonylamino, ethylcarbonylamino, and the like.
“Alkylcarbonyl” means a -C(O)RZ radical where Rz is alkyl as defined herein, e.g., methylcarbonyl, ethylcarbonyl, and the like.
“Alkylcarbonyloxy” means a -OC(O) Rz radical where Rz is alkyl as defined above, e.g., methylcarbonyloxy, ethylcarbonyloxy, and the like.
“Amino” means a -NH2.
“Alkylamino” means -NHRZ radical where Rz is alkyl is as defined above e.g., methylamino, ethylamino, propylamino, and the like.
“Aryl” means a monovalent monocyclic or bicyclic aromatic hydrocarbon radical of 6 to 10 ring atoms e.g., phenyl or naphthyl.
“Arylene” means a divalent aryl (as defined above) radical e.g., phenylene or naphthylene.
“Aralkyl” means a -(alkylene)-Rz radical where Rz is aryl as defined above. Examples include, but are not limited to, benzyl, phenethyl, and the like.
“Bicyclic heterocyclyl” means a saturated monovalent fused bicyclic ring of 8 to 12 ring atoms in which one or two ring atoms are heteroatom independently selected from N, O, and S(O)n, where n is an integer from 0 to 2, the remaining ring atoms being C, unless stated otherwise. Additionally, one or two ring carbon atoms in the heterocyclyl ring can optionally be replaced by a -CO- group. More specifically the term bicyclic heterocyclyl includes, but is not limited to, hexahydro- IH-pyrrolizinyl, and the like.
“Bicyclic heterocyclylalkyl” means a -(alkylene)-R radical where R is bicyclic heterocyclyl as defined above. Examples include, but are not limited to, hexahydro- 1H- pyrrolizinylmethyl, hexahydro- IH-pyrrolizinylethyl, and the like.
“Bicyclic heterocyclylene” means a saturated or unsaturated divalent fused bicyclic group of 9 to 12 ring atoms in which one, two, or three ring atoms are heteroatom independently selected from N, O, and S(O)n, where n is an integer selected from 0 to 2 inclusive, the remaining ring atoms being carbon, unless stated otherwise. Additionally, one or two ring carbon atoms of the bicyclic heterocyclylene ring can optionally be replaced by a -CO- group. More specifically the term bicyclic heterocyclylene includes, but is not limited to, hexahydrofuro[3,2-b]furan-3,6-diyl, and the like. When the heterocyclylene ring is unsaturated it can contain one or two ring double bonds provided that the ring is not aromatic.
“Bridged heterocyclyl” means a saturated monovalent bicyclic ring having 5 to 8 ring carbon ring atoms in which two non-adjacent ring atoms are linked by a (CRzRz’)n group where n is an integer selected from 1 to 3 inclusive and Rz and Rz’ are independently H or methyl (also may be referred to herein as “bridging” group) and further wherein one or two ring carbon atoms, including an atom in the bridging group, is replaced by a heteroatom selected from N, O, and S(O)n, where n is an integer selected from 0 to 2 inclusive. Examples include, but are not limited to, 3,8-diazabicyclo[3.2. l]octa-3-yl, and the like.
“Bridged heterocyclylene” means a saturated divalent bicyclic ring having 5 to 8 ring carbon ring atoms in which two non-adjacent ring atoms are linked by a (CRzRz’)n group where n is an integer selected from 1 to 3 inclusive and Rz and Rz’ are independently H or methyl (also may be referred to herein as “bridging” group) and further wherein one or two ring carbon atoms, including an atom in the bridging group, is replaced by a heteroatom selected from N, O, and S(O)n, where n is an integer selected from 0 to 2 inclusive. Bridged heterocyclylene is optionally substituted with one or two substituents independently selected from alkyl, halo, alkoxy, hydroxy, and cyano unless stated otherwise. Examples include, but are not limited to, 3,8- diazabicyclo[3.2.1]octa-3,8-diyl, and the like.
“Cycloalkyl” means a monocyclic saturated monovalent hydrocarbon radical of three to ten carbon atoms. Examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
“Cycloalkylalkyl” means an -(alkylene)-Rz radical where Rz is cycloalkyl as defined above. Examples include, but are not limited to, cyclopropylmethyl cyclobutylethyl, cyclopentylmethyl, cyclohexylmethyl, and the like.
“Cycloalkylene” means a divalent saturated hydrocarbon radical of three to six carbon atoms, otherwise e.g., 1,1 -cyclopropylene, 1,1 -cyclobutylene, 1 ,4-cyclohexylene, and the like.
“Cycloalkyloxy” or “cycloalkoxy” means a -OR radical where R is cycloalkyl as defined above. Examples include, but are not limited to, cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy, and the like.
“Cyanoalkyl” means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with cyano e.g., cyanomethyl, cyanoethyl, and the like. “Cyanoalkenyl” means an alkenyl radical as defined above where one of the hydrogen atom in the alkynyl chain is replace by a cyano. Examples include, but are not limited to, -C=C(CN), -CH2C=C(CN), and the like.
“Cyanoalkynyl” means an alkynyl radical as defined above where one of the hydrogen atom in the alkynyl chain is replace by a cyano. Examples include, but are not limited to, -C≡C(CN), -CH2C≡C(CN), and the like.
“Carboxy” means -COOH.
“Cyclylaminylene” means a saturated divalent monocyclic ring of 4 to 8 ring atoms in which one ring atom is nitrogen, the remaining ring atoms being C. More specifically, the term cyclylaminyl includes, but is not limited to, pyrrolidinylene, piperidinylene, homopiperidinylene, and the like.
“Deuterium” mean refers to 2H or D.
“Deuterioalkyl” mean alkyl as defined above, which is substituted with one, two, or three deuterium.
“Alkylidenyl” means refers to a group of formula R= where R is deuterioalkyl as defined above. Examples include, but are not limited to, methylidenyl (D2C=), ethylidenyl (CH3,CD=), hexylidenyl (CH3(CH2 )4CD=) , and the like.
“Deuterohaloalkyl” mean haloalkyl as defined herein, which is substituted with one, two, or three deuterium.
“Dialkylamino” means -NRz’ Rz’ radical where Rz’ and Rz" is alkyl as defined above e.g., dimethylamino, diethylamino, methylpropylamino, and the like.
“Fused bicyclic heterocyclyl” means a saturated monovalent fused bicyclic ring of 8 to 10 ring atoms in which one or two ring atoms are heteroatoms independently selected from N, O, and S(O)n, where n is an integer from 0 to 2, one ring atom can be -CO-, and the remaining ring atoms being C, unless stated otherwise, and where two adjacent ring atoms of the bicyclic ring are fused to two adjacent ring atoms of phenyl or a five or six membered heteroaryl, each as defined herein, unless stated otherwise. More specifically the term fused bicyclic heterocyclyl includes, but is not limited to, 2,3-dihydro-lH-pyrrolo[2,l-a]isoindol-9b(5H)-yl, 2,3-dihydro- lH-pyrrolo[1,2-a]indol-9a(9H)-yl, 1,3b,4,5,6,8-hexahydropyrrolo[3,2-a]pyrrolizin-3b-yl, and the like.
“Fused bicyclic heterocyclylalkyl” means a -(alkylene)-R radical where R is fused bicyclic heterocyclyl as defined above. Examples include, but are not limited to, hexahydro- 1H- pyrrolizinylmethyl, hexahydro- IH-pyrrolizinylethyl, 2,3-dihydro-lH-pyrrolo[2, l-a]isoindol- 9b(5H)-ylmethyl, 2,3-dihydro-lH-pyrrolo[l,2-a]indol-9a(9H)-ylmethyl, and the like.
“Fused cycloalkyl” as used herein, means cycloalkyl as defined above where two adjacent ring atoms of the cycloalkyl ring are fused to two adjacent ring atoms of phenyl or a five or six membered heteroaryl, each as defined herein, unless stated otherwise. The fused cycloalkyl can be attached at any atom of the ring. Non limiting examples of the fused cycloalkyl include bicyclo[4. 1.0]hepta-l,3,5-triene, bicyclo[4.2.0]octa-l,3,5-triene, and the like.
“Fused spiro cycloalkyl” means spiro cycloalkyl as defined herein where two adjacent ring atoms of the spiro cycloalkyl are fused to two adjacent ring atoms of a phenyl or a five or six membered heteroaryl, each as defined herein.
“Fused heterocyclyl” as used herein, means a saturated monovalent monocyclic ring of 4 to 7 ring atoms having from one to three heteroatoms independently selected from N, O, and S and the remaining ring atoms being carbon, and further wherein two adjacent ring atoms of the monocyclic ring is fused to two adjacent ring members of a phenyl or a five or six membered heteroaryl, each as defined herein, unless stated otherwise. The nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quatemized and one or two carbon atoms of the fused ring atoms in the saturated monocyclic ring includes the two common ring vertices shared with the fused phenyl or five or six membered heteroaryl. The fused heterocyclyl can be attached at any atom of the ring. Non limiting examples of the fused heterocycloalkyl include 2,3-dihydrobenzo[b][l,4]-dioxinyl, 2-oxabicyclo[3.1.0]hexanyl, indolin-2-one- 1 -yl, indolinyl, and the like.
“Fused heterocyclylene” as used herein, refers to a divalent bicyclic ring in which two adjacent ring atoms of a saturated monocyclic ring of 4 to 7 ring atoms having one or two heteroatoms independently selected from N, O, and S(O)n (where n is 0, 1, or 2) and the remaining ring atoms being carbon, are fused to two adjacent ring members of a phenyl or a five or six membered heteroaryl, each as defined herein, unless stated otherwise. The nitrogen atom is optionally oxidized or quatemized 1. The fused heterocyclylene can be attached at any two atoms of the ring. Representative examples include, but are not limited to, 1,2,3,4-tetrahydroquinolin- 1,4-diyl, 3,4-dihydro-2H-benzo[b][l,4]oxazin-5,8-diyl, and the like.
“Fused heterocyclylalkyl” as used herein, means a -(alkylene)-R radical where R is fused heterocyclyl, as defined herein.
“Fused heteroaryl” means fused bicyclic heteroaryl, as defined herein, where two adjacent ring atoms of the heteroaryl ring are fused to two adjacent ring atoms of phenyl. “Halo” means fluoro, chloro, bromo, or iodo, preferably fluoro or chloro.
“Haloalkyl” means alkyl radical as defined above, which is substituted with one or more halogen atoms, e.g., one to five halogen atoms, such as fluorine or chlorine, including those substituted with different halogens, e.g., -CH2CI, -CF3, -CHF2, -CH2CF3, -CF2CF3, -CF(CH3)2, and the like. When the alkyl is substituted with only fluorine atom(s), it can be referred to in this Application as fluoroalkyl.
“Haloalkoxy” means a -ORZ radical where Rz is haloalkyl as defined above e.g., -OCF3, -OCHF2, and the like. When Rz is haloalkyl where the alkyl is substituted with only fluorine atom(s), it is referred to in this Application as fluoroalkoxy.
“Haloalkoxyalkyl” means a -(alkylene)ORz radical where Rz is haloalkyl as defined above, e.g., trifluoromethoxyalkyl, and the like.
“Halocarbonyl” means a -C(O)RZ radical where Rz is haloalkyl, as defined herein, e.g., trifluoromethylcarbonyl, difluoromethylcarbonyl, and the like.
“Hydroxyalkyl” means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with one or two hydroxy groups, provided that if two hydroxy groups are present, they are not both present on the same carbon atom. Representative examples include, but are not limited to, hydroxymethyl, 2-hydroxy-ethyl, 2-hydroxypropyl, 3 -hydroxypropyl, l-(hydroxymethyl)-2-methylpropyl, 2-hydroxybutyl, 3 -hydroxybutyl, 4-hydroxybutyl, 2,3-dihydroxypropyl, 1 -(hydroxymethyl)-2- hydroxyethyl, 2,3-dihydroxybutyl, 3,4-dihydroxybutyl and 2-(hydroxymethyl)-3-hydroxypropyl, preferably 2-hydroxyethyl, 2,3-dihydroxypropyl, and l-(hydroxymethyl)-2-hydroxyethyl.
“Heteroaryl” means a monovalent monocyclic or fused bicyclic aromatic radical of 5 to 10 ring atoms, unless otherwise stated, where one or more, (in one embodiment, one, two, or three), ring atoms are heteroatom selected from N, O, and S, the remaining ring atoms being carbon. Representative examples include, but are not limited to, pyrrolyl, thienyl, thiazolyl, imidazolyl, furanyl, indolyl, isoindolyl, oxazolyl, isoxazolyl, benzothiazolyl, benzoxazolyl, quinolinyl, isoquinolinyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, tetrazolyl, and the like. As defined herein, the terms “heteroaryl” and “aryl” are mutually exclusive. When the heteroaryl ring contains 5 or 6 ring atoms and is a monocyclic ring, it is also referred to herein as 5 -or 6-membered monocyclic heteroaryl or monocyclic heteroaryl. When the heteroaryl ring contains 9- or 10 ring atoms and is a fused bicyclic ring, it is also referred to herein as 9-or 10- membered fused bicyclic heteroaryl. “Heteroaralkyl” means a -(alkylene)-R radical where R is heteroaryl as defined above, e.g., pyridinylmethyl, and the like. When the heteroaryl ring in heteroaralkyl contains 5- or 6 ring atoms it is also referred to herein as 5 -or 6-membered heteroaralkyl.
“Heteroarylene” means a divalent heteroaryl radical as defined above, unless stated otherwise. Representative examples include, but are not limited to, benzimidazoldiyl e.g., benzimidazole-l,5-diyl, and the like. When the heteroarylene ring contains 5 or 6 ring atoms and is a monocyclic ring and is also referred to herein as monocyclic heteroarylene or as 5 -or 6-membered monocyclic heteroarylene e.g., pyrazolyl-1.4-diyl. When the heteroarylene ring contains 9 or 10 ring atoms and is a fused bicyclic ring, it is also referred to herein as 9-or 10-membered fused bicyclic heteroarylene.
“Heteroaralkyl” means a -(alkylene)-Rz radical where Rz is heteroaryl as defined above, e.g., pyridinylmethyl, and the like. When the heteroaryl ring in heteroaralkyl contains 5 or 6 ring atoms it is also referred to herein as 5 -or 6-membered heteroaralkyl or monocyclic heteroaralkyl.
“Heterocyclyl” means a saturated or unsaturated monovalent monocyclic group of 4 to 8 ring atoms in which one or two ring atoms are heteroatom independently selected from N, O, and S(O)n, where n is an integer selected from 0 to 2 inclusive, the remaining ring atoms being C, unless stated otherwise. Additionally, one or two ring carbon atoms in the heterocyclyl ring can optionally be replaced by a -CO- group. More specifically the term heterocyclyl includes, but is not limited to, pyrrolidino, piperidino, homopiperidino, 2-oxopyrrolidinyl, 2-oxopiperidinyl, morpholino, piperazino, tetrahydro-pyranyl, thiomorpholino, and the like. When the heterocyclyl ring is unsaturated it can contain one or two ring double bonds provided that the ring is not aromatic. When the heterocyclyl group contains at least one nitrogen atom, it is also referred to herein as heterocycloamino and is a subset of the heterocyclyl group.
“Heterocyclylalkyl” or “heterocycloalkyl” means a -(alkylene)-Rz radical where Rz is heterocyclyl ring as defined above e.g., tetraydrofiiranylmethyl, piperazinylmethyl, morpholinylethyl, and the like.
“Heterocyclylene” means a saturated divalent monocyclic group of 4 to 6 ring atoms in which one or two ring atoms are heteroatom independently selected from N, O, and S(O)n, where n is an integer selected from 0 to 2 inclusive, the remaining ring atoms being C, unless stated otherwise. Additionally, one or two ring carbon atoms in the heterocyclylene ring can optionally be replaced by a -CO- group. More specifically, the term heterocyclylene includes, but is not limited to,
Figure imgf000025_0001
, piperidin- 1 ,4-diyl, azetidin- 1,3 -diyl, and the like. “Heteroalkyl” mean alkyl radical as defined above wherein one or two carbon atoms are replaced by O, NR (R is H or alkyl), or S, provided the heteroalkyl group is attached to the remainder of the molecule via a carbon atom, e.g., methoxymethyl, methylethylaminoethyl, and the like.
“Phenylene” refers to divalent phenyl.
The term “oxo,” as used herein, alone or in combination, refers to =(O).
“Optionally substituted aryl” means aryl as defined above, that is optionally substituted with one, two, or three substituents independently selected from alkyl, hydroxyl, cycloalkyl, carboxy, alkoxycarbonyl, hydroxy, alkoxy, alkylthio, alkylsulfonyl, amino, alkylamino, dialkylamino, halo, haloalkyl, haloalkoxy, and cyano. In some embodiments, optionally substituted aryl is optionally substituted phenyl.
“Optionally substituted aralkyl” means -(alkylene)-Rz where Rz is optionally substituted aryl as defined above.
“Optionally substituted heteroaryl” means heteroaryl as defined above that is optionally substituted with one, two, or three substituents independently selected from alkyl, alkylthio, alkylsulfonyl, hydroxyl, cycloalkyl, carboxy, alkoxycarbonyl, hydroxy, alkoxy, halo, haloalkyl, haloalkoxy, amino, alkylamino, dialkylamino, and cyano.
“Optionally substituted heteroaralkyl” means -(alkylene) -Rz where Rz is optionally substituted heteroaryl as defined above.
“Optionally substituted heterocyclyl” means heterocyclyl as defined above that is optionally substituted with one, two, or three substituents independently selected from alkyl, alkylthio, alkylsulfonyl, alkylcarbonyl, hydroxyl, cycloalkyl, cycloalkylalkyl, carboxy, alkoxycarbonyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, cyanoalkyl, halo, haloalkyl, haloalkoxy, and cyano, unless stated otherwise.
“Optionally substituted heterocyclylalkyl” means -(alkylene)-Rz where Rz is optionally substituted heterocyclyl as defined above.
The phrase “optionally” or “optional” as used herein means that the subsequently described event or circumstance may but need not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not. For example, the phrase “heteroaryl optionally substituted with alkyl” is intended to cover heteroaryl that is unsubstituted with alkyl and heteroaryl that is substituted with alkyl.
“Spiro cycloalkyl" means a saturated bicyclic monovalent ring having 5 to 10 ring atoms in in which the rings are connected through only one atom, the connecting atom is also called the spiroatom, most often a quaternary carbon ("spiro carbon"). Unless stated otherwise, spiro cycloalkyl is optionally substituted with one or two substituents independently selected from alkyl, halo, alkoxy, hydroxy, and cyano. Examples include, but are not limited to, Representative examples include, but are not limited to, spiro[3.3]heptane, spiro[3.4]octane, spiro [3.5] -nonane, and the like.
“Spiro cycloalkylene” means a saturated bicyclic divalent hydrocarbon ring having 6 to 12 ring atoms wherein the rings are connected through only one atom, the connecting atom is also called the spiroatom, most often a quaternary carbon (“spiro carbon”). Spiro cycloalkylene is optionally substituted with one or two substituents independently selected from alkyl, halo, alkoxy, hydroxy, and cyano, unless stated otherwise. Representative examples include, but are not limited to, spiro[3,5]nonandiyl e.g., spiro[3.5]nonane-2,7-diyl, and the like.
“Spiro heterocyclyl" means a saturated bicyclic monovalentvalent ring having 6 to 10 ring atoms in which one, two, or three ring atoms are heteroatom selected from N, O, and S(O)n, where n is an integer selected from 0 to 2 inclusive, the remaining ring atoms being C and the rings are connected through only one atom, the connecting atom is also called the spiroatom, most often a quaternary carbon (“spiro carbon”).
“Spiro heterocyclylene" means a saturated bicyclic divalent ring having 6 to 10 ring atoms in which one, two, or three ring atoms are heteroatom selected from N, O, and S(O)n, where n is an integer selected from 0 to 2 inclusive, the remaining ring atoms being C and the rings are connected through only one atom, the connecting atom is also called the spiroatom, most often a quaternary carbon (“spiro carbon”).
“Tricyclic heterocyclyl” means a saturated monovalent fused tricyclic ring of 9 to 14, preferably 12 to 14, ring atoms in which one, two, or three ring atoms are heteroatom independently selected from N, O, and S(O)n, where n is an integer from 0 to 2, one ring atom can be -CO-, and the remaining ring atoms being C, unless stated otherwise. Additionally, one or two ring carbon atoms in the heterocyclyl ring can optionally be replaced by a -CO- group.
The term tricyclic heterocyclyl includes, but is not limited to,
Figure imgf000027_0001
, and the like. “Tricyclic heterocyclylalkyl” means a -(alkylene)-R radical where R is tricyclic heterocyclyl as defined above. Examples include, but are not limited to,
Figure imgf000028_0001
The present disclosure also includes protected derivatives of compounds of Formula (IA) or (I). For example, when compounds of Formula (IA) or (I) contain groups such as hydroxy, carboxy, or any group containing a nitrogen atom(s), these groups can be protected with suitable protecting groups. A comprehensive list of suitable protective groups can be found in T.W. Greene, Protective Groups in Organic Synthesis, 5th Ed., John Wiley & Sons, Inc. (2014), the disclosure of which is incorporated herein by reference in its entirety. The protected derivatives of compounds of the present disclosure can be prepared by methods well known in the art.
The present disclosure also includes polymorphic forms and deuterated forms of the compound of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof.
The term “prodrug” refers to a compound that is made more active in vivo. Certain compounds Formula (IA) or (I) may also exist as prodrugs, as described in Hydrolysis in Drug and Prodrug Metabolism: Chemistry, Biochemistry, and Enzymology (Testa, Bernard and Mayer, Joachim M. Wiley- VHCA, Zurich, Switzerland 2003). Prodrugs of the compounds described herein are structurally modified forms of the compound that readily undergo chemical changes under physiological conditions to provide the active compound. Prodrugs are often useful because, in some situations, they may be easier to administer than the compound, or parent drug. They may, for instance, be bioavailable by oral administration whereas the parent drug is not. A wide variety of prodrug derivatives are known in the art, such as those that rely on hydrolytic cleavage or oxidative activation of the prodrug. An example, without limitation, of a prodrug would be a compound which is administered as an ester (the “prodrug”), but then is metabolically hydrolyzed to the carboxylic acid, the active entity. Additional examples include peptidyl derivatives of a compound.
A “pharmaceutically acceptable salt” of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound. Such salts include: acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as formic acid, acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1 ,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, glucoheptonic acid, 4,4’-methylenebis-(3-hydroxy- 2-ene- 1 -carboxylic acid), 3 -phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, /V-incthylgliicai inc, and the like. It is understood that the pharmaceutically acceptable salts are non-toxic. Additional information on suitable pharmaceutically acceptable salts can be found in Remington ’s Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, PA, 1985, which is incorporated herein by reference in its entirety.
The compounds of Formula (IA) or (I) (and any embodiment thereof disclosed herein including specific compounds) may have asymmetric centers. Compounds of Formula (IA) or (I) (and any embodiment thereof disclosed herein including specific compounds) containing an asymmetrically substituted atom may be isolated in optically active or racemic forms. Individual stereoisomers of compounds can be prepared synthetically from commercially available starting materials which contain chiral centers or by preparation of mixtures of enantiomeric products followed by separation such as conversion to a mixture of diastereomers followed by separation or recrystallization, chromatographic techniques, direct separation of enantiomers on chiral chromatographic columns, or any other appropriate method known in the art. All chiral, diastereomeric, all mixtures of chiral or diastereomeric forms, and racemic forms are within the scope of this disclosure, unless the specific stereochemistry or isomeric form is specifically indicated. It will also be understood by a person of ordinary skill in the art that when a compound is denoted as (R) stereoisomer, it may contain the corresponding (S) stereoisomer as an impurity and vice versa.
Certain compounds of Formula (IA) or (I) (and any embodiment thereof disclosed herein including specific compounds) can exist as tautomers and/or geometric isomers. All possible tautomers and cis and trans isomers, as individual forms and mixtures thereof are within the scope of this disclosure. Additionally, as used herein the term alkyl includes all the possible isomeric forms of said alkyl group albeit only a few examples are set forth. Furthermore, when the cyclic groups such as aryl is substituted, it includes all the positional isomers albeit only a few examples are set forth. Furthermore, all hydrates of a compound of Formula (IA) or (I) (and any embodiment thereof disclosed herein including specific compounds) are within the scope of this disclosure.
The compounds of Formula (IA) or (I) (and any embodiment thereof disclosed herein including specific compounds) may also contain unnatural amounts of isotopes at one or more of the atoms that constitute such compounds. Unnatural amounts of an isotope may be defined as ranging from the amount found in nature to an amount 100% of the atom in question that differ only in the presence of one or more isotopically enriched atoms. Exemplary isotopes that can be incorporated into compounds of the present disclosure, such as a compound of Formula (IA) or (I) (and any embodiment thereof disclosed herein including specific compounds) include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, and iodine, such as 2H, 3H, 11C, 13C, 14C, 13N, 15N, 15O, 17O, 18O, 32P, 33P, 35S, 18F, 36C1, 123I, and 1251, respectively. Isotopically labeled compounds (e.g., those labeled with 3H and 14C) can be useful in compound or substrate tissue distribution assays. Tritiated (i.e., 3H) and carbon-14 (i.e., 14C) isotopes can be useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements). In some embodiments, in compounds of Formula (IA) or (I) (and any embodiment thereof disclosed herein including specific compounds) one or more hydrogen atoms are replaced by 2H or 3H, or one or more carbon atoms are replaced by 13C- or 14C- enriched carbon. Positron emitting isotopes such as 15O, 13N, 11C, and 15F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy. Isotopically labeled compounds can generally be prepared by following procedures analogous to those disclosed in the Schemes or in the Examples herein, by substituting an isotopically labeled reagent for a non- isotopically labeled reagent.
A “pharmaceutically acceptable carrier or excipient” means a carrier or an excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes a carrier or an excipient that is acceptable for veterinary use as well as human pharmaceutical use. “A pharmaceutically acceptable carrier/excipient” as used in the specification and claims includes both one and more than one such excipient.
The term “about,” as used herein, is intended to qualify the numerical values which it modifies, denoting such a value as variable within a margin of error. When no particular margin of error, such as a standard deviation to a mean value given in a chart or table of data, is recited, the term “about” should be understood to mean that range which would encompass ± 10%, preferably ± 5%, wherein the recited value and the range (including the endpoints) are included.
The phrase alkylene optionally substituted with one or two fluoro in the definition of X1, X2, X3’ and X4 in Formula (IA) or (I) (and any embodiment thereof disclosed herein) (and similar phrases used to define other groups in Formula (IA) or (I)) is intended to cover alkylene that is unsubstituted and alkylene that is substituted one or two fluoro.
Certain structures provided herein are drawn with one or more floating substituents. Unless provided otherwise or otherwise clear from the context, the substituent(s) may be present on any atom of the ring to which it is attached, where chemically feasible and valency rules permitting. For example, in the structure:
For example, in the structure of Formula (la):
Figure imgf000031_0001
R2 and R3 groups are floating substituents and can replace the hydrogen atom of any one of U, V, and W of the
Figure imgf000031_0002
portion of the quinazoline ring ring when U, V, and W are CH; and in the structure the Raa substituent of Raa, Rbb and X1, and similarly
Figure imgf000031_0003
the Rbb and X1 substituents, can replace hydrogen of any CH that is part of the benzo portion of the bicyclic ring that is not already substituted with Rbb and X1, and similarly Raa and X1, and Raa and Rbb substituents with respect to Rbb and X1, respectively.
Additionally, as used throughout the application, including in the embodiments, when a group is drawn out as divalent, the left bond of the divalent group is attached to the group which is to its left in the remainder of the molecule, and the right bond of the divalent group is attached to the group which is to its right in the remainder of the molecule. For example, in the following divalent groups
Figure imgf000032_0005
the bond on the left of (a), (b) and (c) is attached to Z6 of L in the following structure :
Figure imgf000032_0001
and the
Figure imgf000032_0002
on the right side of (a), (b), and (c) (i.e., X1, X2, and X3) is attached to ring:
Figure imgf000032_0003
Similarly, for L i.e, -Z1-Z2-Z3-Z4-Z5-Z6-, the left side in L (i.e., Z1) is attached to an atom of an R4 group as denoted in this Application and Z6 is attached to X1, X2, X3, or X4. For example, when L is a group of formula:
Figure imgf000032_0004
and Degron is a group of formula (a), i.e.,
Figure imgf000033_0001
, the left bond of L (i.e., the -NH- group) is attached to X2 and the right hand bond of L (i.e., -SO2-) is attached to an R4 group as denoted in this Application.
The term “disease” as used herein is intended to be generally synonymous, and is used interchangeably with, the terms “disorder,” “syndrome,” and “condition” (as in medical condition), in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptoms, and causes the human or animal to have a reduced duration or quality of life.
The term “combination therapy” means the administration of two or more therapeutic agents to treat a disease or disorder described in the present disclosure. Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each active ingredient. In addition, such administration also encompasses use of each type of therapeutic agent in a sequential manner. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the conditions or disorders described herein.
The term “patient” is generally synonymous with the term “subject” and includes all mammals including humans. Examples of patients include humans, livestock such as cows, goats, sheep, pigs, and rabbits, and companion animals such as dogs, cats, rabbits, and horses. Preferably, the patient is a human.
“Treating” or “treatment” of a disease includes:
(1) preventing the disease, i.e., causing the clinical symptoms of the disease not to develop in a mammal that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease;
(2) inhibiting the disease, i.e., delaying, arresting (i.e., stabilizing) or reducing the development or severity of the disease or its clinical symptoms; or
(3) relieving the disease, i.e., causing regression of the disease or its clinical symptoms.
In one embodiment, treating or treatment of a disease includes inhibiting the disease, i.e., delaying, arresting or reducing the development or severity of the disease or its clinical symptoms; or relieving the disease, i.e., causing regression of the disease or its clinical symptoms. A “therapeutically effective amount” means the amount of a compound of the present disclosure or a pharmaceutically acceptable salt thereof that, when administered to a patient for treating a disease, is sufficient to affect such treatment for the disease. The “therapeutically effective amount” will vary depending on the compound, the disease and its severity and the age, weight, etc., of the mammal to be treated. The therapeutically effective amount of a K-ras inhibitor disclosed herein can be administered to the patient in a single dosage form or multiples thereof. For example, 600 mg dose of a K-ras inhibitor can be administered in a single 600 mg tablet or two 300 mg tablets.
The terms "inhibiting" and "reducing," or any variation of these terms in relation of K-Ras G12D, includes any measurable decrease or complete inhibition to achieve a desired result. For example, there may be a decrease of about, at most about, or at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or more, or any range derivable therein, reduction of K-Ras G12D GTPase activity; a decrease of K- Ras G12D GTP binding affinity or an increase of G12D GDP binding affinity; an increase of GTP off rate or a decrease of GDP off rate; a decrease of signaling transduction molecules levels downstream in the K-Ras pathway, e.g., a decrease in pERK level; and/or a decrease of K-Ras complex binding to downstream signaling molecules compared to normal.
Representative compounds of the disclosure made are disclosed in Compound Table 1 and 2 below:
Compound Table 1
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Compound Table 2
Figure imgf000053_0002
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0002
Contemplated compounds of Formula IA are disclosed in table 3 and 4 below
Compound Table 3
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Table 4.
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
5
Additional contemplated compounds of Formula (I) are disclosed below.
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Embodiments:
In embodiments 1 to 300, the present disclosure includes:
1A. In embodiment 1A, the compound of Formula (IA) or a pharmaceutically acceptable salt thereof is a compound according to structure (IA’):
Figure imgf000122_0001
where R1, R1a, R1b, R2, R3, R4, R5, n, m, L, U, V, W, and Degron are as defined in the first aspect of the Summary.
1. In embodiment 1, the compound of Formula (I) or a pharmaceutically acceptable salt thereof is a compound according to structure (la):
Figure imgf000122_0002
where R1, R1a, R1b, R2, R3, R4, R5, n, m, L, U, V, W, and Degron are as defined in the second aspect of the Summary. la. In embodiment la, the compound of embodiment IA or 1, a pharmaceutically acceptable salt is a compound according to structure (lb):
Figure imgf000122_0003
where R1, R1a, R1b, R2, R3, R4, R5, n, m, L and Degron are as defined in the first or second aspect of the Summary, respectivley. lb. In embodiment lb, the compound of embodiment 1 A or 1, or a pharmaceutically acceptable salt is a compound according to structure (Ic):
Figure imgf000123_0001
where R1, R1a, R1b, R2, R3, R4, R5, n, m, L and Degron are as defined in the first or second aspect of the Summary, respectively.
2. In embodiment 2, the compound of embodiment 1A, 1, la, or lb, or a pharmaceutically acceptable salt thereof, is wherein R6 is fused heterocyclylalkyl where fused heterocyclyl of fused heterocyclylalkyl is substituted with Ra, Rb, and Rc as defined therein.
3. In embodiment 3, the compound of embodiment 1 A, 1, la, or lb, or a pharmaceutically acceptable salt thereof, is wherein the fused heterocyclyl of fused heterocyclylalkyl of R6 is isoindolinyl substituted with Ra, Rb, and Rc as defined therein.
4. In embodiment 4, the compound of any one of embodiments 1 A to 3, or a pharmaceutically acceptable salt thereof, is wherein the fused heterocyclylalkyl of R6 is a ring of formula:
Figure imgf000123_0002
are as defined therein.
5. In embodiment 5, the compound of embodiment 1A, 1, la, or lb, or a pharmaceutically acceptable salt thereof, is wherein R6 is heterocyclylalkyl, bicyclic heterocyclyl, or bicyclic heterocylalkylalkyl, where heterocyclyl of heterocyclylalkyl and bicyclic heterocyclyl, by itself or as part of bicyclic heterocyclylalkyl are substituted with Rd, Re, and Rfas defined therein.
6. In embodiment 6, the compound of embodiment 1A, 1, 1a, lb, or 5, or a pharmaceutically acceptable salt thereof, is wherein R6 is heterocyclylalkyl where heterocyclyl of heterocyclylalkyl of R6 is substituted with Rd, Re, and Rf as defined therein. 7. In embodiment 7, the compound of any one of embodiments 1A, 1, la, lb, 5 and 6, or a pharmaceutically acceptable salt thereof, is wherein the heterocyclylalkyl of R6 is pyrrolidin-2- ylmethyl, piperidin-2-ylmethyl, or piperi din-3 -ylmethyl, preferably pyrrolidin-2-ylmethyl substituted with Rd, Re, and Rf as defined therein.
8. In embodiment 8, the compound of any one of embodiments 1A, 1, 1a, lb, and 5 to 7, or a pharmaceutically acceptable salt thereof, is wherein the heterocyclylalkyl of R6 is pyrrolidin-2- ylmethyl of structure
Figure imgf000124_0001
substituted with Re and Rf, preferably R6 is pyrrolidin-2-ylmethyl of structure
Figure imgf000124_0002
hydrogen.
9. In embodiment 9, the compound of any one of embodiments 1A, 1, 1a, lb, and 5, or a pharmaceutically acceptable salt thereof, is wherein R6 is bicyclic heterocyclalkylalkyl substituted with Rd, Re, and Rf as defined therein.
10. In embodiment 10, the compound of any one of embodiments 1A, 1, 1a, lb, 5, and 9, or a pharmaceutically acceptable salt thereof, is wherein the bicyclic heterocyclylalkyl of R6 is hexahydro- 1 H-pyrrolizin-7 a-ylalkyl-d2, preferably, hexahydro- 1 H-pyrrolizin-7a-ylmethyl-d2, where hexahydro- lH-pyrrolizin-7a-yl is substituted with Rd, Re, and Rf as defined therein.
11. In embodiment 11, the compound of any one of embodiments 1A, 1, 1a, lb, 5, 9, and 10, or a pharmaceutically acceptable salt thereof, is wherein the bicyclic heterocyclylalkyl is a ring of formula:
Figure imgf000124_0003
substituted with Re, and Rf as defined therein. 12. In embodiment 12, the compound of any one of embodiments 1A, 1, la, lb, 5, and 9 to
11 , or a pharmaceutically acceptable salt thereof, is wherein the bicyclic heterocyclylalkyl of R6 is a ring of formula:
Figure imgf000125_0001
where Rd, Re, Rf, are as defined therein.
13. In embodiment 13, the compound of any one of embodiments 1A, 1, la, lb, 5, and 9 to 12, or a pharmaceutically acceptable salt thereof, is wherein the bicyclic heterocyclylalkyl of R6 is a ring of formula:
, preferably is
Figure imgf000125_0003
where Rd and Rf are as defined therein.
Figure imgf000125_0002
14. In embodiment 14, the compound of embodiment 1A, 1, la, or lb, or a pharmaceutically acceptable salt thereof, is wherein R6 is fused bicyclic heterocyclyl, fused bicyclic heterocyclylalkyl, heterocyclyl fused bicyclic heterocyclyl, heterocyclyl fused bicyclic heterocyclylalkyl, tricyclic heterocyclyl, or tricyclic heterocyclylalkyl, wherein fused bicyclic heterocyclyl, by itself or as part of fused bicyclic heterocyclylalkyl, heterocyclyl fused bicyclic heterocyclyl, by itself or as part of heterocyclyl fused bicyclic heterocyclylalkyl, or tricyclic heterocyclyl, by itself or as part of tricyclic heterocyclylalkyl are independently substituted with Rg, Rh, and R1 as defined therein.
15 In embodiment 15, the compound of embodiment 1A, 1, la, lb, or 14, or a pharmaceutically acceptable salt thereof, is wherein R6 is fused bicyclic heterocyclyl substituted with Rg, Rh, , and R1 as defined therein.
16. In embodiment 16, the compound of embodiment 1A, 1, la, lb, or 15, or a pharmaceutically acceptable salt thereof, is wherein the fused bicyclic heterocyclyl of R6 is a ring of formula: where ring Al is phenyl or 5- or 6-membered heteroaryl and the
Figure imgf000126_0001
fused bicyclic heterocyclyl is additionally substituted with Rh and R1 where Rg, Rh and R1 are as defined as defined therein, preferably ring Al is phenyl or 5- or 6-membered heteroaryl substituted with Rh and R1 as defined therein.
17. In embodiment 17, the compound of embodiment 1A, 1, la, lb, 15, or 16, or a pharmaceutically acceptable salt thereof, is wherein the fused bicyclic heterocyclyl of R6 is a ring of formula:
Figure imgf000126_0002
where ring Al is phenyl, pyrazolyl, pyridinyl, or pyrimidinyl, each ring substituted with Rh and R1 as defined therein.
18. In embodiment 18, the compound of any one of embodiments 1A, 1, la, lb, and 14, or a pharmaceutically acceptable salt thereof, is wherein R6 is fused bicyclic heterocylalkylalkyl where fused bicyclic heterocyclyl of fused bicyclic heterocyclylalkyl is substituted with Rg, Rh, and R1 as defined therein.
19. In embodiment 19, the compound of embodiment 1A, 1, la, lb, or 14, or a pharmaceutically acceptable salt thereof, is wherein fused bicyclic heterocyclyl of fused bicyclic heterocyclylalkyl of R6 is a ring of formula:
Figure imgf000126_0003
where ring Al is phenyl or 5- or 6-membered heteroaryl and the fused bicyclic heterocyclyl is additionally substituted with Rh and R1 where Rg, Rh and R1 are as defined as defined therein.
20. In embodiment 20, the compound of embodiment 1A, 1, la, lb, 14, or 19, or a pharmaceutically acceptable salt thereof, is wherein the fused bicyclic heterocyclyl of fused bicyclic heterocyclylalkyl of R6 is a ring of formula:
Figure imgf000127_0001
where ring Al is phenyl, pyrazolyl, pyridinyl, or pyrimidinyl, each ring substituted with Rb and R1 as defined therein.
21. In embodiment 21, the compound of any one of embodiments 1A, 1, la, lb, and 14 to 20, or a pharmaceutically acceptable salt thereof, is wherein the fused bicyclic heterocyclyl and fused bicyclic heterocyclyl of fused bicyclic heterocyclylalkyl of R6 is 2,3 -dihydro- lH-pyrrolo[2,l- a]isoindol-9b(5H)-yl, 2,3-dihydro-lH-pyrrolo[l,2-a]indol-9a(9H)-yl, 1,3b, 4,5,6, 8-hexahydropyrrolo- [3,2-a]pyrrolizin-3b-yl, l-methyl-l,3b,4,5,6,8-hexahydropyrrolo[4,3-a]pyrrolizin-3b-yl, 4b, 6,7,9- tetrahydro-5H-pyrido[3,2-a]-pyrrolizin-4b-yl, 3,3a,4,5-tetrahydro-2H-pyrano[4,3,2-cd]isoindol-5-yl, or l,2,3,5,10,10a-hexahydropyrrolo[l,2-b]isoquinolin-10a-yl, each ring substituted with Rg, Rh, and R1 as defined therein.
22. In embodiment 22, the compound of any one of embodiments 1A, 1, la, lb, and 14, or a pharmaceutically acceptable salt thereof, is wherein R6 is tricyclic heterocyclyl substituted with Rg, Rb, and R1 as defined therein.
23. In embodiment 23, the compound of any one of embodiments 1A, 1, la, lb, and 14, or a pharmaceutically acceptable salt thereof, is wherein R6 is tricyclic heterocyclylalkyl where tricyclic heterocyclyl of tricyclic heterocyclylalkyl is substituted with Rg, Rb, and R1 as defined therein.
24. In embodiment 24, the compound of any one of embodiments 1 A to 23, or a pharmaceutically acceptable salt thereof, is wherein R7, R9, and R11 are independently hydrogen, deuterium, alkyl, fluoro, or haloalkyl, Rb, Re, and Rb are a bond to L, and Rc, Rf, and R1 are independently hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, or hydroxy.
25. In embodiment 25, the compound of any one of embodiments 1 A to 23, or a pharmaceutically acceptable salt thereof, is wherein R7, R9, and R11 are independently hydrogen, deuterium, alkyl, fluoro, or haloalkyl, Rb, Re, and Rb are independently hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, hydroxy, -(alkylene)n1-OC(O)NR13R13a and Rc, Rf, and R1 are independently a bond to L.
26. In embodiment 26, the compound of any one of embodiments 1A to 23, or a pharmaceutically acceptable salt thereof, is wherein R7, R9, and R11 are independently a bond to L, Rb, Re, and Rb are independently hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, hydroxy, or -(alkyl ene)ni- OC(O)NR13R13a and Rc, Rf, and R1 are independently hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, or hydroxy.
27. In embodiment 27, the compound of any one of embodiments 1A to 25, or a pharmaceutically acceptable salt thereof, is wherein R7, R9, and R11 are independently hydrogen, deuterium, methyl, ethyl, propyl, fluoro, difluoromethyl, or trifluoromethyl.
28. In embodiment 28, the compound of any one of embodiments 1A to 25, and 27, or a pharmaceutically acceptable salt thereof, is wherein R7, R9, and R11 are independently hydrogen.
29. In embodiment 29, the compound of any one of embodiments 1A to 25, and 27, or a pharmaceutically acceptable salt thereof, is wherein R7, R9, and R11 are independently deuterium.
30. In embodiment 30, the compound of any one of embodiments 1A to 25, or a pharmaceutically acceptable salt thereof, is wherein R7, R9, and R11 are independently alkyl, preferably methyl, ethyl, or propyl, preferably methyl.
31. In embodiment 31 , the compound of any one of embodiments 1 A to 25, and 27, or a pharmaceutically acceptable salt thereof, is wherein R7, R9, and R11 are independently fluoro.
32. In embodiment 32, the compound of any one of embodiments 1A to 25, or a pharmaceutically acceptable salt thereof, is wherein R7, R9, and R11 are independently haloalkyl, preferably fluoromethyl, difluoromethyl, or trifluoromethyl, preferably trifluoromethyl.
33. In embodiment 33, the compound of any one of embodiments 1A to 32, or a pharmaceutically acceptable salt thereof, is wherein R8, R10, and R12 are independently hydrogen, deuterium, alkyl, halo, haloalkyl, cyano, alkyloxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, phenyl, or heteroaryl.
34. In embodiment 34, the compound of any one of embodiments 1A to 33, or a pharmaceutically acceptable salt thereof, is wherein R8, R10, and R12 are independently hydrogen, deuterium, fluoro, methyl, cyano, methoxymethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclopropylethyl, cyclobutylethyl, cyclopentylethyl, phenyl, pyrrolidinyl, furanyl, pyranyl, piperidinyl, morpholinyl, or 5- or 6- membereing heteroaryl (such as pyridyl, pyrimidinyl, or pyrazolyl).
35. In embodiment 35, the compound of any one of embodiments 1A to 34, or a pharmaceutically acceptable salt thereof, is wherein R8, R10, and R12 are independently hydrogen. 36. In embodiment 36, the compound of any one of embodiments 1A to 34, or a pharmaceutically acceptable salt thereof, is wherein R8, R10, and R12 are independently fluoro ormethyl.
37. In embodiment 37, the compound of any one of embodiments 1A to 34, or a pharmaceutically acceptable salt thereof, is wherein R8, R10, and R12 are independently hydrogen or cyano.
38. In embodiment 38, the compound of any one of embodiments 1A to 34, or a pharmaceutically acceptable salt thereof, is wherein R8, R10, and R12 are methoxymethyl.
39. In embodiment 39, the compound of any one of embodiments 1A to 34, or a pharmaceutically acceptable salt thereof, is wherein R8, R10, and R12 are independently cyclopropyl, cyclobutyl, cyclopentyl, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclopropylethyl, cyclobutylethyl, or cyclopentylethyl.
40. In embodiment 40, the compound of any one of embodiments 1A to 34, or a pharmaceutically acceptable salt thereof, is wherein R8, R10, and R12 are phenyl, pyrrolidinyl, furanyl, pyranyl, piperidinyl, morpholinyl, or 5- or 6-membereing heteroaryl (such as pyridyl, pyrimidinyl, or pyrazolyl).
41. In embodiment 41, the compound of any one of embodiments 1 A to 23, or a pharmaceutically acceptable salt thereof, is wherein, independently of each other, R7 andR8, R9 and R10, and R11 and R12 together with the carbon atom to which they are attached form cycloalkylene optionally substituted with alkyl, halo, alkoxy, or hydroxy, Rb, Re, and Rb are a bond to L, and Rc, Rf, and R1 are independently hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, or hydroxy.
42. In embodiment 42, the compound of any one of embodiments 1A to 23, or a pharmaceutically acceptable salt thereof, is wherein, independently of each other, R7 andR8, R9 and R10, and R11 and R12 together with the carbon atom to which they are attached form cycloalkylene optionally substituted with alkyl, halo, alkoxy, or hydroxy, Rb, Re, and Rb are independently hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, hydroxy, -(alkylene)n1-OC(O)NR13R13a and Rc, Rf, and R1 are independently a bond to L.
43. In embodiment 43, the compound of any one of embodiments 1A to 23, 41, and 42, or a pharmaceutically acceptable salt thereof, is wherein R7 and R8, R9 and R10, and R11 and R12 together with the carbon atom to which they are attached form cyclopropyl, cyclobutylene, or cyclopentylene, each ring optionally substituted with methyl, fluoro, or methoxy. 44. In embodiment 44, the compound of any one of embodiments 1A to 23, 25 to 40, 42, and 43, or a pharmaceutically acceptable salt thereof, is wherein Rb, Re, and Rb are independently hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, or hydroxy.
45. In embodiment 45, the compound of any one of embodiments 1A to 23, 25 to 40, and 42 to 44, or a pharmaceutically acceptable salt thereof, is wherein Rb, Re, and Rb are independently hydrogen, methyl, fluoro, methoxy, ethoxy, or methoxymethyl.
46. In embodiment 46, the compound of any one of embodiments 1A to 23, 25 to 40, and 42 to 45, or a pharmaceutically acceptable salt thereof, is wherein Rb, Re, and Rb are hydrogen.
47. In embodiment 47, the compound of any one of embodiments 1A to 23 and 25 to 45, or a pharmaceutically acceptable salt thereof, is wherein Rb, Re, and Rb are alkyl, preferably methyl.
48. In embodiment 48, the compound of any one of embodiments 1 A to 23 and 25 to 45, or a pharmaceutically acceptable salt thereof, is wherein Rb, Re, and Rb are halo, preferably fluoro.
49. In embodiment 49, the compound of any one of embodiments 1A to 23 and 25 to 45, or a pharmaceutically acceptable salt thereof, is wherein Rb, Re, and Rb are alkoxy, preferably methoxy or ethoxy.
50. In embodiment 50, the compound of any one of embodiments 1A to 23, 25 to 40, and 42 to 45, or a pharmaceutically acceptable salt thereof, is wherein Rb, Re, and Rb are alkoxyalkyl, preferably methoxymethyl.
51. In embodiment 51 , the compound of any one of embodiments 1 A to 23, 25 to 40, 42 and 43, or a pharmaceutically acceptable salt thereof, is wherein Rb, Re, and Rb are independently - ( alkyl ene)ni-OC(O)NR13R13a.
52. In embodiment 52, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51, or a pharmaceutically acceptable salt thereof, is wherein nl is 0.
53. In embodiment 53, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51, or a pharmaceutically acceptable salt thereof, is wherein nl is 1.
54. In embodiment 54, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51, and 53, or a pharmaceutically acceptable salt thereof, is wherein alkylene is methylene, ethylene, -CH(CH3)-, or -C(CH3)2-, preferably methylene.
55. In embodiment 55, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 54, or a pharmaceutically acceptable salt thereof, is wherein R13 is hydrogen, deuterium, alkyl, deuterioalkyl, haloalkyl, haloalkoxyalkyl, or alkoxyalkyl and R13a is deuterioalkyl, cycloalkyl, alkoxy, alkoxyalkyl, haloalkyl, haloalkoxyalkyl, or heterocyclyl optionally substituted with one or two substituents independently selected from alkyl, halo, alkoxy, haloalkoxy, and haloalkyl.
56. In embodiment 56, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 55, or a pharmaceutically acceptable salt thereof, is wherein R13 is hydrogen or deuterium.
57. In embodiment 57, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 55, or a pharmaceutically acceptable salt thereof, is wherein R13 is alkyl.
58. In embodiment 58, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 55, or a pharmaceutically acceptable salt thereof, is wherein R13 is haloalkyl.
59. In embodiment 59, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 55, or a pharmaceutically acceptable salt thereof, is wherein R13 is haloalkoxyalkyl.
60. In embodiment 60, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 55, or a pharmaceutically acceptable salt thereof, is wherein R13 is alkoxyalkyl.
61. In embodiment 61, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 55, or a pharmaceutically acceptable salt thereof, is wherein R13 is deuterioalkyl.
62. In embodiment 62, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 61, or a pharmaceutically acceptable salt thereof, is wherein R13a is alkoxy.
63. In embodiment 63, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 61, or a pharmaceutically acceptable salt thereof, is wherein R13a is alkoxyalkyl.
64. In embodiment 64, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 61, or a pharmaceutically acceptable salt thereof, is wherein R13a is haloalkyl.
65. In embodiment 65, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 61, or a pharmaceutically acceptable salt thereof, is wherein R13a is haloalkoxyalkyl.
66. In embodiment 66, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 61, or a pharmaceutically acceptable salt thereof, is wherein R13a is cycloalkyl.
67. In embodiment 67, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 61, or a pharmaceutically acceptable salt thereof, is wherein R13a is deuterioalkyl.
68. In embodiment 68, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 61, or a pharmaceutically acceptable salt thereof, is wherein R13a is heterocyclyl substituted with Ri and Rk independently selected from alkyl, halo, alkoxy, haloalkoxy, and haloalkyl. 69. In embodiment 69, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 68, or a pharmaceutically acceptable salt thereof, is wherein R13 is hydrogen, methyl, methyl-d3, methoxyethyl, ethoxyethyl, or propoxyethyl; and R13ais methyl-d3, cyclopropyl, 2,2- difluoroethyl, 2,2,2-trifluoroethyl, 2,2-difluoroethoxy, 2,2,2-trifluoroethoxy, methoxyethyl, ethoxyethyl, oxetan-3-yl, tetrahydrofuranyl, or tetrahydropyranyl.
70. In embodiment 70, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, and 51 to 54, or a pharmaceutically acceptable salt thereof, is wherein R13 and R13a together with the nitrogen atom to which they are attached form heterocyclyl, bicyclic heterocyclyl, bridged heterocyclyl, fused heterocyclyl, or spiroheterocyclyl wherein (a) heterocyclyl is substituted with Rm, Rn, R°, and Rp and (b) bicyclic heterocyclyl, bridged heterocyclyl, fused heterocyclyl, and spiroheterocyclyl are independently substituted with Rq, Rr, and Rs.
71. In embodiment 71, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54 and 70, or a pharmaceutically acceptable salt thereof, is wherein R13 and R13a together with the nitrogen atom to which they are attached form heterocyclyl substituted with Rm, Rn, R°, and RP.
72. In embodiment 72, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54 and 70, or a pharmaceutically acceptable salt thereof, is wherein R13 and R14 together with the nitrogen atom to which they are attached form bicyclic heterocyclyl, bridged heterocyclyl, fused heterocyclyl, or spiroheterocyclyl, each ring independently substituted with Rr, Rs, and Rt.
73. In embodiment 73, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, 70, and 71, or a pharmaceutically acceptable salt thereof, is wherein R13 and R13a together with the nitrogen atom to which they are attached form azetidin-l-yl, pyrrolidin- 1 -yl, piperidin- 1 -yl, piperazin-l-yl, morpholin-l-yl, or homomorpholin- 1 -yl, each ring substituted with Rm, Rn, R°, and Rp.
74. In embodiment 74, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, 70, 71, and 73, or a pharmaceutically acceptable salt thereof, is wherein R13 and R13a together with the nitrogen atom to which they are attached form azetidin-l-yl, pyrrolidin- 1 -yl, piperidin- 1 -yl, piperazin-l-yl, morpholin-l-yl, or homomorpholin- 1 -yl, each ring substituted with Rm, Rn, R°, and Rp where Rm and Rn are independently selected from hydrogen, deuterium, methyl, fluoro, difluoromethyl, trifluoromethyl, difluoromethoxy, trifluoromethoxy, cyano, or methoxy, R° is hydrogen, deuterium, methoxymethyl, or fluoro, and Rp is hydrogen, deuterium, or fluoro. 75. In embodiment 75, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, 70, 71, 73, and 74, or a pharmaceutically acceptable salt thereof, is wherein Rm, Rn, R°, and Rp are hydrogen.
76. In embodiment 76, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, 70, 71, 73, and 74, or a pharmaceutically acceptable salt thereof, is wherein R13 and R13a together with the nitrogen atom to which they are attached form 3-methoxymethyl-azetidin-l-yl, 2- methoxymethyl-piperidin-l-yl, 3,3,4,4-tetrafluoropyrrolidin-l-yl, morpholin-l-yl. 2,6- dimethylmorpholin-4-yl, 2,2-dimethylmorpholin-4-yl, 2-(trifluoromethyl)morpholin-4-yl, 2,2- difluoromorpholin-4-yl, 2-methylmorpholin-4-yl, 3-methylmorpholin-4-yl, or 2- (difluoromethyl)morpholin-4-yl.
77. In embodiment 77, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, 70, 71, and 73 to 76, or a pharmaceutically acceptable salt thereof, is wherein R13 and R13a together with the nitrogen atom to which they are attached form morpholin-l-yl. 2,6- dimethylmorpholin-4-yl, 2,2-dimethylmorpholin-4-yl, 2-(trifluoromethyl)morpholin-4-yl, 2,2- difluoromorpholin-4-yl, 2-methylmorpholin-4-yl, 3-methylmorpholin-4-yl, or 2- (difluoromethyl)morpholin-4-yl.
78. In embodiment 78, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, and 70, or a pharmaceutically acceptable salt thereof, is wherein R13 and R13a together with the nitrogen atom to which they are attached form bicyclic heterocyclyl substituted with Rq, Rr, and Rs.
79. In embodiment 79, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, and 70, or a pharmaceutically acceptable salt thereof, is wherein R13 and R13a together with the nitrogen atom to which they are attached form bridged heterocyclyl substituted with Rq, Rr, and Rs.
80. In embodiment 80, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, and 70, or a pharmaceutically acceptable salt thereof, is wherein R13 and R13a together with the nitrogen atom to which they are attached form fused heterocyclyl substituted with Rq, Rr, and Rs.
81. In embodiment 8q, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, and 70, or a pharmaceutically acceptable salt thereof, is wherein R13 and R13a together with the nitrogen atom to which they are attached form spiroheterocyclyl substituted with Rq, Rr, and
Rs.
82. In embodiment 82, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 51 to 54, and 70, or a pharmaceutically acceptable salt thereof, is wherein R13 and R13a together with the nitrogen atom to which they are attached form a ring selected from:
Figure imgf000134_0001
each ring substituted with Rq, Rr, and Rs .
83. In embodiment 83, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 70 and 78 to 82, or a pharmaceutically acceptable salt thereof, is wherein Rs is hydrogen.
84. In embodiment 84, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 70 and 78 to 83, or a pharmaceutically acceptable salt thereof, is wherein Rrand Rs are hydrogen.
85. In embodiment 85, the compound of any one of embodiments 1A to 23, 25 to 40, 42, 43, 70 and 78 to 84, or a pharmaceutically acceptable salt thereof, is wherein Rq, Rr, and Rs (where applicable) are independently selected from hydrogen, methyl, methoxy, or fluoro.
86. In embodiment 86, the compound of any one of embodiments 1A to 24 and 26 to 85, or a pharmaceutically acceptable salt thereof, is wherein Rc, Rf, and R1 are hydrogen.
87. In embodiment 87, the compound of any one of embodiments 1A to 24 and 26 to 85, or a pharmaceutically acceptable salt thereof, is wherein Rc, Rf, and R1 are deuterium.
88. In embodiment 88, the compound of any one of embodiments 1A to 24 and 26 to 85, or a pharmaceutically acceptable salt thereof, is wherein Rc, Rf, and R1 are independently methyl, methoxy, methyloxy, chloro, or fluoro. 89. In embodiment 89, the compound of any one of embodiments 1A to 24 and 26 to 85, or a pharmaceutically acceptable salt thereof, is wherein Rc, Rf, and R1 are independently chloro or fluoro, preferably, Rc, Rf, and R1 are fluoro.
90. In embodiment 90, the compound of any one of embodiments 1A to 89, or a pharmaceutically acceptable salt thereof, is wherein Z is O.
91. In embodiment 91, the compound of any one of embodiments 1A to 90, or a pharmaceutically acceptable salt thereof, is wherein m and n are each 1 , or one of m and n is i and the other of m and n is 2.
92. In embodiment 92, the compound of any one of embodiments 1A to 90, or a pharmaceutically acceptable salt thereof, is wherein m and n are each 1 , or one of m and n is i.
93. In embodiment 93, the compound of any one of embodiments 1A to 90, or a pharmaceutically acceptable salt thereof, is wherein one of m and n is 1 and the other of m and n is 2.
94. In embodiment 94, the compound of any one of embodiments 1A to 90, or a pharmaceutically acceptable salt thereof, is wherein m and n are each 1.
95. In embodiment 95, the compound of any one of embodiments 1A to 90, or a pharmaceutically acceptable salt thereof, is wherein m is 1 and n is 3.
96. In embodiment 06, the compound of any one of embodiments 1A to 95, or a pharmaceutically acceptable salt thereof, is wherein R1 and R1a are independently selected from hydrogen, methyl, and ethyl, and R1b and Rlc are hydrogen.
97. In embodiment 97, the compound of any one of embodiments 1A to 95, or a pharmaceutically acceptable salt thereof, is wherein R1 is cyanomethyl and R1a is hydrogen, methyl, or ethyl, preferably R1b is hydrogen, and R1b and Rlc are hydrogen.
98. In embodiment 98, the compound of any one of embodiments 1A to 95, or a pharmaceutically acceptable salt thereof, is wherein R1 and R1a are attached to the carbon atoms of the ring that are opposite or diagonal to each other and combine to form -(CH2)Z- where z is 1, 2, or 3, preferably z is 2, and R1b and Rlc are hydrogen.
99. In embodiment 99, the compound of any one of embodiments 1A to 95, or a pharmaceutically acceptable salt thereof, is wherein R1 and R1a are attached to the carbon atoms of the ring that are opposite or diagonal to each other and combine to form -(CH2)Z- where z is 1, 2, or 3, preferably 2, Rlc is hydrogen and R1b is attached to a carbon of the -(CH2)Z- group and is alkylidenyl, preferably =CH2. 100. In embodiment 100, the compound of any one of embodiments 1A to 92, 94, and 98, or a pharmaceutically acceptable salt thereof, is wherein
Figure imgf000136_0002
is:
Figure imgf000136_0001
.
101. In embodiment 101, the compound of any one of embodiments 1A to 100, or a pharmaceutically acceptable salt thereof, is wherein R5 is -Q-R14 where Q is bond and R14 is cycloalkyl, fused cycloalkyl, fused spirocycloalkyl, aryl, heteroaryl, or fused heteroaryl, wherein aryl, heteroaryl, and fused heteroaryl are substituted with Rt Ru, Rv, and Rw wherein Rt and Ru are independently selected from hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, hydroxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, and cyano, Rvis hydrogen, alkenyl, alkynyl, cyanoalkenyl, cyanoalkynyl, or halo, and Rw is hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, alkoxy, alkylthio, heteroalkyl, hydroxyalkyl, amino, cyano, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, or optionally substituted heterocyclylalkyl.
102. In embodiment 102, the compound of any one of embodiments 1A to 100, or a pharmaceutically acceptable salt thereof, is wherein R5 is -Q-R14 where Q is alkylene and R14is cycloalkyl, aryl, or fused heteroaryl, wherein aryl, fused heteroaryl, and heteroaryl are substituted with Rt Ru, Rv, and Rwwherein Rt and Ru are independently selected from hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, hydroxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, and cyano, Rv is hydrogen, alkynyl, or halo, and Rwis hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, cyano, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, or optionally substituted heterocyclylalkyl.
103. In embodiment 103, the compound of any one of embodiments 1A to 100, or a pharmaceutically acceptable salt thereof, is wherein R5 is -Q-R14 where Q is -C(O)- and R14 is cycloalkyl, aryl, fused heteroaryl, or heteroaryl, wherein aryl, fused heteroaryl and heteroaryl are substituted with Rt Ru, Rv, and Rwwherein Rt and Ru are independently selected from hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, hydroxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, and cyano, Rv is hydrogen, alkynyl, or halo, and Rw is hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, cyano, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, or optionally substituted heterocyclylalkyl. 104. In embodiment 104, the compound of any one of embodiments 1 A to 101, or a pharmaceutically acceptable salt thereof, is wherein R14 is cycloalkyl, fused cycloalkyl, aryl, or heteroaryl wherein aryl, and heteroaryl are substituted with Rt Ru, Rv, and Rw wherein Rt and Ru are independently selected from hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, hydroxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, and cyano, Rv is hydrogen, alkenyl, alkynyl, cyanoalkenyl, cyanoalkynyl, or halo, and Rw is hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, cyano, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, or optionally substituted heterocyclylalkyl.
105. In embodiment 105, the compound of any one of embodiments 1 A to 101, or a pharmaceutically acceptable salt thereof, is wherein R5 is -Q-R14 where Q is bond and R14 is phenyl or naphthyl substituted with Rt Ru, Rv, and Rw.
105a. In embodiment 105a, the compound of embodiment 105, or a pharmaceutically acceptable salt thereof, is wherein Rt and Ru are independently selected from hydrogen, halo, and hydroxyl, Rw is independently selected from hydrogen or halo, and Rv is hydrogen or alkynyl.
105b. In embodiment 105b, the compound of embodiment 105, or a pharmaceutically acceptable salt thereof, is wherein R14 is:
Figure imgf000137_0001
, substituted with Rt Ru, and Rw where Rv is alkynyl.
105c. In embodiment 105c, the compound of embodiment 105, or a pharmaceutically acceptable salt thereof, is wherein R14 is:
Figure imgf000137_0002
, substituted with Rt Ru, and Rw wherein Rv is ethenyl.
106. In embodiment 122, the compound of any one of embodiments 1A to 101 and 105, or a pharmaceutically acceptable salt thereof, is wherein R5 is -Q-R14 where Q is bond and R14 is phenyl or naphthyl substituted with Rt Ru, Rv, and Rw where Rt and Ru are independently selected from hydrogen, alkyl, halo, haloalkyl, hydroxy, alkoxy, haloalkoxy, cycloalkyl, amino, cyano, and hydroxyalkyl, Rv is hydrogen, fluoro, alkynyl, and Rwis hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, cyano, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, or optionally substituted heterocyclylalkyl.
107. In embodiment 107, the compound of any one of embodiments 1A to 105 and 106, or a pharmaceutically acceptable salt thereof, is wherein Rt and Ru independently selected from hydrogen, methyl, ethyl, fluoro, chloro, trifluoromethyl, difluoromethyl, trifluoromethoxy, hydroxy, methyl, ethoxy, cyclopropyl, amino, cyano, and hydroxymethyl, Rv is hydrogen, ethynyl, 2- cyano vinyl, 2-cyanoethyn-l-yl, or fluoro, and Rwis hydrogen, methyl, fluoro, amino, or cyclopropyl.
108. In embodiment 108, the compound of any one of embodiments 1A to 101, or a pharmaceutically acceptable salt thereof, is wherein R5 is -Q-R14 where Q is bond and R14 is heteroaryl or fused heteroaryl substituted with Rt Ru, Rv, and Rw.
109. In embodiment 109, the compound of any one of embodiments 1A to 101 and 108, or a pharmaceutically acceptable salt thereof, is wherein R5 is -Q-R14 where Q is bond and R14 is a monocyclic heteroaryl (e.g., pyridyl, pyrimidinyl, benzothiazolyl) substituted with Rt Ru, Rv, and Rw.
110. In embodiment 110, the compound of any one of embodiments 1A to 101 and 108, or a pharmaceutically acceptable salt thereof, is wherein R5 is -Q-R14 where Q is bond and R14 is bicyclic heteroaryl (e,g, quinolinyl, isoquinolinyl, or indazolyl), substituted with Rt Ru, Rv, and Rw.
111. In embodiment 111, the compound of any one of embodiments 1A to 101 and 108 to
110, or a pharmaceutically acceptable salt thereof, is wherein the heteroaryl is substituted with Rt Ru, Rv, and Rw where Rt and Ruare independently selected from hydrogen, alkyl, halo, haloalkyl, hydroxy, alkoxy, haloalkoxy, cycloalkyl, amino, cyano, and hydroxyalkyl and Rwis hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, cyano, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, or optionally substituted heterocyclylalkyl and Rv is hydrogen.
112. In embodiment 112, the compound of any one of embodiments 1A to 101 and 108 to
110, or a pharmaceutically acceptable salt thereof, is wherein Rt and Ru are independently selected from hydrogen, methyl, ethyl, fluoro, chloro, trifluoromethyl, difluoromethyl, trifluoromethoxy, hydroxy, methyl, ethoxy, cyclopropyl, amino, cyano, and hydroxymethyl, Rv is hydrogen or fluoro, and Rwis hydrogen, methyl, fluoro, amino, or cyclopropyl.
113. In embodiment 113, the compound of any one of embodiments 1A to 101 and 104 to
112, or a pharmaceutically acceptable salt thereof, is wherein R5 is -Q-R14 where Q is bond and R14 is:
Figure imgf000139_0001
113. In embodiment 113, the compound of any one of embodiments 1A to 101, 103-105, and 112, or a pharmaceutically acceptable salt thereof, is wherein R5 is -Q-R14 where Q is bond and R14 is:
Figure imgf000140_0001
114. In embodiment 114, the compound of any one of embodiments 1A to 101, 103-105,
112, and 113, or a pharmaceutically acceptable salt thereof, is wherein R5 is -Q-R14 where Q is bond and R14 is:
Figure imgf000140_0002
115. In embodiment 115, the compound of any one of embodiments 1 A to 114, or a pharmaceutically acceptable salt thereof, is wherein R2 is hydrogen, halo, or alkyl, and R3 is hydrogen, halo, cycloalkyl, cycloalkyloxy, or alkyl.
116. In embodiment 116, the compound of any one of embodiments 1A to 115, or a pharmaceutically acceptable salt thereof, is wherein R2 and R3 are each hydrogen.
117. In embodiment 117, the compound of any one of embodiments 1A to 115, or a pharmaceutically acceptable salt thereof, is wherein R2 is hydrogen or chloro and R3 is hydrogen, fluoro, cyclopropyl, or cyclopropyloxy.
118. In embodiment 118, the compound of any one of embodiments 1 A to 115, or a pharmaceutically acceptable salt thereof, is wherein R2 is hydrogen and R3 is fluoro or cyclopropyl.
119. In embodiment 119, the compound of any one of embodiments 1 A to 115 and 118, or a pharmaceutically acceptable salt thereof, is wherein R2 is hydrogen and R3 is fluoro or cyclopropyl and R3 is attached to C-8, the carbon substituted with R5 being C-7.
120. In embodiment 120, the compound of any one of embodiments 1A to 119, or a pharmaceutically acceptable salt thereof, is wherein the Degron is an E3 ligase ligand of formula (i):
Figure imgf000140_0003
(i).
121. In embodiment 121, the compound of any one of embodiments 1A to 120, or a pharmaceutically acceptable salt thereof, is wherein ring A of the E3 ligase ligand of formula (i) is a group of formula (a):
Figure imgf000141_0001
122. In embodiment 122, the compound of any one of embodiments 1 A to 121, or a pharmaceutically acceptable salt thereof, is wherein R20 and R21 are independently hydrogen or alkyl.
123. In embodiment 123, the compound of any one of embodiments 1A to 122, or a pharmaceutically acceptable salt thereof, is wherein R20 and R21 are hydrogen.
124. In embodiment 124, the compound of any one of embodiments 1A to 122, or a pharmaceutically acceptable salt thereof, is wherein R20 is hydrogen and R21 is methyl.
125. In embodiment 125, the compound of any one of embodiments 1 A to 121, or a pharmaceutically acceptable salt thereof, is wherein R20 and R21 together with the carbon to which they are attached form >C=O.
126. In embodiment 126, the compound of any one of embodiments 1A to 120, or a pharmaceutically acceptable salt thereof, is wherein the ring A of the E3 ligase ligand of formula (i) is a group of formula (b):
Figure imgf000141_0002
127. In embodiment 127, the compound of any one of embodiments 1A to 120 and 126, or a pharmaceutically acceptable salt thereof, is wherein R22 is hydrogen.
128. In embodiment 128, the compound of any one of embodiments 1A to 120 and 126, or a pharmaceutically acceptable salt thereof, wherein R22 is alkyl, preferably methyl. 129. In embodiment 129, the compound of any one of embodiments 1A to 120, or a pharmaceutically acceptable salt thereof, is wherein ring A of the E3 ligase ligand of formula (i) is a group of formula (c):
Figure imgf000142_0001
130. In embodiment 130, the compound of any one of embodiments 1A to 128, or a pharmaceutically acceptable salt thereof, is wherein ring A of the E3 ligase ligand of formula (i) is:
Figure imgf000142_0002
131. In embodiment 131, the compound of any one of embodiments 1A to 128 and 130, or a pharmaceutically acceptable salt thereof, is wherein ring A of the E3 ligase ligand of formula (i) is:
Figure imgf000142_0003
132. In embodiment 132, the compound of any one of embodiments 1A to 128, 130, and
131 , or a pharmaceutically acceptable salt thereof, is wherein ring A of the E3 ligase ligand of formula (i) is:
Figure imgf000143_0001
132a. In embodiment 132a, the compound of any one of embodiments 1A to 128, 130, and
131 , or a pharmaceutically acceptable salt thereof, is wherein ring A of the E3 ligase ligand of formula (i) is:
Figure imgf000143_0002
133. In embodiment 133, the compound of any one of embodiments 1A to 128, 130, and
131 , or a pharmaceutically acceptable salt thereof, is wherein ring A of the E3 ligase ligand of formula (i) is:
Figure imgf000143_0003
134. In embodiment 134, the compound of any one of embodiments 1A to 128, 130, and
131 , or a pharmaceutically acceptable salt thereof, is wherein ring A of the E3 ligase ligand of formula (i) is:
Figure imgf000143_0004
135. In embodiment 135, the compound of any one of embodiments 1A to 128, 130, and
131 , or a pharmaceutically acceptable salt thereof, is wherein ring A of the E3 ligase ligand of formula (i) is:
Figure imgf000144_0001
136. In embodiment 136, the compound of any one of embodiments 1A to 128, 130, and
131 , or a pharmaceutically acceptable salt thereof, is wherein ring A of the E3 ligase ligand of formula (i) is:
Figure imgf000144_0002
137. In embodiment 137, the compound of any one of embodiments 1A to 128, 130, and
131 , or a pharmaceutically acceptable salt thereof, is wherein ring A of the E3 ligase ligand of formula (i) is:
Figure imgf000144_0003
138. In embodiment 138, the compound of any one of embodiments 1A to 128, and 130 to 134, or a pharmaceutically acceptable salt thereof, is wherein Raa, Rbb, Rcc, and Rdd are independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy.
139. In embodiment 139, the compound of any one of embodiments 1 to 128, and 130 to 134, or a pharmaceutically acceptable salt thereof, is wherein Raa, Rbb, Rcc, and Rdd are independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, and cyano.
140. In embodiment 140, the compound of any one of embodiments 1A to 128, 130 to 134, and 138, or a pharmaceutically acceptable salt thereof, is wherein Raa, Rbb, Rcc, and Rdd are independently selected from hydrogen, methyl, methoxy, ethoxy, fluoro, trifluoromethyl, difluoromethyl, and trifluoromethoxy. 141. In embodiment 141, the compound of any one of embodiments 1A to 128, 130 to 134, and 138, or a pharmaceutically acceptable salt thereof, is wherein Raa, Rbb, Rcc, and Rdd are independently selected from hydrogen and methyl.
142. In embodiment 142, the compound of any one of embodiments 1A to 128, 130 to 134, and 138, or a pharmaceutically acceptable salt thereof, is wherein Raa, Rbb, Rcc, and Rdd are independently selected from hydrogen and methoxy.
143. In embodiment 143, the compound of any one of embodiments 1A to 128, 130 to 134, and 138, or a pharmaceutically acceptable salt thereof, is wherein Raa, Rbb, Rcc, and Rdd are independently selected from hydrogen and fluoro.
143a. In embodiment 143a, the compound of any one of embodiments 1A to 128, 130 to 134, and 138, or a pharmaceutically acceptable salt thereof, is wherein Raa, Rbb, Rcc, and Rdd are hydrogen.
144. In embodiment 144, the compound of any one of embodiments 1A to 128, 130 to 134, and 138, or a pharmaceutically acceptable salt thereof, is wherein Raa, Rbb, Rcc, and Rdd are independently selected from hydrogen, trifluoromethyl, and difluoromethyl.
145. In embodiment 145, the compound of any one of embodiments 1A to 128, 130 to 134, and 138, or a pharmaceutically acceptable salt thereof, is wherein Raa, Rbb, Rcc, and Rdd are independently selected from hydrogen and trifluoromethoxy.
146. In embodiment 146, the compound of any one of embodiments 1A to 128, 130 to 134, and 138, or a pharmaceutically acceptable salt thereof, is wherein Raa, Rbb, Rcc, and Rdd are independently selected from hydrogen, fluoro, and trifluoromethyl.
146a. In embodiment 146a, the compound of any one of embodiments 1A to 146, or a pharmaceutically acceptable salt thereof, is wherein R15 is hydrogen.
147. In embodiment 147, the compound of any one of embodiments 1A to 119, or a pharmaceutically acceptable salt thereof, is wherein the Degron is an E3 ligase ligand of formula (ii):
Figure imgf000145_0001
(ii).
148. In embodiment 148, the compound of any one of embodiments 1A to 119 and 147, or a pharmaceutically acceptable salt thereof, is wherein Ya is CH. 149. In embodiment 149, the compound of any one of embodiments 1A to 119 and 147, or a pharmaceutically acceptable salt thereof, is wherein Ya is N.
150. In embodiment 150, the compound of any one of embodiments 1A to 119, and 147-
149, or a pharmaceutically acceptable salt thereof, is wherein Za is a bond, -NH-, O, or -NHC(O)-.
151. In embodiment 152, the compound of any one of embodiments 1A to 119, and 147-
150, or a pharmaceutically acceptable salt thereof, is wherein Za is a bond, -NH-, or -NHC(O)-.
152. In embodiment 152, the compound of any one of embodiments 1A to 119, and 147- 150, or a pharmaceutically acceptable salt thereof, is wherein Za is a bond.
153. In embodiment 153, the compound of any one of embodiments 1A to 119, and 147- 150, or a pharmaceutically acceptable salt thereof, is wherein Za is -NH-, or -NHC(O)-.
154. In embodiment 154, the compound of any one of embodiments 1A to 119, 147, 148, and 150, or a pharmaceutically acceptable salt thereof, is wherein Za is -NH-.
154a. In embodiment 154a, the compound of any one of embodiments 1A to 119, and 147- 150, or a pharmaceutically acceptable salt thereof, is wherein Za is -NHC(O)-.
155. In embodiment 155, the compound of any one of embodiments 1A to 119, and 147- 154a, or a pharmaceutically acceptable salt thereof, is wherein ring B is phenylene substituted with Ree and Rff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
156. In embodiment 156, the compound of any one of embodiments 1A to 119, and 147- 154a, or a pharmaceutically acceptable salt thereof, is wherein ring B is cyclylaminylene substituted with Ree and Rff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
157. In embodiment 157, the compound of any one of embodiments 1A to 119, and 147- 154a, or a pharmaceutically acceptable salt thereof, is wherein ring B is 5- or 6-membered monocyclic heteroarylene or a 9- or 10-membered fused bicyclic heteroarylene, wherein each heteroarylene ring contains one to three nitrogen ring atoms and each ring is substituted with Ree and Rff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
158. In embodiment 158, the compound of any one of embodiments 1A to 119, 147-154a and 157, or a pharmaceutically acceptable salt thereof, is wherein ring B is 5- or 6-membered monocyclic heteroarylene containing one or two nitrogen ring atoms substituted with Ree and Rff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
159. In embodiment 159, the compound of any one of embodiments 1A to 119, 147-150 and 157, or a pharmaceutically acceptable salt thereof, is wherein ring B is a 9- or 10-membered fused bicyclic heteroarylene containing one to three nitrogen ring atoms and substituted with Ree and Rff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
160. In embodiment 160, the compound of any one of embodiments 1A to 119, 147-150 and 157, or a pharmaceutically acceptable salt thereof, is wherein ring B is a 9- or 10-membered fused bicyclic heteroarylene containing two nitrogen ring atoms and substituted with Ree and Rff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
161. In embodiment 161, the compound of any one of embodiments 1A to 119 and 147 to 160, or a pharmaceutically acceptable salt thereof, is wherein the E3 ligase ligand of formula (ii) is:
Figure imgf000147_0001
162. In embodiment 162, the compound of any one of embodiments 1A to 119 and 147 to
161, or a pharmaceutically acceptable salt thereof, is wherein the E3 ligase ligand of formula (ii) is:
Figure imgf000148_0001
163. In embodiment 163, the compound of any one of embodiments 1A to 119, and 147 to
161, or a pharmaceutically acceptable salt thereof, is wherein the E3 ligase ligand of formula (ii) is:
Figure imgf000148_0002
Figure imgf000149_0001
164. In embodiment 164, the compound of any one of embodiments 1A to 119, and 147 to 163, or a pharmaceutically acceptable salt thereof, is wherein each Ree and Rff are independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, and haloalkoxy.
165. In embodiment 165, the compound of any one of embodiments 1A to 119, and 147 to 163, or a pharmaceutically acceptable salt thereof, is wherein Ree and Rff are independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, and cyano.
166. In embodiment 166, the compound of any one of embodiments 1A to 119, and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein Ree and Rff are independently selected from hydrogen, methyl, ethyl, isopropyl, cyclopropyl, methoxy, ethoxy, fluoro, chloro, trifluoromethyl, 2,2,2-trifluoroethyl, difluoromethyl, difluoromethoxy, trifluoromethoxy, or cyano.
167. In embodiment 167, the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein Ree and Rff are independently selected from hydrogen and methyl, ethyl, or isopropyl.
168. In embodiment A88, the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein Ree and Rff are independently selected from hydrogen and methoxy.
169. In embodiment 169, the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein Ree and Rff are independently selected from hydrogen, methyl, ethyl, isopropyl, chloro, and fluoro.
170 In embodiment 170, the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein one of Ree and Rff is hydrogen or fluoro and the other of Ree and Rff is selected from hydrogen, trifluoromethyl, 2,2,2-trifluoroethyl, and difluoromethyl.
171. In embodiment 171, the compound of any one of embodiments 1A to 119 147 to
163 and 164, or a pharmaceutically acceptable salt thereof, is wherein Ree and Rff are independently selected from hydrogen, difluoromethoxy, and trifluoromethoxy.
172. In embodiment 172, the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein Ree and Rff are independently selected from hydrogen, chloro, fluoro, and trifluoromethyl.
173. In embodiment A93, the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein Ree and Rff are independently hydrogen.
174. In embodiment 174, the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein Ree and Rff are independently chloro.
175. In embodiment 175, the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein Ree and Rff are independently fluoro.
176. In embodiment 176, the compound of any one of embodiments 1A to 119 and 147 to 165, or a pharmaceutically acceptable salt thereof, is wherein Ree and Rff are independently trifluoromethyl or 2,2,2-trifluoroethyl.
177. In embodiment 177, the compound of any one of embodiments 1 A to 119, or a pharmaceutically acceptable salt thereof, is wherein the Degron is an E3 ligase ligand of formula (iii), (iv), (v), (vi), or (vii).
178. In embodiment 178, the compound of any one of embodiments 1A to 119 and 177, or a pharmaceutically acceptable salt thereof, is wherein the Degron is an E3 ligase ligand of formula
(iii).
179. In embodiment 179, the compound of any one of embodiments 1A to 119 and 177, or a pharmaceutically acceptable salt thereof, is wherein the Degron is an E3 ligase ligand of formula
(iv).
180. In embodiment 180, the compound of any one of embodiments 1A to 119 and 177, or a pharmaceutically acceptable salt thereof, is wherein the Degron is an E3 ligase ligand of formula
(v). 181. In embodiment 181, the compound of any one of embodiments 1A to 119 and 177, or a pharmaceutically acceptable salt thereof, is wherein the Degron is an E3 ligase ligand of formula
(vi).
181a. In embodiment 181a, the compound of any one of embodiments 1A to 119 and 177, or a pharmaceutically acceptable salt thereof, is wherein the Degron is an E3 ligase ligand of formula
(vii).
181b. In embodiment 181b, the compound of any one of embodiments 1A to 119, or a pharmaceutically acceptable salt thereof, is wherein the Degron is an E3 ligase ligand of formula
(viii).
181c. In embodiment 181c, the compound of any one of embodiments 1A to 119, or a pharmaceutically acceptable salt thereof, is wherein the Degron is an E3 ligase ligand of formula (ix).
182. In embodiment 182, the compound of any one of embodiments 1A to 119, 177 to 180, and 181 c, or a pharmaceutically acceptable salt thereof, is wherein the Degron is an E3 ligase ligand of formula (iii), (iv), (v), or (ix), where R17, R18, and R19 are 1 -fluorocycloprop- 1 -yl and Wa is bond, S, or methylene.
183. In embodiment 183, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein X1, X2, X3, and X4 are independently a bond.
184. In embodiment 184, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein X1, X2, X3, and X4 are independently -alkylene-, preferably methylene.
185. In embodiment 185, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein X1, X2, X3, and X4 are independently -O-.
186. In embodiment 186, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein X1, X2, X3, and X4 are independently -(O- alkylene)-.
187. In embodiment 187, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein X1, X2, X3, and X4 are independently -(alkylene-O)-.
188. In embodiment 188, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein X1, X2, X3, and X4 are independently -(NRgg- alkylene)-. 189. In embodiment 189, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein X1, X2, X3. and X4 are independently -(alkylene - NRhh)-.
190. In embodiment 190, the compound of any one of embodiments 1 to 176, or a pharmaceutically acceptable salt thereof, is wherein X1, X2, X3, and X4 are
Figure imgf000152_0001
191. In embodiment 191, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein X1, X2, X3, and X4 are -NH-.
192. In embodiment 192, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein X1, X2, X3, and X4 are independently -N(alkyl)-.
193. In embodiment 193, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein X1, X2, X3, and X4 are -C(=O)-.
194. In embodiment 194, the compound of any one of embodiments 1 to 176, or a pharmaceutically acceptable salt thereof, is wherein X1, X2, X3, and X4 are independently - NRjJC(=O)-.
195. In embodiment 195, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein X1, X2, X3, and X4 are independently - C(=O)NRkk-.
195a. In embodiment 195a, the compound of any one of embodiments 1A and 2 to 176, or a pharmaceutically acceptable salt thereof, is wherein X1, X2, X3, and X4 are independently alkynylene.
195b. In embodiment 195b, the compound of any one of embodiments 1A and 2 to 176, or a pharmaceutically acceptable salt thereof, is wherein X1, X2, X3, and X4 are independently -(alkylene)- heterocyclylene- or cycloalkylene,.
196. In embodiment 196, the compound of any one of embodiments 1A to 176, 188, 189, 194, and 195, or a pharmaceutically acceptable salt thereof, is wherein Rgg, Rhh, Rjj, and Rkk are independently hydrogen or alkyl.
197. In embodiment 197, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein Z6 is a bond or alkylene.
197a. In embodiment 197a, the compound of any one of embodiments 1A to 197, or a pharmaceutically acceptable salt thereof, is wherein Z1 is -alkylene-O-.
197al. In embodiment 197al, the compound of any one of embodiments 1A to 197, or a pharmaceutically acceptable salt thereof, is wherein Z1 is -methylene-O-. 197a2. In embodiment 197a2, the compound of any one of embodiments 1A to 197, or a pharmaceutically acceptable salt thereof, is wherein Z1 is -O-.
197b. In embodiment 197b, the compound of any one of embodiments 1A to 197a, or a pharmaceutically acceptable salt thereof, is wherein Z2 is -C(O)-.
197c. In embodiment 197c, the compound of any one of embodiments 1A to 197b, or a pharmaceutically acceptable salt thereof, is wherein Z3 is -NR”-, or heterocyclene substituted with Rww and Rxx.
197d. In embodiment 197d, the compound of any one of embodiments 1A to 197c, or a pharmaceutically acceptable salt thereof, is wherein R'™ and Rxx are hydrogen.
197e. In embodiment 197e, the compound of any one of embodiments 1A to 197d, or a pharmaceutically acceptable salt thereof, is wherein R” is hydrogen. 197f. In embodiment 197f, the compound of any one of embodiments 1A to 197e, or a pharmaceutically acceptable salt thereof, is wherein Z4 is a bond, alkylene, -(O-alkylene)c-, or -(alkylene-O)c-.
197g. In embodiment 197g, the compound of any one of embodiments 1A to 197f, or a pharmaceutically acceptable salt thereof, is wherein Z5 is a bond, alkylene, -(O-alkylene)b-, or -(alkylene-O)b-.
197h. In embodiment 197h, the compound of any one of embodiments 1A to 197g, or a pharmaceutically acceptable salt thereof, is wherein Z4-Z5 is a bond.
197i. In embodiment 197i, the compound of any one of embodiments 1A to 197h, or a pharmaceutically acceptable salt thereof, is wherein Z6 is a bond, alkylene, -(O-alkylene)a-, or -(alkylene-O)a-.
197j . In embodiment 197j , the compound of any one of embodiments 1A to 197i, or a pharmaceutically acceptable salt thereof, is wherein a, b, c, and d is independently an integer selected from 1 to 3.
198. In embodiment 198, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is a bond, alkylene, or -alkylene-O-;
Z2 is a bond, -alkylene, -C(O)-, -C(O)NR-, phenylene, monocyclic heteroarylene, or heterocycylene, where each ring is substituted with R'™ and Rxx independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy; Z3 is a bond, alkylene, -NR”-, -(O-alkylene)d-, -(alkylene-O)d-, phenylene, heteroarylene, heterocyclylene, bridged heterocyclylene, or spiro heterocyclylene, where each ring is substituted with Ruu and Rvv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
Z4 is a bond, alkylene, -C(O)NR-, -(O-alkylene)c-, or -(alkylene-O)c-;
Z5 is a bond, alkylene, -C(O)-, -C(O)N(R)-, -NR’(CO)-, -(O-alkylene)b-, -(alkylene-O)b-, - 0(CH2)7-, -O(CH2)8-, or heterocyclylene, where each ring is substituted with Rqq and R" independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
Z6 is a bond, alkylene, -C(O)NR-, -NR’(CO)-, -S(O)2NR-, -NR’S(O)2-, -(O-alkylene)a-, - (alkylene-O)a-, phenylene, monocyclic heteroarylene, or heterocyclylene, where each ring is substituted with R00 and Rpp independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy; and where each alkylene of -Z1-Z2-Z3-Z4-Z5-Z6- is independently one to eight carbon atoms, R, R’ and R” is independently hydrogen or alkyl, each a, b, c, and d is independently an integer selected from 1 to 6, and each alkylene of -Z1-, -Z2-, -Z3-, -Z4-, -Z5- and
-Z6- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano
198. In embodiment 198, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is a bond, alkylene, or -alkylene-O-;
Z2 is a bond, -alkylene, -C(O)-, or -C(O)NR-;
Z3 is a bond, alkylene, -NR”-, -(alkylene-O)d-, phenylene, heteroarylene, or heterocyclylene, where each ring is substituted with Ruu and Rvv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
Z4 is a bond, alkylene, -C(O)NR-, -(O-alkylene)c-, or -(alkylene-O)c-;
Z5 is a bond, alkylene, -C(O)-, or heterocyclylene, where the ring is substituted with Rqq and R" independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
Z6 is a bond or alkylene; and where each alkylene of -Z1-Z2-Z3-Z4-Z5-Z6- is independently one to eight carbon atoms, R, R’ and R” is independently hydrogen or alkyl, each a, b, c, and d is independently an integer selected from 1 to 6, and each alkylene of -Z1-, -Z2-, -Z3-, -Z4-, -Z5- and
-Z6- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
199. In embodiment 199, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -alkylene-O-;
Z2 is -C(O)NR-;
Z3, Z4 and Z5 is a bond; and
Z6 is alkylene; and where each alkylene of -Z1 and -Z6- is independently one to eight carbon atoms, R is hydrogen or alkyl, and each alkylene of -Z1- and -Z6- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
200. In embodiment 200, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -alkylene-O-;
Z2 is -C(O)NR-;
Z3 is -(alkylene-O)d-;
Z4 and Z5 are bond and
Z6 is alkylene; and where each alkylene of -Z1-, -Z3-, and -Z6- is independently one to eight carbon atoms, R is hydrogen or alkyl, d is an integer selected from 1 to 6, and each alkylene of -Z1-, -Z3-, and -Z6- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
201. In embodiment 201, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -alkylene-O-;
Z2 is -C(O)-;
Z3 is heterocyclylene substituted with Ruu and Rvv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
Z4 and Z5 are bond; and
Z6 is alkylene; and where each alkylene of -Z1- and -Z2- is independently one to eight carbon atoms, and each alkylene of -Z1- and -Z2- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
202. In embodiment 202, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -alkylene-O-;
Z2, Z3, Z4, and Z5 are a bond;
Z6 is alkylene; and where each alkylene of -Z1- and -Z6- is independently one to eight carbon atoms, and each alkylene of -Z1- and -Z6- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
203. In embodiment 203, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -alkylene-O-; Z2 is a bond;
Z3 is -(alkylene-O)d-;
Z4 and Z5 is a bond;
Z6 is alkylene; and where each alkylene of -Z1-, -Z2-, and -Z6- is independently one to eight carbon atoms.
204. In embodiment 204, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
Z1, Z2, Z3, Z4 and Z5 are a bond; and
Z6 is alkylene; and where alkylene of -Z6- is one to eight carbon atoms.
205. In embodiment 205, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is alkylene;
Z2 is -C(O)NR-;
Z3, Z4, and Z5 are a bond;
Z6 is alkylene; and where each alkylene of -Z1- and -Z6- is independently one to eight carbon atoms, R is independently hydrogen or alkyl, and each alkylene of -Z1- and -Z6- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
206. In embodiment 206, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is alkylene;
Z2 is a bond;
Z3 is phenylene, heterocyclylene or heteroarylene, each ring substituted with Ruu and R" independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
Z4 and Z5 is a bond;
Z6 is alkylene; and where each alkylene of -Z1- and -Z6- is independently one to eight carbon atoms, and each alkylene of -Z1- and -Z6- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
207. In embodiment 207, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -alkylene-O-;
Z2 is a bond;
Z3 is heterocyclylene substituted with Ruu and Rvv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
Z4 and Z5 is a bond;
Z6 or alkylene; and where each alkylene of -Z1- and -Z6- is independently one to eight carbon atoms, and each alkylene of -Z1- and -Z6- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
208. In embodiment 208, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
Z1, Z2, Z3, Z4 and Z5 is a bond, and
Z6 is alkylene of one to eight carbon atom and the alkylene of -Z6- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
209. In embodiment 209, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein: Z1 is -alkylene-O-;
Z2 is -C(0)-;
Z3 is heterocyclylene substituted with Ruu and Rvv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
Z4 is or -(O-alkylene)c- or -(alkylene-O)c-;
Z5 is a bond;
Z6 is a bond or alkylene; and where each alkylene of -Z1-, -Z4-, and -Z6- is independently one to eight carbon atoms, each c is independently an integer selected from 1 to 6, and each alkylene of -Z1-, -Z4-, and -Z6- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
210. In embodiment 210, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -alkylene-O-;
Z2 is -C(O)-;
Z3 is heterocyclylene substituted with Ruu and Rvv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
Z4 is alkylene;
Z5 is heterocyclylene substituted with Rqq and Rrr independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
Z6 is a bond or alkylene; and where each alkylene of -Z1-, -Z4-, and -Z6- is independently one to eight carbon atoms, and each alkylene of -Z1-, -Z4-, and -Z6- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
210a. In embodiment 210a, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein:
Z' is O;
Z2 is alkylene;
Z3 is phenylene (preferred p-phenylene)
Z4 is monocyclic heteroarylene (preferred 1 ,4-triazolyl, 1,4-imidazolyl, 1,4-pyrazolyl, 1,4- pyrrolyl)
Z5 and Z6 are bond; and where each alkylene of -Z3- is independently one to eight carbon atoms, Ris independently hydrogen or alkyl, and alkylene of -Z2- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano.
211. In embodiment 211, the compound of any one of embodiments 1 A to 210, or a pharmaceutically acceptable salt thereof, is wherein each alkylene of -Z1-, -Z2-, -Z3-, -Z4-, -Z5-, and - Z6- is independently selected from methylene, ethylene, propylene butylene, pentylene, or hexylene, preferably methylene, ethylene, or propylene.
212. In embodiment 212, the compound of any one of embodiments 1 A to 211, or a pharmaceutically acceptable salt thereof, is wherein each heterocyclylene of -Z2-, -Z3-, -Z4-, -Z5-, and -Z6- is independently selected from azetidindiyl, pyrrolidindiyl, piperidindiyl, or piperazindiyl.
213. In embodiment 212, the compound of any one of embodiments 1 to 212, or a pharmaceutically acceptable salt thereof, is wherein each heterocyclylene of -Z2-, -Z3-, -Z4-, -Z5-, and -Z6- is independently selected from 1,3- azetidindiyl, 1,3-pyrrolidindiyl, 1,4-piperidinyl, or 1,4- piperazindiyl.
214. In embodiment 214, the compound of any one of embodiments 1 A to 213, or a pharmaceutically acceptable salt thereof, is wherein each heteroarylene of -Z2-, -Z3-, -Z4-, -Z5-, and - Z6- is independently selected from 2,4, or 3,5-pyridindiyl, 2,4, or 2,5-pyrimidindiyl, or 1,3, 1,4- pyrazoldiyl. 215. In embodiment 215, the compound of any one of embodiments 1 A to 214, or a pharmaceutically acceptable salt thereof, is wherein each alkylene of -Z1-Z2-Z3-Z4-Z5-Z6-, as part of another group (e.g, -(O-alkylene)a, -(alkylene-O)a-, -(alkylene -NR”)-) and when present, is ethylene or propylene.
216. In embodiment 216, the compound of any one of embodiments 1A to 215, or a pharmaceutically acceptable salt thereof, is wherein each alkylene of -Z1-Z2-Z3-Z4-Z5-Z6-, as part of another group (e.g, -(O-alkylene)a, -(alkylene-O)a-, -(alkylene -NR”)-) and when present, is ethylene.
217. In embodiment 217, the compound of any one of embodiments 1 to 216, or a pharmaceutically acceptable salt thereof, is wherein each R, R’ and R” of -Z1-Z2-Z3-Z4-Z5-Z6-, when present, is independently hydrogen or methyl.
218. In embodiment 218, the compound of any one of embodiments 1A to 217, or a pharmaceutically acceptable salt thereof, is wherein each R, R’ and R” of -Z1-Z2-Z3-Z4-Z5-Z6-, when present, is hydrogen.
219. In embodiment 219, the compound of any one of embodiments 1 A to 217, or a pharmaceutically acceptable salt thereof, is wherein each R, R’ and R” of -Z1-Z2-Z3-Z4-Z5-Z6-, when present, is methyl.
220. In embodiment 220, the compound of any one of embodiments 1 A to 219, or a pharmaceutically acceptable salt thereof, is wherein phenylene of -Z1-Z2-Z3-Z4-Z5-Z6-, when present, is independently selected from 1,3-phenylene and 1 ,4-phenylene unless stated otherwise in any of the embodiment above.
221. In embodiment 221, the compound of any one of embodiments 1A to 220, or a pharmaceutically acceptable salt thereof, is wherein heterocyclylene, bridged heterocyclylene, and spiro heterocyclylene, of -Z1-Z2-Z3-Z4-Z5-Z6-, when present, are independently selected from
Figure imgf000162_0001
222. In embodiment 222, the compound of any one of embodiments 1 A to 221, or a pharmaceutically acceptable salt thereof, is wherein L (when the Degron is a group of formula (iii) to (vi)), -X'-L-, -X2-L-, -X3-L- and -X4-L- are independently selected from:
Figure imgf000162_0002
Figure imgf000163_0001
Figure imgf000164_0001
Figure imgf000165_0001
223. In embodiment 223, the compound of any one of embodiments 1A to 222, or a pharmaceutically acceptable salt thereof, is wherein R15 and R16 are hydrogen.
224. In embodiment 224, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein L is where:
Z1 is -alkylene-O; and
Z2 is -C(O)- or -C(O)NR-.
225. In embodiment 225, the compound of any one of embodiments 1A to 196 and 224, or a pharmaceutically acceptable salt thereof, is wherein Z2 is -C(O)- .
226. In embodiment 225, the compound of any one of embodiments 1A to 196 and 224, or a pharmaceutically acceptable salt thereof, is wherein Z2 is -C(O)NR-, preferably -CONH-.
227. In embodiment 227, the compound of any one of embodiments 1A to 182 and 225, or a pharmaceutically acceptable salt thereof, is wherein Z3 is heterocyclylene, bridged heterocyclylene, or spiro heterocyclylene, where each ring is substituted with Ruu and Rvv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, cyanoalkyl, hydroxy, hydroxyalkyl, alkoxyalkyl, amino, alkylamino, and dialkylamino.
228. In embodiment 228, the compound of any one of embodiments 1 to 182 and 225, or a pharmaceutically acceptable salt thereof, is wherein Z3 is heterocyclylene, bridged heterocyclylene, or spiro heterocyclylene, where each ring is substituted with Ruu and Rvv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy amino, alkylamino, and dialkylamino.
229. In embodiment 229, the compound of any one of embodiments 1A to 182 and 226, or a pharmaceutically acceptable salt thereof, is wherein Z3 is alkylene, alkynylene, -(alkylene -NR”)-, - (alkylene-O)a-, cycloalkylene, or spiro cyclolalkylene.
230. In embodiment 230, the compound of any one of embodiments 1A to 182 and 224 to
229, or a pharmaceutically acceptable salt thereof, is wherein Z4 is a bond, alkylene, alkenylene, alkynylene, -NR’(CO)-, -O-, -(O-alkylene)c-, -(alkylene-O)c-, phenylene, or monocyclic heteroarylene, where each ring is substituted with Rss and Rt independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy.
231. In embodiment 231 , the compound of any one of embodiments 1 A to 182 and 224 to
230, or a pharmaceutically acceptable salt thereof, is wherein Z4 is a bond. 231a. In embodiment 231a, the compound of any one of embodiments 1A to 182 and 224 to 230, or a pharmaceutically acceptable salt thereof, is wherein Z4 is alkylene.
23 lb. In embodiment 231b, the compound of any one of embodiments 1 A to 182 and 224 to 230, or a pharmaceutically acceptable salt thereof, is wherein Z4 is alkynylene.
232. In embodiment 232, the compound of any one of embodiments 1A to 182 and 224 to 230, or a pharmaceutically acceptable salt thereof, is wherein Z4 is -NR’(CO)-, -O-, -(O-alkylene)c-, - (alkylene-O)c-, phenylene, or monocyclic heteroarylene, where each ring is substituted with Rss and Rt independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy.
233. In embodiment 233, the compound of any one of embodiments 1A to 182 and 224 to 230, or a pharmaceutically acceptable salt thereof, is wherein Z4 is -O-.
234. In embodiment 234, the compound of any one of embodiments 1A to 182 and 224 to 230, or a pharmaceutically acceptable salt thereof, is wherein Z4 is -(O-alkylene)c.
235. In embodiment 235, the compound of any one of embodiments 1A to 182 and 224 to 230, or a pharmaceutically acceptable salt thereof, is wherein Z4 is -(alkylene-O)c.
236. In embodiment 236, the compound of any one of embodiments 1A to 182 and 224 to 230, or a pharmaceutically acceptable salt thereof, is wherein Z4 is phenylene substituted with Rss and Rt independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy.
237. In embodiment 237, the compound of any one of embodiments 1A to 182 and 224 to 230, or a pharmaceutically acceptable salt thereof, is wherein Z4 is monocyclic heteroarylene substituted with Rss and Rt independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy.
238. In embodiment 238, the compound of any one of embodiments 1A to 182 and 224 to
237, or a pharmaceutically acceptable salt thereof, is wherein Z5 is a bond, alkylene, -SO2-, -NR’SO2-, -C(O)-, -NR’(CO)-, -(O-alkylene)b-, or -(alkylene-O)b-.
239. In embodiment 239, the compound of any one of embodiments 1A to 182 and 224 to
238, or a pharmaceutically acceptable salt thereof, is wherein Z5 is a bond.
240. In embodiment 240, the compound of any one of embodiments 1A to 182 and 224 to 238, or a pharmaceutically acceptable salt thereof, is wherein Z5 is a alkylene.
241. In embodiment 241, the compound of any one of embodiments 1A to 182 and 224 to 238, or a pharmaceutically acceptable salt thereof, is wherein Z5 is a -SO2-. 242. In embodiment 242, the compound of any one of embodiments 1A to 182 and 224 to 238, or a pharmaceutically acceptable salt thereof, is wherein Z5 is a -NR’SO2-.
243. In embodiment 243, the compound of any one of embodiments 1A to 182 and 224 to 238, or a pharmaceutically acceptable salt thereof, is wherein Z5 is -C(O)-.
244. In embodiment 244, the compound of any one of embodiments 1A to 182 and 224 to 238, or a pharmaceutically acceptable salt thereof, is wherein Z5 is a -NR’(CO)-.
245. In embodiment 245, the compound of any one of embodiments 1A to 182 and 224 to 238, or a pharmaceutically acceptable salt thereof, is wherein Z5 is a -(O-alkylene)b-.
246. In embodiment 246, the compound of any one of embodiments 1A to 182 and 224 to 238, or a pharmaceutically acceptable salt thereof, is wherein Z5 is a -(alkylene-O)b.
247. In embodiment 247, the compound of any one of embodiments 1A to 182 and 224 to
246, or a pharmaceutically acceptable salt thereof, is wherein Z6 is a bond, alkylene, -C(O)NR-, - NR’(CO)-, -(alkylene-O)a-, phenylene, heterocyclylene, bicyclic heterocyclylene, spiro heterocyclylene, -O-heterocyclylene-, or -heterocyclylene-C(O)-, where each ring is substituted with R00 and Rpp independently selected from hydrogen, deuterium, alkyl, alkoxy, cyano, hydroxy, carboxy, halo, haloalkyl, and haloalkoxy.
248. In embodiment 248, the compound of any one of embodiments 1A to 182 and 224 to
247, or a pharmaceutically acceptable salt thereof, is wherein Z6 is a bond.
249. In embodiment 249, the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof, is wherein Z6 is alkylene.
250. In embodiment 248, the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof, is wherein Z6 is a -C(O)NR-.
251. In embodiment 251, the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof, is wherein Z6 is -NR’(CO)-.
252. In embodiment 252, the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof, is wherein Z6 is a -(alkylene-O)a-.
253. In embodiment 253, the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof, is wherein Z6 is phenylene substituted with R00 and Rpp independently selected from hydrogen, deuterium, alkyl, alkoxy, cyano, hydroxy, carboxy, halo, haloalkyl, and haloalkoxy. 254. In embodiment 254, the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof, is wherein Z6 is heterocyclylene substituted with R00 and Rpp independently selected from hydrogen, deuterium, alkyl, alkoxy, cyano, hydroxy, carboxy, halo, haloalkyl, and haloalkoxy.
255. In embodiment 255, the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof, is wherein Z6 is bicyclic heterocyclylene substituted with R00 and Rpp independently selected from hydrogen, deuterium, alkyl, alkoxy, cyano, hydroxy, carboxy, halo, haloalkyl, and haloalkoxy.
256. In embodiment 256, the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof, is wherein Z6 is spiro heterocyclylene substituted with R00 and Rpp independently selected from hydrogen, deuterium, alkyl, alkoxy, cyano, hydroxy, carboxy, halo, haloalkyl, and haloalkoxy.
257. In embodiment 257, the compound of any one of embodiments 1A to 182 and 224 to 247, or a pharmaceutically acceptable salt thereof, is wherein Z6 is -O-heterocyclylene- or - heterocyclylene-C(O)-, where each ring is substituted with R00 and Rpp independently selected from hydrogen, deuterium, alkyl, alkoxy, cyano, hydroxy, carboxy, halo, haloalkyl, and haloalkoxy.
258. In embodiment 258, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)NR;
Z3 is alkylene, alkynylene, -(alkylene -NR”)-, or -(alkylene-O)a; and Z4, Z5, Z6, X1, X2, X3, and X4 are a bond.
259. In embodiment 259, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)-;
Z3 is a heterocyclylene; and
Z4, Z5, Z6, X1, X2, X3, and X4 are a bond.
259a. In embodiment 259a, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-; Z2 is -C(0)-;
Z3 is a heterocyclylene; and
Z4 is alkylene, and
Z5, Z6, X1, X2, X3, and X4 are a bond.
260. In embodiment 260, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)NR-;
Z3 is alkylene;
Z6 is -C(O)NR- or heterocyclylene; and
Z4, Z5, X1, X2, X3, and X4 are a bond.
260a. In embodiment 260a, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)NR-;
Z3 is alkylene; and
Z4, Z5, Z6, X1, X2, X3, and X4 are a bond.
261. In embodiment 261, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)-;
Z3 is heterocyclylene or spiro heterocyclylene;
Z6 is alkylene, -C(O)NR-, -(alkylene-O)a-, or phenylene; and
Z4, Z5, X1, X2, X3, and X4 are a bond.
262. In embodiment 262, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)-;
Z3 is heterocyclylene;
Z4 is alkynylene, phenylene, or -(alkylene-O)c; and
Z5, Z6, X1, X2, X3, and X4 are a bond. 263. In embodiment 263, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is a -C(O)NR-;
Z3 is a -(alkylene-O)a- or cycloalkylene;
Z4 is alkylene or -O-; preferably -O-;
Z6 is -C(O)NR- or - heterocyclylene; and
Z5, X1, X2, X3, and X4 are a bond.
264. In embodiment 264, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O);
Z3 is a heterocyclylene;
Z4 is alkylene, -(O-alkylene)c, -O-, or heteroarylene;
Z6 is -C(O)NR-, heterocyclylene, phenylene, -O-heterocyclylene, or -heterocyclylene-CO-; and
Z5, X1, X2, X3, and X4 are a bond.
265. In embodiment 265, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is a -C(O)NR-;
Z3 is alkylene, -(alkylene-O)i-3, cycloalkylene, or spiro cycloalkylene;
Z5 is alkylene, NHCO, or -CO-;
Z6 is -C(O)NR- or heterocyclylene; and
Z4, X1, X2, X3, and X4 are a bond.
266. In embodiment 266, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is a -C(O); Z3 is a heterocyclylene, spiro heterocyclylene, bridged heterocycylene, or heterocyclylene substituted with cyano, methoxy, methoxymethyl, hydroxymethyl, cyanomethyl, or 1 or 2 methyl;
Z5 is CO or -(alkylene-O)1-3-;
Z6 is heterocyclylene, bicyclic heterocycylene, spiro heterocycylene, -O- heterocyclylene, or heterocyclylene substituted with carboxy, cyano, hydroxy, methyl, methoxy, or fluoro; and
Z4, X1, X2, X3, and X4 are a bond.
267. In embodiment 267, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)-;
Z3 is heterocyclylene;
Z4 is alkylene or -O-alkylene-;
Z5 is CO;
Z6 is heterocyclylene; and
X1 , X2, X3, and X4 are a bond.
268. In embodiment 268, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)NR-;
Z3 is -(alkylene-O) 1-3;
X1, X2, X3, and X4 are -alkyl ene-NH-; and
Z4, Z5, and Z6 are bond.
269. In embodiment 269, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)-;
Z3 is a heterocyclylene or spiro heterocyclylene;
X1, X2, X3, and X4 are -alkyl ene-NH-; and
Z4, Z5, and Z6 are a bond. 270. In embodiment 270, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)NR-;
Z3 is heterocyclylene;
Z5 is -C(O)-;
X1, X2, X3, and X4 are cycloalkylene; and
Z4 and Z6 are a bond.
271. In embodiment 271, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is a -C(O)-;
Z3 is a heterocyclylene;
Z5 is NHCO, -NHSO2-, alkylene, -(O-alkylene)-, or C(O)-;
X1 , X2, X3, and X4 are -NH- or cycloalkylene; and
Z4 and Z6 are a bond.
272. In embodiment 272, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)NR-;
Z3 is a heterocyclylene;
Z5 is CO or SO2;
Z6 is phenylene;
X1, X2, X3, and X4 are -alkyl ene-heterocycylene-; and
Z4 is a bond.
273. In embodiment 273, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O);
Z3 is heterocyclylene;
Z5 is CO; 76 is heterocyclylene;
X1, X2, X3, and X4 are -NH- or O and Z4 is a bond.
274. In embodiment 274, the compound of any one of embodiments 1A to 176, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is a -C(O);
Z3 is heterocyclylene;
Z4 is O;
Z6 is heterocyclylene;
X1, X2, X3, and X4 are alkynylene -; and Z5 is a bond.
275. In embodiment 275, the compound of any one of embodiments 1A to 119 and 177 to 181b, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)- or -CONH;
Z3 is cycloalkylene, heterocyclylene, spiro heterocycylene, or alkylene; and Z3, Z4, Z5, X1, X2, X3, and X4 are bond.
276. In embodiment 276, the compound of any one of embodiments 1A to 119 and 177 to 181b, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)- or -CONH;
Z3 is (alkylene-O)i-3, heterocyclylene, spiro heterocycylene, bridged heterocyclylene, or alkylene;
Z4 is heteroarylene, alkylene, phenylene, or NHCO; and
Z5, Z6, X1, X2, X3, and X4 are a bond.
277. In embodiment 277, the compound of any one of embodiments 1A to 119 and 177 to 181b, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)- or -CONH;
Z3 is heterocyclylene or alkylene; Z6 is -heterocyclylene-CO-; and
Z5, Z5, X1, X2, X3, and X4 are a bond.
278. In embodiment 278, the compound of any one of embodiments 1A to 119 and 177 to 181b, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)-;
Z3 is heterocyclylene;
Z5 is alkylene or CO;
Z6 is NHCO, alkylene, phenylene, or -heterocyclylene-CO-; and
Z4, X1, X2, X3, and X4 are a bond.
279. In embodiment 279, the compound of any one of embodiments 1A to 119 and 177 to 181b, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)NH-;
Z3 is alkylene;
Z4 is phenylene;
Z5 is alkylene; -; and
Z6, X1, X2, X3, and X4 are a bond.
280. In embodiment 280, the compound of any one of embodiments 1A to 119 and 177 to 181b, or a pharmaceutically acceptable salt thereof, is wherein:
Z1 is -(alkylene-O)-;
Z2 is -C(O)- or -CONH-;
Z3 is heterocyclylene or alkylene;
Z4 is alkylene;
Z6 is heterocyclylene-CO or -NHCO—; and
Z5, X1, X2, X3, and X4 are a bond.
281. In embodiment 281, the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof, is wherein each alkylene of -Z1-, -Z3-, -Z4-, -Z5-, and -Z6-, when present, is independently selected from methylene, ethylene, propylene butylene, pentylene, or hexylene, preferably methylene, ethylene, or propylene. 282. In embodiment 282, the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof, is wherein each heterocyclylene of -Z3- and -Z6- is independently selected from azetidindiyl, pyrrolidindiyl, piperidindiyl, or piperazindiyl.
283. In embodiment 283, the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof, is wherein each heterocyclylene of -Z3- and -Z6- is independently selected from 1,3- azetidindiyl, 1,3 -pyrrolidindiyl, 1 ,4-piperidinyl, or 1,4- piperazindiyl.
284. In embodiment 284, the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof, is wherein each alkylene of -Z1-Z2-Z3-Z4-Z5-Z6-, as part of another group (e.g, -(O-alkylene)a, -(alkylene-O)a-, -(alkylene -NR”)-) and when present, is ethylene or propylene.
285. In embodiment 285, the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof, is wherein each alkylene of -Z1-Z2-Z3-Z4-Z5-Z6-, as part of another group (e.g, -(O-alkylene)a, -(alkylene-O)a-, -(alkylene -NR”)-) and when present, is ethylene.
286. In embodiment 286, the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof, is wherein each R, R’ and R” of -Z1-Z2-Z3-Z4-Z5-Z6-, when present, is independently hydrogen or methyl.
287. In embodiment 287, the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof, is wherein each R, R’ and R” of -Z1-Z2-Z3-Z4-Z5-Z6-, when present, is hydrogen.
288. In embodiment 288, the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof, is wherein each R, R’ and R” of -Z1-Z2-Z3-Z4-Z5-Z6-, when present, is methyl.
289. In embodiment 289, the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof, is wherein phenylene of -Z1-Z2-Z3-Z4-Z5-Z6-, when present, is independently selected from 1,3-phenylene and 1 ,4-phenylene unless stated otherwise in any of the embodiments above.
290. In embodiment 290, the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof, is wherein heterocyclylene, bridged heterocyclylene, and spiro heterocyclylene, of -Z1-Z2-Z3-Z4-Z5-Z6-, when present, are independently selected from
Figure imgf000177_0001
291. In embodiment 291, the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof, is wherein alkenylene of -Z1-Z2-Z3-Z4-Z5-Z6-, when present, are independently selected from ethenylene, propenylene butenylene, pentyenylene, or hexenylene.
292. In embodiment 292, the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof, is wherein alkynylene of -Z1-Z2-Z3-Z4-Z5-Z6-, when present, are independently selected from ethynylene, propynylene butynylene, pentynylene, or hexynylene.
293. In embodiment 293, the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof, is wherein cycloalkylene of -Z1-Z2-Z3-Z4-Z5-Z6-, when present, are independently selected from cycloprolylene, cyclobutylene, cyclopentylene, cyclohexylene, and cycloheptylene.
294. In embodiment 294, the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof, is wherein cycloalkylene of -Z1-Z2-Z3-Z4-Z5-Z6-, when present are cyclohexylene.
295. In embodiment 295, the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof, is wherein spiro cycloalkylene of -Z1-Z2-Z3-Z4-Z5-Z6-, when present, are independently selected from spiro[5.5]undecanylene, spiro[4.5]decanylene, spiro[3.5]nonanylene, spiro[4.4]nonanylene, and spiro[3.4]octanylene. 296. In embodiment 296, the compound of any one of embodiments 224 to 280, or a pharmaceutically acceptable salt thereof, is wherein spiro cycloalkylene of -Z1-Z2-Z3-Z4-Z5-Z6-, when present, are spiro[5.5]undecanylene.
297. In embodiment 297, the compound of any one of embodiments 1 A to 119, or a pharmaceutically acceptable salt thereof, is wherein L-Degron is:
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
298. In embodiment 298, the compound of any one of embodiments 1A to 119, or a pharmaceutically acceptable salt thereof, is wherein L-Degron is:
Figure imgf000181_0002
Figure imgf000182_0001
299. In embodiment 299, the compound of any one of embodiments 1A to 196, or a pharmaceutically acceptable salt thereof, is wherein L is:
Figure imgf000182_0002
Figure imgf000183_0001
Figure imgf000184_0001
300. In embodiment 300, provided is a pharmaceutical composition comprising a compound of any one of embodiments 1A to 299, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
General Synthetic Scheme
Compounds Formula (IA) or (I) can be made by the methods depicted in the reaction schemes shown below.
The starting materials and reagents used in preparing these compounds are either available from commercial suppliers such as Aldrich Chemical Co., (Milwaukee, Wis.), Bachem (Torrance, Calif.), or Sigma (St. Louis, Mo.) or are prepared by methods known to those skilled in the art following procedures set forth in references such as Fieser and Fieser’s Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd’s Chemistry of Carbon Compounds, Volumes 1-5 and Suppiementals (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1- 40 (John Wiley and Sons, 1991), March’s Advanced Organic Chemistry, (John Wiley and Sons, 4th Edition) and Larock’s Comprehensive Organic Transformations (VCH Publishers Inc., 1989). These schemes are merely illustrative of some methods by which the compounds Formula (IA) or (I) can be synthesized, and various modifications to these schemes can be made and will be suggested to one skilled in the art reading this disclosure. The starting materials and the intermediates, and the final products of the reaction may be isolated and purified if desired using conventional techniques, including but not limited to filtration, distillation, crystallization, chromatography and the like. Such materials may be characterized using conventional means, including physical constants and spectral data. Unless specified to the contrary, the reactions described herein take place at atmospheric pressure over a temperature range from about -78 °C to about 150 °C, such as from about 0 °C to about 125 °C and further such as at about room (or ambient) temperature, e.g., about 20 °C. Compounds of Formula (IA) or (I) can be prepared by methods known in the art. For example, compound of Formula (IA) or (I) where R4 is -O-R6 and other groups are as defined in the Summary can be prepared as illustrated and described in Scheme 1 below.
Scheme 1
Figure imgf000185_0001
Chlorination of a compound of formula 1-a where Xa is a halogen, and other groups as defined in the Summary with a suitable chlorination reagent such as POCh optionally in presence of a base such as DIPEA provides a 2,4-dichloro compound of formula 1-b. Compounds of formula l~a is either commercially available or they can be prepared by method well known in the art e.g., see Methods 1 and 2 below.
Method 1
Compounds of Formula 1-a where Xa is halogen, U is CH, V is N, W is CH, R2 and R3 are as defined in the Summary (or any embodiments thereof) can be prepared as illustrated and described below.
Figure imgf000186_0001
Iodination of a compound of formula 1 where Xa is a halo and R2 and R3 are as defined in the Summary, with NIS and a suitable acid such as TsOH provides a compound of formula 2. The iodine in 2 can be converted to ethyl carboxylate under carbonylation condition including Pd catalyst such as Pd(PPh3)4 in carbon monoxide atmosphere and ethanol solvent to provide a compound of formula 3. Compound 3 can react with triphosgene to provide tri chloroacetamido compound of formula 4, which upon treatment with ammonia in an organic solvent such as methanol, undergoes cyclization to provide compound of formula 1-a. Compounds of formula 1 are either commercially available or can be made by methods known in the art. For example, 2-chloro-3-fluoropyridin-4-amine and 2- chloropyridin-4-amine are commercially available.
Method 2:
Compounds of Formula 1-a where Xa is halogen, U, V and W are CH, R2 and R3 are as defined in the Summary (or any embodiments thereof) can be by reacting a compound of formula
Figure imgf000186_0002
urea at elevated temperature. Compounds of formula 5 are either commercially available or can be made by methods known in the art. For example, 2-amino-4-bromo-5 -chi oro-3 - fluorobenzoic acid, 2-amino-4-bromo-3 -fluorobenzoic acid and 2-amino-4-bromobenzoic acid are commercially available.
Treatment of compound 1-b with an amine of formula (a”) where m, n, R1, R1a, R1b, and Rlc are as defined in the Summary or a precursor group thereof and PG is a suitable amino protecting group such as Boc, CBz, and the like, in the presence of a base such as DEA or DBU and the like, provides a 2-chloro compound of formula 1-c. Displacement of chloro group at C-2 position in compound 1-c with an alcohol of formula 1-f where groups are as defined in Summary provides a compound of formula 1-d.
Amines of formula (a”) are either commercially available or can be made by methods known in the art. For example, benzyl 2-(cyanomethyl)piperazine-l -carboxylate, tert-butyl 2- (cyanomethyl)piperazine- 1 -carboxylate, benzyl 2,5-dimethylpiperazine-l -carboxylate, tert-butyl 2- methylpiperazine- 1 -carboxylate, tert-butyl piperazine- 1 -carboxylate, benzyl piperazine- 1 -carboxylate are commercially available. Others can be prepared by methods well known in the art.
Reaction of a compound of formula 1-d with a suitable organometallic reagent of formula R5- M where R5 is cycloalkyl, aryl or heteroaryl as defined in the Summary and M is boronic acid, boronic ester, or stannane, under Suzuki, Negeshi, and Stille reaction conditions provides compound of formula 1-e.
Removal of amino protecting group PG in 1-e under standard reaction condition provides a compound of Formula (IA) or (I).
Alternatively, compounds of Formula (IA) or (I) where R4 is -O-R6, and other groups are as defined in the Summary can be prepared as illustrated and described in Scheme 2 below.
Scheme 2
Figure imgf000187_0001
Coupling reaction between compound 1-a and a suitable organometallic reagent of formula
R5-M where R5 is cycloalkyl, aryl or heteroaryl as defined in the Summary or a precursor group thereof and M is boronic acid, boronic ester, or stannane, under Suzuki, Negeshi, and Stille reaction conditions respectively, to provide a compound of formula 2-a. Chlorination of a compound of formula 2-a with a suitable chlorination reagent such as POCl3 optionally in presence of a base such as DIPEA provides compound of formula 2-b. Compound 2-b is converted to a compound of Formula (IA) or (I) as described in Scheme 1 above.
Figure imgf000188_0001
NR" or heterocyclylene can be prepared by a precursor group of formula as
Figure imgf000188_0002
illustrated and described in Scheme 3 below.
Figure imgf000188_0003
Treatment of a compound of formula 3-a (prepared by example 1 or 2) with a coulping agent such as CDI, and the like, treatment of the resulting intermediate with an amine of Z3-Z4-Z5-Z6- Degron precursor, provides a compound of formula 3-b which can the converted to compound of Formula (IA) or (I) as described above.
Utility The present disclosure provides treatment of cancer mediated by K-ras, in particular with K- ras G12D mutants. In some embodiments, the cancer is pancreatic cancer, colorectal cancer, lung cancer, gall bladder cancer, thyroid cancer, and bile duct cancer. In certain embodiments the lung cancer is a non- small cell lung carcinoma (NSCLC), for example adenocarcinoma, squamous-cell lung carcinoma or large-cell lung carcinoma. In some embodiments, the lung cancer is a small cell lung carcinoma. Other lung cancers treatable with the disclosed compounds include, but are not limited to, glandular tumors, carcinoid tumors and undifferentiated carcinomas.
K-ras G12D mutations are observed in hematological malignancies that affect blood, bone marrow, and/or lymph nodes. As such the compounds of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof can be used for the treatment of acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), chronic myelogenous leukemia (CML), acute monocytic leukemia (AMoL) and/ or other leukemias, lymphomas such as all subtypes of Hodgkins lymphoma or non-Hodgkins lymphoma, plasma cell malignancies such as multiple myeloma, mantle cell lymphoma, and Waldenstrom’s macroglubunemia.
The compounds of Formula (IA) or (I), or a pharmaceutically acceptable salt thereof can be used for the treatment of a hyperproliferative disorder or metastasis in human who suffers from a cancer such as acute myeloid leukemia, cancer in adolescents, adrenocortical carcinoma childhood, AIDS related cancers (e.g. Lymphoma and Kaposi's Sarcoma), anal cancer, appendix cancer, astrocytomas, atypical teratoid, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumors, Burkitt lymphoma, carcinoid tumor, atypical teratoid, embryonal tumors, germ cell tumor, primary lymphoma, cervical cancer, childhood cancers, chordoma, cardiac tumors, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myleoproliferative disorders, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, extrahepatic ductal carcinoma in situ (DCIS), embryonal tumors, CNS cancer, endometrial cancer, ependymoma, esophageal cancer, esthesioneuroblastoma, ewing sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor, eye cancer, fibrous histiocytoma of bone, gall bladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumors (GIST), germ cell tumor, gestational trophoblastic tumor, hairy cell leukemia, head and neck cancer, heart cancer, liver cancer, Hodgkin lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumors, pancreatic neuroendocrine tumors, kidney cancer, laryngeal cancer, lip and oral cavity cancer, liver cancer, lobular carcinoma in situ (LCIS), lung cancer, lymphoma, metastatic squamous neck cancer with occult primary, midline tract carcinoma, mouth cancer, multiple endocrine neoplasia syndromes, multiple myeloma/plasma cell neoplasm, mycosis fimgoides, myelodysplastic syndromes, myelodysplastic/myeloproliferative neoplasms, multiple myeloma, merkel cell carcinoma, malignant mesothelioma, malignant fibrous histiocytoma of bone and osteosarcoma, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-hodgkin lymphoma, non-small cell lung cancer (NSCLC), oral cancer, lip and oral cavity cancer, oropharyngeal cancer, ovarian cancer, pancreatic cancer, papillomatosis, paraganglioma, paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pleuropulmonary blastoma, primary central nervous system (CNS) lymphoma, prostate cancer, rectal cancer, transitional cell cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, skin cancer, stomach (gastric) cancer, small cell lung cancer, small intestine cancer, soft tissue sarcoma, T-Cell lymphoma, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer, transitional cell cancer of the renal pelvis and ureter, trophoblastic tumor, unusual cancers of childhood, urethral cancer, uterine sarcoma, vaginal cancer, vulvar cancer, or viral-induced cancer. The compounds of Formula (IA) or (I), or a pharmaceutically acceptable salt thereof can also be used for the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
Testing
The K-Ras G12D activity of the compounds of Formula (IA) or (I), or a pharmaceutically acceptable salt thereof can be tested using the in vitro assay described in Biological Examples 1 below.
Pharmaceutical Compositions
In general, the compounds Formula (IA) or (I) (unless stated otherwise, reference to compound/compounds of Formula (IA) or (I) herein includes any embodiments thereof described herein or a pharmaceutically acceptable salt thereof) will be administered in a therapeutically effective amount by any of the accepted modes of administration for agents that serve similar utilities. Therapeutically effective amounts of compounds Formula (IA) or (I) may range from about 0.01 to about 500 mg per kg patient body weight per day, which can be administered in single or multiple doses. A suitable dosage level may be from about 0.1 to about 250 mg/kg per day; about 0.5 to about 100 mg/kg per day. A suitable dosage level may be about 0.01 to about 250 mg/kg per day, about 0.05 to about 100 mg/kg per day, or about 0.1 to about 50 mg/kg per day. Within this range the dosage can be about 0.05 to about 0.5, about 0.5 to about 5 or about 5 to about 50 mg/kg per day. For oral administration, the compositions can be provided in the form of tablets containing about 1.0 to about 1000 milligrams of the active ingredient, particularly about 1, 5, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, 500, 600, 750, 800, 900, and 1000 milligrams of the active ingredient. The actual amount of the compound Formula (IA) or (I), i.e., the active ingredient, will depend upon numerous factors such as the severity of the disease to be treated, the age and relative health of the patient, the potency of the compound being utilized, the route and form of administration, and other factors.
In general, compounds Formula (IA) or (I) will be administered as pharmaceutical compositions by any one of the following routes: oral, systemic (e.g., transdermal, intranasal or by suppository), or parenteral (e.g., intramuscular, intravenous or subcutaneous) administration. The preferred manner of administration is oral using a convenient daily dosage regimen, which can be adjusted according to the degree of affliction. Compositions can take the form of tablets, pills, capsules, semisolids, powders, sustained release formulations, solutions, suspensions, elixirs, aerosols, or any other appropriate compositions.
The choice of formulation depends on various factors such as the mode of drug administration (e.g., for oral administration, formulations in the form of tablets, pills or capsules, including enteric coated or delayed release tablets, pills or capsules are preferred) and the bioavailability of the drug substance.
The compositions are comprised of in general, a compound of Formula (IA) or (I) in combination with at least one pharmaceutically acceptable excipient. Acceptable excipients are generally non-toxic, aid administration, and do not adversely affect the therapeutic benefit of the compound of Formula (IA) or (I). Such excipient may be any solid, liquid, semi-solid or, in the case of an aerosol composition, gaseous excipient that is generally available to one of skill in the art.
Solid pharmaceutical excipients include starch, cellulose, talc, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, magnesium stearate, sodium stearate, glycerol monostearate, sodium chloride, dried skim milk and the like. Liquid and semisolid excipients may be selected from glycerol, propylene glycol, water, ethanol and various oils, including those of petroleum, animal, vegetable or synthetic origin, e.g., peanut oil, soybean oil, mineral oil, sesame oil, etc. Preferred liquid carriers, particularly for injectable solutions, include water, saline, aqueous dextrose, and glycols.
The compounds of Formula (IA) or (I) may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in powder form or in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, saline or sterile pyrogen-free water, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
Formulations for parenteral administration include aqueous and non-aqueous (oily) sterile injection solutions of the active compounds which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
In addition to the formulations described previously, the compounds of Formula (IA) or (I) may also be formulated as a depot preparation. Such long -acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
For buccal or sublingual administration, the compositions may take the form of tablets, lozenges, pastilles, or gels formulated in conventional manner. Such compositions may comprise the active ingredient in a flavored basis such as sucrose and acacia or tragacanth. The compounds of Formula (IA) or (I) may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter, polyethylene glycol, or other glycerides.
Certain compounds of Formula (IA) or (I) may be administered topically, that is by non- systemic administration. This includes the application of a compound of Formula (IA) or (I) externally to the epidermis or the buccal cavity and the instillation of such a compound into the ear, eye and nose, such that the compound does not significantly enter the blood stream. In contrast, systemic administration refers to oral, intravenous, intraperitoneal and intramuscular administration.
Formulations suitable for topical administration include liquid or semi-liquid preparations suitable for penetration through the skin to the site of inflammation such as gels, liniments, lotions, creams, ointments or pastes, and drops suitable for administration to the eye, ear or nose. The active ingredient for topical administration may comprise, for example, from 0.001% to 10% w/w (by weight) of the formulation. In certain embodiments, the active ingredient may comprise as much as 10% w/w. In other embodiments, it may comprise less than 5% w/w. In certain embodiments, the active ingredient may comprise from 2% w/w to 5% w/w. In other embodiments, it may comprise from 0.1% to 1% w/w of the formulation.
For administration by inhalation, compounds of Formula (IA) or (I) may be conveniently delivered from an insufflator, nebulizer pressurized packs or other convenient means of delivering an aerosol spray. Pressurized packs may comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Alternatively, for administration by inhalation or insufflation, the compounds of Formula (IA) or (I) may take the form of a dry powder composition, for example a powder mix of the compound and a suitable powder base such as lactose or starch. The powder composition may be presented in unit dosage form, in for example, capsules, cartridges, gelatin or blister packs from which the powder may be administered with the aid of an inhalator or insufflator. Other suitable pharmaceutical excipients and their formulations are described in Remington’s Pharmaceutical Sciences, edited by E. W. Martin (Mack Publishing Company, 20th ed., 2000).
The level of the compound of Formula (IA) or (I) in a formulation can vary within the full range employed by those skilled in the art. Typically, the formulation will contain, on a weight percent (wt. %) basis, from about 0.01-99.99 wt. % of a compound of Formula (IA) or (I) based on the total formulation, with the balance being one or more suitable pharmaceutical excipients. For example, the compound is present at a level of about 1-80 wt. %.
Combinations and Combination Therapies
The compounds of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof may be used in combination with one or more other drugs in the treatment of diseases or conditions for which compounds of Formula (IA) or (I) or the other drugs may have utility. Such other drug(s) may be administered, by a route and in an amount commonly used therefore, contemporaneously or sequentially with a compound of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof. When a compound of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof is used contemporaneously with one or more other drugs, a pharmaceutical composition in unit dosage form containing such other drugs and the compound of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof can be used. Accordingly, the pharmaceutical compositions of the present disclosure also include those that contain one or more other drugs, in addition to a compound of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof. The combination therapy may also include therapies in which the compound of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof and one or more other drugs are administered on different overlapping schedules. It is also contemplated that when used in combination with one or more other active ingredients, the compounds of Formula (IA) or (I) and the other active ingredients may be used in lower doses than when each is used singly. The weight ratio of the compound of this disclosure to the second active ingredient may be varied and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used.
Where the subject in need is suffering from or at risk of suffering from cancer, the patient can be treated with a compound of Formula (IA) or (I) or a pharmaceutically acceptable salt thereof in any combination with one or more other anti-cancer agents including but not limited to:
MAP kinase pathway (RAS/RAF/MEK/ERK) inhibitors including but not limited to: Vemurafanib (PLX4032, CAS No. 918504-65-1), Dabrafenib (CAS No. 1195765-45-7), Encorafenib (LGX818 CAS No. 1269440-17-6), TQ-B3233, XL-518 (Cas No. 1029872- 29-4, available from ACC Corp); trametinib (CAS No. 871700-17-3), selumetinib (AZD6244 CAS No. 606143-52-6), TQ- B3234, PD184352 (CAS No. 212631-79-3), PD325901 (CAS No. 391210-10-9), TAK-733 (CAS No. 1035555-63-5), pimasertinib (CAS No. 1236699-92-5), binimetinib (CAS No. 606143-89-9), refametinib (CAS No. 923032-37-5), cobimetinib (GDC- 0973 CAS No. 934660-93-2), AZD8330 (CAS No. 869357-68-6), BVD-523 (CAS No. 869886-67-9), LTT462 (CAS No. 869886-67-9), , AMG510 (CAS No. 2296729-00-3), ARS853 (CAS No. 1629268-00-3), and any RAS inhibitors disclosed in patents WO2016049565, W02016164675,W02016168540, WO2017015562, WO2017058728, WO2017058768, WO2017058792, W02017058805,W02017058807, W02017058902, WO2017058915, W02017070256, WO2017087528, W02017100546, WO20 17172979, W02017201161, WO2018064510, WO2018068017, and WO2018119183;
SHP2 inhibitors including but not limited to: SHP099 (CAS No. 2200214-93-1), TNO155 (CAS No. 1801765-04-7), RMC4630, JAB-3312, JAB-3068 and ERAS-601;
S0S1 inhibitors including but not limited to BI 1701963 and BAY-293;
CSF1R inhibitors (PLX3397, LY3022855,) and CSF1R antibodies (IMC-054, RG7155);
TGF beta receptor kinase inhibitor such as LY2157299;
BTK inhibitor such as ibrutinib; BCR-ABL inhibitors: Imatinib (CAS No. 152459-95-5); Inilotinib hydrochloride; Nilotinib (CAS No. 923288-95-3); Dasatinib (BMS-345825 CAS No. 302962-49-8); Bosutinib (SKI-606 CAS No. 380843-75-4); Ponatinib (AP24534 CAS No. 943319- 70-8); Bafetinib (INNO406 CAS No. 859212-16-1); Danusertib (PHA-739358 CAS No. 827318-97- 8), AT9283 (CAS No. 896466-04-9); Saracatinib (AZD0530 CAS No. 379231-04-6); and PF- 03814735 (CAS 942487-16-3);
ALK inhibitors: PF-2341066 (XALKOPJ® ; crizotinib); 5-chloro-N4-(2- (isopropyl- sulfonyl)phenyl)-N2-(2-methoxy-4-(4-(4-methylpiperazin-l-yl)piperidin-l-yl)phenyl)pyrimidine-2,4- diamine; GSK1838705A (CAS No. 1116235-97-2); CH5424802 (CAS No. 1256580-46-7); Ceritinib (ZYKADIA CAS No. 1032900-25-6); TQ-B3139, and TQ-B3101;
PI3 K inhibitors : 4- [2-( 1 H-indazol-4-yl)-6- [ [4-(methylsulfonyl)-piperazin-l- yl]methyl]thieno[3,2-d]pyrimidin-4-yl]morpholine (also known as GDC 0941 and described in PCT Publication Nos. WO 09/036082 and WO 09/055730), BEZ235 or NVP-BEZ235 (CAS No. 915019- 65-7), disclosed in PCT Publication No. WO 06/122806);
Vascular Endothelial Growth Factor (VEGF) receptor inhibitors: Bevacizumab (sold under the trademark Avastin® by Genentech/Roche), axitinib, (N-methyl-2-[[3-[(E)-2- pyridin-2- ylethenyl]- lH-indazol-6-yl]sulfanyl]benzamide, also known as AG013736, and described in PCT Publication No. WO 01/002369), Brivanib Alaninate ((S)-((R)-l-(4-(4-fluoro-2-methyl-4H-indol- 5-yloxy)-5- methylpyrrolo[2,l-f][l,2,4]triazin-6-yloxy)propan-2-yl)2-aminopropanoate, also known as BMS- 582664), motesanib (N-(2,3-dihydro-3,3- dimethyl-lH-indol-6-yl)-2-[(4- pyridinylmethyl)amino]-3- pyridinecarboxamide, and described in PCT Publication No. WO 02/066470), pasireotide (also known as SOM230, and described in PCT Publication No. WO 02/010192), sorafenib (sold under the tradename Nexavar®, CAS No. 284461-73-0); or AL-2846;
MET inhibitor such as foretinib (CAS No. 849217-64-7), cabozantinib (CAS No. 1140909- 48-3), capmatinib (CAS No. 1029712-80-8), tepotinib (CAS No. 1100598-32-0), savolitinib (CAS No. 1313725-88-0, or crizotinib (CAS No. 877399-52-5);
FLT3 inhibitors - sunitinib malate (CAS No. 341031-54-7, sold under the tradename Sutent® by Pfizer); PKC412 (CAS No. 120685-11-2, midostaurin); tandutinib (CAS No. 387867-13-2), sorafenib (CAS No. 284461-73-0), lestaurtinib (CAS No.: 111358-88-4), KW-2449 (CAS No. 1000669-72-6), quizartinib (AC220, CAS No. 950769-58-1), or crenolanib (CAS No. 670220-88-9);
Epidermal growth factor receptor (EGFR) inhibitors: Gefitnib (sold under the tradename Iressa®), N- [4- [(3 -chloro-4-fluorophenyl)amino] -7- [ [(3 S)-tetrahydro-3 - furanyl] oxy] -6- quinazolinyl]-4(dimethylamino)-2-butenamide, sold under the tradename Tovok® by Boehringer Ingelheim), cetuximab (sold under the tradename Erbitux® by Bristol-Myers Squibb), or panitumumab (sold under the tradename Vectibix® by Amgen);
HER2 receptor inhibitors: Trastuzumab (sold under the trademark Herceptin® by Genentech/Roche), neratinib (also known as HKI-272, (2E)-N-[4-[[3-chloro-4-[(pyridin-2- yl)methoxy]phenyl]amino]-3-cyano-7-ethoxyquinolin-6-yl]-4-(d imethylamino)but-2- enamide, and described PCT Publication No. WO 05/028443), lapatinib (CAS No. 231277-92-2) or lapatinib ditosylate (CAS No: 388082-77-7) (sold under the trademark Tykerb® by GlaxoSmithKline); or Trastuzumab emtansine (in the United States, ado-trastuzumab emtansine, trade name Kadcyla) - an antibody-drug conjugate consisting of the monoclonal antibody trastuzumab (Herceptin) linked to the cytotoxic agent mertansine (DM1);
HER dimerization inhibitors: Pertuzumab (sold under the trademark Omnitarg®, by Genentech);
FGFR inhibitors: Erdafitinib (CAS No. 1346242-81-6), Pemigatinib (CAS No. 1513857-77-6) or Infigratinib (CAS No. 872511-34-7)
Aurora kinase inhibitors: TAS-119 (CAS No. 1453099-83-6), LY3295668 (CAS No. 1919888-06-4), or alisertib (CAS No. 1028486-01-2); CD20 antibodies: Rituximab (sold under the trademarks Riuxan® and MabThera® by Genentech/Roche), tositumomab (sold under the trademarks Bexxar® by GlaxoSmithKline), or ofatumumab (sold under the trademark Arzerra® by GlaxoSmithKline);
Tyrosine kinase inhibitors: Erlotinib hydrochloride (CAS No. 183319-69-9, sold under the trademark Tarceva® by Genentech/Roche), Linifanib (N-[4-(3-amino-lH-indazol-4-yl)phenyl]-N'-(2- fluoro-5- methylphenyl)urea, also known as ABT 869, available from Genentech), sunitinib malate (CAS No. 341031-54-7, sold under the tradename Sutent® by Pfizer), bosutinib (4-[(2,4-dichloro-5- methoxyphenyl)amino]-6- methoxy-7-[3-(4-methylpiperazin4-yl)propoxy]quinoline-3-carbonitrile, also known as SKI-606, and described in US Patent No. 6,780,996), dasatinib (CAS No. 302962-49- 8, sold under the tradename Sprycel® by Bristol-Myers Squibb), armala (CAS No. 444731-52-6, also known as pazopanib, sold under the tradename Votrient® by GlaxoSmithKline), imatinib (CAS No. 152459-95-5) and imatinib mesylate (CAS No. 220127-57-1) (sold under the tradenames Gilvec® and Gleevec® by Novartis);
DNA Synthesis inhibitors: Capecitabine (CAS No. 154361-50-9) (sold under the trademark Xeloda® by Roche), gemcitabine hydrochloride (CAS No. 122111-03-9) (sold under the trademark Gemzar® by Eli Lilly and Company), or nelarabine ((2R3S,4R,5R)-2-(2-amino-6-methoxypurin-9- yl)-5-(hydroxymethyl)oxolane-3,4- diol, sold under the tradenames Arranon® and Atriance® by G laxo SmithKline) ;
Antineoplastic agents: oxaliplatin (CAS No. 61825-94-3) (sold under the tradename Eloxatin® ay Sanofi- Aventis and described in US Patent No. 4,169,846);
Human Granulocyte colony-stimulating factor (G-CSF) modulators: Filgrastim (sold under the tradename Neupogen® by Amgen);
Immunomodulators: Afutuzumab (available from Roche®), pegfilgrastim (sold under the tradename Neulasta® by Amgen), lenalidomide (CAS No. 191732-72-6, also known as CC-5013, sold under the tradename Revlimid®), or thalidomide (CAS No. 50-35-1, sold under the tradename Thalomid®);
CD40 inhibitors: Dacetuzumab (also known as SGN-40 or huS2C6, available from Seattle Genetics, Inc);
Pro-apoptotic receptor agonists (PARAs): Dulanermin (also known as AMG-951, available from Amgen/Genentech); Hedgehog antagonists: 2-chloro-N-[4-chloro-3-(2-pyridinyl)phenyl]-4-(methylsulfony 1)- benzamide (also known as GDC-0449, and described in PCT Publication No. WO 06/028958);
Phospholipase A2 inhibitors: Anagrelide (CAS No. 58579-51-4, sold under the tradename Agrylin®);
BCL-2 inhibitors: 4-[4-[[2-(4-chlorophenyl)-5,5-dimethyl-l-cyclohexen-l-yl]met hyl]- 1- piperazinyl]-N-[[4-[[(lR)-3-(4-morpholinyl)-l-[(phenylthio)m ethyl]propyl]amino]-3- [(trifluoromethyl)sulfonyl]phenyl]sulfonyl]benzamide (also known as ABT-263 and described in PCT Publication No. WO 09/155386);
MCL-1 inhibitors: MIK665 (CAS No. 1799631-75-6, S64315), AMG 397, and AZD5991 (CAS No. 2143010-83-5); Aromatase inhibitors: Exemestane (CAS No. 107868-30-4, sold under the trademark Aromasin® by Pfizer), letrozole (CAS No. 112809-51-5, sold under the tradename Femara® by Novartis), or anastrozole (CAS No. 120511-73-1, sold under the tradename Arimidex®);
Topoisomerase I inhibitors: Irinotecan (CAS No. 97682-44-5, sold under the trademark Camptosar® by Pfizer), topotecan hydrochloride (CAS No. 119413-54-6, sold under the tradename Hycamtin® by GlaxoSmithKline);
Topoisomerase II inhibitors: etoposide (CAS No. 33419-42-0, also known as VP-
16 and Etoposide phosphate, sold under the tradenames Toposar®, VePesid® and Etopophos®), or teniposide (CAS No. 29767-20-2, also known as VM-26, sold under the tradename Vumon®); mTOR inhibitors: Temsirolimus (CAS No. 162635-04-3, sold under the tradename Torisel® by Pfizer), ridaforolimus (CAS No. 572924-54-0, formally known as deferolimus, AP23573 and MK8669, and described in PCT Publication No. WO 03/064383), or everolimus (CAS No. 159351- 69-6, sold under the tradename Afmitor® by Novartis);
Proteasome inhibitor such as carfilzomib (CAS No. 868540-17-4), MLN9708 (CAS No. 1201902-80-8), delanzomib (CAS No. 847499-27-8), or bortezomib (CAS No. 179324-69-7);
BET inhibitors such as INCB054329 (CAS No. 1628607-64-6), OTX015 (CAS No. 202590- 98-5), or CPI-0610 (CAS No. 1380087-89-7);
LSD1 inhibitors such as GSK2979552, or INCB059872;
HIF-2a inhibitors such as PT2977 (1672668-24-4), NKT2152, or PT2385 (CAS No. 1672665- 49-4); Osteoclastic bone resorption inhibitors: l-hydroxy-2-imidazol-l-yl-phosphonoethyl) phosphonic acid monohydrate (sold under the tradename Zometa® by Novartis);
CD33 Antibody Drug Conjugates: Gemtuzumab ozogamicin (sold under the tradename Mylotarg® by Pfizer/Wyeth);
CD22 Antibody Drug Conjugates: Inotuzumab ozogamicin (also referred to as CMC-544 and WAY-207294, available from Hangzhou Sage Chemical Co., Ltd.);
CD20 Antibody Drug Conjugates: Ibritumomab tiuxetan (sold under the tradename Zevalin®);
Somatostain analogs: octreotide (also known as octreotide acetate, sold under the tradenames Sandostatin® and Sandostatin LAR®);
Synthetic Interleukin- 11 (IL-1 1): oprelvekin (sold under the tradename Neumega® by Pfizer/Wyeth);
Synthetic erythropoietin: Darbepoetin alfa (sold under the tradename Aranesp® by Amgen);
Receptor Activator for Nuclear Factor k B (RANK) inhibitors: Denosumab (sold under the tradename Prolia® by Amgen);
Thrombopoietin mimetic peptibodies: Romiplostim (sold under the tradename
Nplate® by Amgen;
Cell growth stimulators: Palifermin (sold under the tradename Kepivance® by Amgen);
Anti-insulin-like Growth Factor-1 receptor (IGF-1R) antibodies: Figitumumab (also known as CP-751,871, available from ACC Corp), robatumumab (CAS No. 934235-44-6);
Anti-CSl antibodies: Elotuzumab (HuLuc63, CAS No. 915296-00-3);
CD52 antibodies: Alemtuzumab (sold under the tradename Campath®);
Histone deacetylase inhibitors: Voninostat (sold under the tradename Zolinza® by Merck);
Alkylating agents: Temozolomide (sold under the tradenames Temodar® and Temodal® by Schering-Plough/Merck), dactinomycin (also known as actinomycin-D and sold under the tradename Cosmegen®), melphalan (also known as L-PAM, L-sarcolysin, and phenylalanine mustard, sold under the tradename Alkeran®), altretamine (also known as hexamethylmelamine (HMM), sold under the tradename Hexalen®), carmustine (sold under the tradename BiCNU®), bendamustine (sold under the tradename Treanda®), busulfan (sold under the tradenames Busulfex® and Myleran®), carboplatin (sold under the tradename Paraplatin®), lomustine (also known as CCNU, sold under the tradename CeeNU®), cisplatin (also known as CDDP, sold under the tradenames Platinol® and Platinol®-AQ), chlorambucil (sold under the tradename Leukeran®), cyclophosphamide (sold under the tradenames Cytoxan® and Neosar®), dacarbazine (also known as DTIC, DIC and imidazole carboxamide, sold under the tradename DTIC -Dome®), altretamine (also known as hexamethylmelamine (HMM) sold under the tradename Hexalen®), ifosfamide (sold under the tradename Ifex®), procarbazine (sold under the tradename Matulane®), mechlorethamine (also known as nitrogen mustard, mustine and mechloroethamine hydrochloride, sold under the tradename Mustargen®), streptozocin (sold under the tradename Zanosar®), thiotepa (also known as thiophosphoamide, TESPA and TSPA, sold under the tradename Thioplex®;
Biologic response modifiers: bacillus calmette-guerin (sold under the tradenames theraCys® and TICE® BCG), or Denileukin diftitox (sold under the tradename Ontak®);
Anti-tumor antibiotics: doxorubicin (sold under the tradenames Adriamycin® and Rubex®), bleomycin (sold under the tradename lenoxane®), daunorubicin (also known as dauorubicin hydrochloride, daunomycin, and rubidomycin hydrochloride, sold under the tradename Cerubidine®), daunorubicin liposomal (daunorubicin citrate liposome, sold under the tradename DaunoXome®), mitoxantrone (also known as DHAD, sold under the tradename Novantrone®), epirubicin (sold under the tradename Ellence™), idarubicin (sold under the tradenames Idamycin®, Idamycin PFS®), or mitomycin C (sold under the tradename Mutamycin®);
Anti -microtubule agents: Estramustine (CAS No. 52205-73-9, sold under the tradename Emcyl®);
Cathepsin K inhibitors: Odanacatib (CAS No. 603139-19-1, also know as MK-0822 available from Lanzhou Chon Chemicals, ACC Corp., and Chemi eTek, and described in PCT Publication no. WO 03/075836);
Epothilone B analogs: Ixabepilone (CAS No. 219989-84-1, sold under the tradename Lxempra® by Bristol- Myers Squibb);
Heat Shock Protein (HSP) inhibitors: Tanespimycin (17-allylamino-17- demethoxygeldanamycin, also known as KOS-953 and 17-AAG, available from SIGMA, and described in US Patent No. 4,261,989), NVP-HSP990 (CAS No. 934343-74-5), AUY922 (CAS No. 747412-49-3), AT13387 (CAS No. 912999-49-6), STA-9090 (CAS No. 888216-25-9), Debio 0932, KW-2478 (CAS No. 819812-04-9), XL888 (CAS No. 1149705-71-4), CNF2024 (CAS No. 848695- 25-0), and TAS-116 (CAS No. 1260533-36-5); TpoR agonists: Eltrombopag (sold under the tradenames Promacta® and Revolade® by G laxo SmithKline) ;
Anti-mitotic agents: Docetaxel (CAS No. 114977-28-5, sold under the tradename Taxotere® by Sanofi- Aventis); Adrenal steroid inhibitors: aminoglutethimide (CAS No. 125-84-8, sold under the tradename Cytadren®);
Anti-androgens: Nilutamide (CAS No. 63612-50-0, sold under the tradenames Nilandron® and Anandron®), bicalutamide (CAS No. 90357-06-5, sold under tradename Casodex®), or flutamide (CAS No. 13311-84-7, sold under the tradename Fulexin™);
Androgens: Fluoxymesterone (CAS No. 76-43-7, sold under the tradename Halotestin®);
CDK (CDK1, CDK2, CDK3, CDK5, CDK7, CDK8, or CDK9) inhibitors including but not limited to: Alvocidib (CAS No. 146426-40-6, pan-CDK inhibitor, also known as flovopirdol or HMR-1275, 2-(2-chlorophenyl)-5,7-dihydroxy-8-[(3S,4R)-3-hydroxy-l-methyl -4-piperidinyl]-4- chromenone, and described in US Patent No. 5,621,002);
CDK2 inhibitor PF-07104091;
CDK4/6 inhibitors: pabociclib (CAS No. 827022-33-3), ribociclib (CAS No. 1211441-98-3), abemaciclib (CAS No. 1231929-97-7), PF-06873600 (CAS No. 2185857-97-8), NUV-422 and Trilaciclib (CAS No. 1374743-00-6);
CDK7 inhibitors CT7001 (CAS No. 1805789-54-1) and SY- 1365 (CAS No. 1816989-16-8);
CDK9 inhibtiors AZD 4573 (CAS No. 2057509-72-3), P276-00 (CAS No. 920113-03-7), AT7519 (CAS No. 844442-38-2), CYC065 (CAS No. 1070790-89-4) or TP-1287;
Gonadotropin-releasing hormone (GnRH) receptor agonists: Leuprolide or leuprolide acetate (sold under the tradenames Viadure® by Bayer AG, Eligard® by Sanofi-Aventis and Lupron® by Abbott Lab);
Taxane anti-neoplastic agents: Cabazitaxel (l-hydroxy-7,10 -dimethoxy-9-oxo-5,20- epoxytax-1 l-ene-2a,4,13a-triyl-4-acetate-2-benzoate-13-[(2R,3S)-3-{ [(tert- butoxy)carbonyl]- amino}-2-hydroxy-3-phenylpropanoate), or larotaxel ((2a,3x,4a,5b,7a,10b,13a)- 4,10-bis(acetyloxy)- 13-({ (2R,3S)-3-[(tert-butoxycarbonyl)amino]-2-hydroxy-3- phenylpropanoyl}oxy)-l-hydroxy-9-oxo- 5,20-epoxy-7,19-cyclotax-l l-en-2-ylbenzoate);
5HTla receptor agonists: Xaliproden (also known as SR57746, l-[2-(2- naphthyl)ethyl]-4-[3- (trifluoromethyl)phenyl]-l,2,3,6-tetrahydropyridine, and described in US Patent No. 5,266,573);
HPC vaccines: Cervarix® sold by GlaxoSmithKline, Gardasil® sold by Merck; Iron Chelating agents: Deferasinox (CAS No. 201530-41-8, sold under the tradename Exjade® by Novartis);
Anti -metabolites: Claribine (2-chlorodeoxyadenosine, sold under the tradename leustatin®), 5 -fluorouracil (sold under the tradename Adrucil®), 6-thioguanine (sold under the tradename Purinethol®), pemetrexed (sold under the tradename Alimta®), cytarabine (also known as arabinosylcytosine (Ara-C), sold under the tradename Cytosar-U®), cytarabine liposomal (also known as Liposomal Ara-C, sold under the tradename DepoCyt™), decitabine (sold under the tradename Dacogen®), hydroxyurea (sold under the tradenames Hydrea®, Droxia™ and Mylocel™), fludarabine (sold under the tradename Fludara®), floxuridine (sold under the tradename FUDR®), cladribine (also known as 2-chlorodeoxyadenosine (2-CdA) sold under the tradename Leustatin™), methotrexate (also known as amethopterin, methotrexate sodim (MTX), sold under the tradenames Rheumatrex® and Trexall™), or pentostatin (sold under the tradename Nipent®);
Bisphosphonates: Pamidronate (CAS No. 57248-88-1, sold under the tradename Aredia®), zoledronic acid CAS No. 118072-93-8 (sold under the tradename Zometa®);
Demethylating agents: 5-azacitidine (CAS No. 320-67-2, sold under the tradename Vidaza®), decitabine (CAS No. 2353-33-5, sold under the tradename Dacogen®);
Plant Alkaloids: Paclitaxel protein-bound (sold under the tradename Abraxane®), vinblastine (also known as vinblastine sulfate, vincaleukoblastine and VLB, sold under the tradenames Alkaban- AQ® and Velban®), vincristine (also known as vincristine sulfate, LCR, and VCR, sold under the tradenames Oncovin® and Vincasar Pfs®), vinorelbine (sold under the tradename Navelbine®), or paclitaxel (sold under the tradenames Taxol and Onxal™);
Retinoids: Ali tretinoin (sold under the tradename Panretin®), tretinoin (all-trans retinoic acid, also known as ATRA, sold under the tradename Vesanoid®), Isotretinoin (13- cis-retinoic acid, sold under the tradenames Accutane®, Amnesteem®, Claravis®, Clarus®, Decutan®, Isotane®, Izotech®, Oratane®, Isotret®, and Sotret®), or bexarotene (sold under the tradename Targretin®);
Glucocorticosteroids: Hydrocortisone (also known as cortisone, hydrocortisone sodium succinate, hydrocortisone sodium phosphate, and sold under the tradenames Ala- Cort®, Hydrocortisone Phosphate, Solu-Cortef®, Hydrocort Acetate® and Lanacort®), dexamethazone ((8S, 9R, 10S, 1 IS, 13S,14S,16R, 17R)-9-fluoro-l 1,17-dihydroxy-17-(2- hydroxyacetyl)- 10, 13,16- trimethyl-6,7,8,9, 10,11,12, 13, 14, 15,16, 17-dodecahydro-3H- cyclopenta[a]phenanthren- 3-one), prednisolone (sold under the tradenames Delta-Cortel®, Orapred®, Pediapred® and Prelone®), prednisone (sold under the tradenames Deltasone®, Liquid Red®, Meticorten® and Orasone®), or methylprednisolone (also known as 6-Methylprednisolone, Methylprednisolone Acetate, Methylprednisolone Sodium Succinate, sold under the tradenames Duralone®, Medralone®, Medrol®, M-Prednisol® and Solu- Medrol®);
Cytokines: interleukin-2 (also known as aldesleukin and IL-2, sold under the tradename Proleukin®), interleukin- 11 (also known as oprevelkin, sold under the tradename Neumega®), alpha interferon alfa (also known as IFN-alpha, sold under the tradenames Intron® A, and Roferon-A®);
Estrogen receptor downregulators: Fulvestrant (CAS No. 129453-61-8, sold under the tradename Faslodex®);
Anti-estrogens: tamoxifen (CAS No. 10540-29-1, sold under the tradename Novaldex®); or Toremifene (CAS No. 89778-27-8, sold under the tradename Fareston®);
Selective estrogen receptor modulators (SERMs): Raloxifene (CAS No. 84449-90-1, sold under the tradename Evista®);
Leutinizing hormone releasing hormone (LFfRH) agonists: Goserelin (CAS No. 145781-92-6, sold under the tradename Zoladex®); Progesterones: megestrol (also known as megestrol acetate, CAS No. 595-33-5, sold under the tradename Megace®);
Miscellaneous cytotoxic agents: Arsenic trioxide (sold under the tradename Trisenox®), or asparaginase (also known as L-asparaginase, Erwinia L-asparaginase, sold under the tradenames Elspar® and Kidrolase®);
Exemplary immune checkpoint inhibitors include inhibitors (smack molecules or biologies) against immune checkpoint molecules such as CD27, CD28, CD40, CD 122, CD96, CD73, CD39, CD47, 0X40, GITR, CSF1R, JAK, PI3K delta, PI3K gamma, TAM kinase, arginase, CD 137 (also known as 4-1BB), ICOS, A2AR, A2BR, HIF-2a, B7-H3, B7- H4, BTLA, CTLA-4, LAG3, TIM3, VISTA, CD96, TIGIT, PD-1, PD-L1 and PD-L2. In some embodiments, the immune checkpoint molecule is a stimulatory checkpoint molecule selected from CD27, CD28, CD40, ICOS, 0X40, GITR, CD 137 and STING. In some embodiments, the immune checkpoint molecule is an inhibitory checkpoint molecule selected from B7-H3, B7-H4, BTLA, CTLA-4, IDO, TDO, Arginase, KIR, LAG3, PD-1, TIM3, CD96, TIGIT and VISTA. In some embodiments, the compounds provided herein can be used in combination with one or more agents selected from KIR inhibitors, TIGIT inhibitors, LAIR1 inhibitors, CD 160 inhibitors, 2B4 inhibitors and TGFR beta inhibitors. In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of PD-1, e.g., an anti-PD-1 monoclonal antibody. In some embodiments, the anti-PD-1 monoclonal antibody is nivolumab, pembrolizumab (also known as MK-3475), pidilizumab, SHR-1210, PDR001, or AMP - 224. In some embodiments, the anti-PD-1 monoclonal antibody is nivolumab, or pembrolizumab or PDR001. In some embodiments, the anti -PD 1 antibody is pembrolizumab.
In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of PD- Ll, e.g., an anti-PD-Ll monoclonal antibody. In some embodiments, the anti-PD-Ll monoclonal antibody is BMS-935559, MEDI4736, MPDL3280A (also known as RG7446), or MSB0010718C. In some embodiments, the anti-PD-Ll monoclonal antibody is MPDL3280A (atezolizumab) or MEDI4736 (durvalumab). In some embodiments, the anti-PD-Ll small molecule inhibitor is INCB86550.
In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of CTLA-4, e.g., an anti-CTLA-4 antibody. In some embodiments, the anti-CTLA-4 antibody is ipilimumab or tremelimumab. In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of LAG3, e.g., an anti-LAG3 antibody. In some embodiments, the anti-LAG3 antibody is BMS-986016 or LAG525. In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of GITR, e.g., an anti-GITR antibody. In some embodiments, the anti-GITR antibody is TRX518 or, MK-4166, INCAGN01876 or MK-1248. In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of 0X40, e.g., an anti-OX40 antibody or OX40L fusion protein. In some embodiments, the anti-OX40 antibody is MED 10562 or, INCAGNO 1949, GSK2831781, GSK-3174998, MOXR-0916, PF-04518600 or LAG525. In some embodiments, the OX40L fusion protein is MEDI6383.
Compounds of the invention can also be used to increase or enhance an immune response, including increasing the immune response to an antigen; to improve immunization, including increasing vaccine efficacy; and to increase inflammation. In some embodiments, the compounds of the invention can be sued to enhance the immune response to vaccines including, but not limited, Listeria vaccines, oncolytic viral vaccines, and cancer vaccines such as GV AX® (granulocytemacrophage colony-stimulating factor (GM-CF) gene- transfected tumor cell vaccine). Anti-cancer vaccines include dendritic cells, synthetic peptides, DNA vaccines and recombinant viruses. Other immune-modulatory agents also include those that block immune cell migration such as antagonists to chemokine receptors, including CCR2 and CCR4; Sting agonists and Toll receptor agonists. Other anti-cancer agents also include those that augment the immune system such as adjuvants or adoptive T cell transfer. Compounds of this application may be effective in combination with CAR (Chimeric antigen receptor) T cell treatment as a booster for T cell activation.
A compound of the invention can also be used in combination with the following adjunct therapies: Anti-nausea drugs: NK-1 receptor antagonists: Casopitant (sold under the tradenames Rezonic® and Zunrisa® by GlaxoSmithKline); and Cytoprotective agents: Amifostine (sold under the tradename Ethyol®), leucovorin (also known as calcium leucovorin, citrovorum factor and folinic acid). The disclosure of the PCT applications referred to herein above are incorporated herein by reference in their entirety.
Examples
The following preparations of Intermediates (References) and compounds of Formula (IA) or (I) (Examples) are given to enable those skilled in the art to more clearly understand and to practice the present disclosure. They should not be considered as limiting the scope of the disclosure, but merely as being illustrative and representative thereof.
Example 1
Synthesis ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen- 1 -yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy) methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(2-((2-(2,6-dioxo piperidin-3-yl)-l ,3-dioxoisoindolin-4- yl)amino)ethyl)piperazine- 1 -carboxylate
Figure imgf000205_0001
Step 1 : (2S)-tert-butyl 2-(hydroxymethyl)-5-methoxypyrrolidine- 1 -carboxylate
Figure imgf000205_0002
To a solution of (S)- 1 -tert-butyl 2-methyl 5-oxopyrrolidine-l,2-dicarboxylate (150 g, 617 mmol) in DCM (900 mL) was added DIBAL-H (1 M, 2.47 L) and the resulting mixture was stirred at -78 °C for 0.5 h, then at 20°C for 2 h. After cooling to 0 °C, methyl alcohol (3000 mL) and 2 M HC1 (5000 mL) were added. The mixture was stirred for 2 h at room temperature and then extracted with EtOAc. The combined organic layer was washed with brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by column chromatography, eluted with EA/PE (0-50%), to afford the title compound (42.0 g, 181 mmol, 36.8% yield) was obtained as a yellow oil.
Step 2 : (5S)-tert-butyl 2-cyano-5-(hydroxymethyl)pyrrolidine-l -carboxylate
Figure imgf000206_0001
To a solution of (2S)-tert-butyl 2-(hydroxymethyl)-5-methoxypyrrolidine-l -carboxylate (48.0 g, 208 mmol) in DCM (500 mL) at -70 °C was added TMSCN (51.5 g, 519 mmol, 64.9 mL) dropwise and then BF3.Et2O (64.8 g, 457 mmol, 56.4 mL). The resulting mixture was then stirred at -70 °C for 1 h and then, then quenched with sat. NaHCO3, aq. solution. The mixture was then extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography, eluted with EA/PE (0-20%), to afford the title compound (31.0 g).
Step3: tert-butyl (2S)-2-[[tert-butyl(diphenyl)silyl]oxymethyl]-5-cyano-pyrrolidine-l -carboxylate
Figure imgf000206_0002
To a solution of tert-butyl (5 S)-2-cyano-5-(hydroxymethyl)pyrrolidine-l -carboxylate (50 g, 220 mmol, 1 eq) in THF (500 mL) was added imidazole (37.6 g, 552.43 mmol, 2.5 eq) and tert-butyl- chlorodiphenylsilane (66.8 g, 243 mmol, 62.4 mL, 1.1 eq) at 0 °C. The mixture was stirred at 20 °C for 12 h and then quenched with H2O, and extracted with EA. The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography, eluted with EA/PE (0-20%), to afford the title compound (69.0 g).
Step 4: (5 S)-l -tert-butyl 2-methyl 5-(((tert-butyldiphenylsilyl)oxy)methyl)pyrrolidine-l,2- dicarboxylate
Figure imgf000207_0003
A mixture of tert-butyl (2S)-2-[[tert-butyl(diphenyl)silyl]oxymethyl]-5-cyano-pyrrolidine-l- carboxylate (69.0 g, 148 mmol, 1 eq) and K2CO3 (26.6 g, 193 mmol, 1.3 eq) in MeOH (500 mL) was stirred at 50 °C for 12 h under N2 atmosphere. The mixture was acidified with 1.0 M HC1 aq. to pH= 4 and then extracted with EA. The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography, eluted with EA/PE (0-15%), to afford the title compound (12 g, 19.53 mmol).
Step 5: 1 -(tert-butyl) 2-methyl (2S,5S)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2-(2- (chloromethyl)allyl)pyrrolidine- 1 ,2-dicarboxylate
Figure imgf000207_0001
To a solution of (5 S)- 1 -tert-butyl 2-methyl 5-(((tert-butyldiphenylsilyl)oxy)methyl)- pyrro li dine- 1 ,2-dicarboxylate (12.0 g, 24.1 mmol, 1 eq) in THF (100 mL) was added LiHMDS (1 M, 48.22 mL, 2 eq) at -70 °C and the mixture was stirred at -70 °C for 15 min. 3-Chloro-2- (chloromethyl)prop-l-ene (4.52 g, 36.1 mmol, 4.19 mL, 1.5 eq) was added dropwise at -70 °C under nitrogen atmosphere and the resulting mixture was stirred 16 h at 20 °C. The mixture was quenched with sat. NaHCO3, aq. and extracted with EtOAc. The combined organic layers were washed with water and brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by column chromatography, eluted with EA/PE (0-10%), to afford the title compound (11 g).
Step 6: methyl (5S,7aS)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2-methylenetetrahydro-lH- pyrrolizine-7a(5H)-carboxylate
Figure imgf000207_0002
To a solution of 1 -(tert-butyl) 2-methyl (2S,5S)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2-(2- (chloromethyl)allyl)pyrrolidine-l,2-dicarboxylate (10.7 g, 18.2 mmol, 1 eq) in DCM (90 mL) was added TFA (45 mL) at 0 °C and the resulting mixture was stirred at 20 °C for 2 h. The reaction was quenched by adding sat. aq. NaHCO3 solution at 0 °C and extracted with DCM. The combined organic layers were washed with brine, dried over Na2SO4 filtered and concentrated. The residue was purified by column chromatography eluted with EA/PE (0-10%), to afford the title compound (3.2 g, crude).
Step 7: ((5S,7aS)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2-methylenetetrahydro-lH-pyrrolizin-
7 a(5H)-yl)methanol
Figure imgf000208_0001
To a solution of LiA1H4 (540 mg, 14.2 mmol, 2 eq) in THF (20 mL) was added dropwise methyl (5S,7aS)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2-methylenetetrahydro-lH-pyrrolizine- 7a(5H)-carboxylate (3.2 g, 7.12 mmol, 1 eq) in THF (10 mL) at -40 °C and the resulting mixture was stirred at -40 °C for 1 h. After cooling the mixture to 0 °C, the mixture was quenched by adding Na2SO4.10H2O. The mixture was then filtered and concentrated under reduced pressure to give the title compound (2.83 g, crude).
Step 8: (lR,5S)-tert-butyl 3 -(2-(((5S,7aS)-5 -(((tert-butyl diphenylsilyl)oxy) methyl)-2-methylene- hexahydro-lH-pyrrolizin-7a-yl)methoxy)-7-chloro-8-fluoropyrido[4,3-d] pyrimidin-4-yl)-3,8- diazabicyclo[3.2.1]octane-8-carboxylate
Figure imgf000208_0002
To a solution of ((5S,7aS)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2-methylene-hexahydro- lH-pyrrolizin-7a-yl)methanol (886 mg, 2.10 mmol, 1.0 eq) in THF (1.0 mL) was added NaH (168.1 mg, 4.20 mmol, 60% in mineral oil, 2.0 eq) in an ice water bath, followed by (lR,5S)-tert-butyl 3- (2,7-dichloro-8-fluoropyrido[4,3-d] pyrimidin-4-yl)-3,8-diazabicyclo-[3.2.1]octane-8-carboxylate (900 mg, 2.10 mmol, 1.0 eq). The resulting mixture was stirred at 20 °C for 2 h. The reaction mixture was quenched with water at 5 °C, and then extracted with EtOAc. The combined organic layers were dried over Na2SO4 and concentrated. The residue was purified by column chromatography, eluted with DCM/MeOH (0-10%), to give the title compound (1.3 g). Step 9: (lR,5S)-tert-butyl 3-(2- (((5S,7aS)-5-(((tert-butyldiphenylsilyl)oxy) methyl)-2-methylenehexahydro-lH-pyrrolizin-7a- yl)methoxy)-8-fluoro-7-(7-fluoro-3-(methoxy methoxy)-8-((triisopropylsilyl)ethynyl)naphthalen-l- yl)pyrido [4,3-d]pyrimidin-4-yl)-3,8-diazabicyclo-[3.2.1]octane-8-carboxylate
Figure imgf000209_0001
A mixture of (lR,5S)-tert-butyl 3-(2-(((5S,7aS)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2- methylenehexahydro-lH-pyrrolizin-7a-yl) methoxy)-7-chloro-8-fluoropyrido[4,3-d]pyrimidin-4-yl)- 3,8-diazabicyclo[3.2.1] octane-8-carboxylate (1.2 g, 1.48 mmol, 1.0 eq), ((2-fluoro-6- (methoxymethoxy)-8-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)naphthalen-l-yl)ethynyl)- triisopropylsilane (986 mg, 1.62 mmol, 1.1 eq), cataCXiumAPdG3 (110.3 mg, 0.15 mmol, 0.1 eq), K2CO3 (610.7 mg, 4.43 mmol, 3.0 eq) in DME (9.0 mL) and H2O (3.00 mL) was stirred at 85 °C for 5 h under N2 atmosphere. The mixture was filtered and concentrated, and the residue was purified by column chromatography, eluted with EtOAc/PE (0-30%), to give the title compound (1.6 g). Step 10: (lR,5S)-tert-butyl 3-(7-(8-ethynyl-7-fluoro-3-(methoxymethoxy) naphthal en-l-yl)-8-fluoro- 2-(((5S,7aS)-5-(hydroxymethyl)-2-methylene hexahydro- lH-pyrrolizin-7a-yl)methoxy)pyrido[4, 3- d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1] octane-8-carboxylate
Figure imgf000209_0002
To a solution of (lR,5S)-tert-butyl 3-(2-(((5S,7aS)-5-(((tert-butyldiphenylsilyl)oxy)-methyl)- 2-methylenehexahydro-lH-pyrrolizin-7a-yl) methoxy)-8-fluoro-7-(7-fluoro-3-(methoxymethoxy)-8- ((triisopropylsilyl)ethynyl) naphthalen-l-yl)pyrido[4,3-d]pyrimidin-4-yl)-3,8- diazabicyclo[3.2.1]octane-8-carboxylate (1.6 g, 1.38 mmol, 1.0 eq) in THF (10.0 mL) was added TBAF (1.0 M, 6.9 mL, 5.0 eq) and the resulting mixture was stirred at rt for 3 h. After cooling in an ice-water bath, the mixture was quenched by adding water and then extracted with EtOAc. The combined organic layers were dried over Na2 SO4, filtered and concentrated. The residue was purified by column chromatography, eluted with DCM/MeOH (0-5%), to give the title compound (980 mg). Step 10: tert-butyl (lR,5S)-3-(7-(8-ethynyl-7-fluoro-3 -(methoxymethoxy) naphthal en-l-yl)-8-fluoro-
2-(((5S,7aS)-2-methylene-5-((((4-nitrophenoxy) carbonyl)oxy)methyl)tetrahydro-lH-pyrrolizin-
7a(5H)-yl)methoxy)pyrido[4,3-d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate
Figure imgf000210_0001
To a solution of tert-butyl (lR,5S)-3-(7-(8-ethynyl-7-fluoro-3-(methoxymethoxy) naphthalen- l-yl)-8-fluoro-2-(((5S,7aS)-5-(hydroxymethyl)-2-methylene tetrahydro- 1 H-pyrrolizin-7a(5H)- yl)methoxy)pyrido[4,3-d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2. l]octane-8-carboxylate (50 mg, 0.07 mmol) in anhydrous THF (1.5 mL) at 0 °C was added triethylamine (0.09 mL, 0.65 mmol), followed by 4-nitrophenyl carbonochloridate (65.62 mg, 0.33 mmol) under argon atmosphere. The resulting mixture was gradually warmed to rt and stirred for 18 h. Additional 2.0 eq of (4-nitrophenyl) carbonochloridate (26.249 mg, 0.1302 mmol) was added and the mixture was stirred for 40 h. Water was then added and the mixture was extracted with EtOAc. The combined organic layers were washed with water, brine, dried over Na2SO4 and concentrated to afford title compound (60 mg). Step 11: tert-butyl (lR,5S)-3-(2-(((5S,7aS)-5-(((4-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3- dioxoisoindolin-4-yl)amino)ethyl)piperazine- 1 -carbonyl)oxy)methyl)-2-methylenetetrahydro- 1 H- pyrrolizin-7a(5H)-yl)methoxy)-7-(8-ethynyl-7-fluoro-3-(methoxymethoxy)naphthalen-l-yl)-8- fluoropyrido[4,3-d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate
Figure imgf000210_0002
A mixture of tert-butyl (lR,5S)-3-(7-(8-ethynyl-7-fluoro-3-(methoxymethoxy)naphthalen-l- yl)-8-fluoro-2-(((5S,7aS)-2-methylene-5-((((4-nitro phenoxy)carbonyl)oxy)methyl)tetrahydro-lH- pyrrolizin-7a(5H)-yl)methoxy)pyrido[4,3-d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1]octane-8- carboxylate (50 mg, crude) and 4-((2-(2-(2-aminoethoxy)ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin- 3-yl)isoindoline-l,3-dione (prepared as described in J Med. Chem. 2022, 65, 13, 9096-9125) (61.90 mg, 0.16 mmol, 3.0 eq), and TEA (43.26 mg, 0.43 mmol, 8.0 eq) in THF (1 mL) was stirred at 25°C for 3 hrs under N2. The reaction mixture was quenched with sat. NaHC'O, aq. solution and then extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2SO4 and concentrated. The residue obtained was purified by flash chromatography, eluted with DCM/MeOH (0-5%) to afford the title compound (8 mg).
Step 12: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl) -6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(2-((2-(2,6-dioxopiperi din-3 -yl)- 1,3 -di oxoisoindolin-4- yl)amino)ethyl)piperazine- 1 -carboxylate
Figure imgf000211_0001
To a solution of tert-butyl (lR,5S)-3-(2-(((5S,7aS)-5-(12-((2-(2,6-dioxopiperidin-3-yl)-l,3- dioxoisoindolin-4-yl)amino)-3 -oxo-2, 7,10-trioxa-4-azadodecyl)-2-methylenetetrahydro- 1H- pyrrolizin-7a(5H)-yl)methoxy)-7-(8-ethynyl-7-fluoro-3-(methoxymethoxy)naphthalen-l-yl)-8- fluoropyrido[4,3-d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate (8 mg, 6.8 pmol) in DCM (1.0 mL) was added a solution of HC1 in dioxane (4.0 M, 0.5 mL) at 5 °C and the resulting mixture was stirred at 5 °C for 3 hs. The mixture was concentrated in reduced pressure and the residue was basified to pH = 8 by adding a solution of ammonia in MeOH. After volatiles were removed under reduced pressure, the residue was purified by HPLC to afford the title compound (2.7 mg). MS (ES, m/z): [M+H]+ =1036.3.
Example 2
Synthesis ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen- 1 -yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl) -6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)ethoxy)ethoxy)ethyl) carbamate
Figure imgf000212_0001
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 4-((2-(2-(2-aminoethoxy)ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione (ref. J. Med. Chem. 2019, 62, 448 - 466) instead of 4-((2-(2-(2- aminoethoxy)ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione in Step 11. MS (ES, m/z): [M+H]+ =1055.4.
Example 3
Synthesis ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen- 1 -yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy) methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (5-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)pentyl)carbamate
Figure imgf000212_0002
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 4-((2-(2-(2-aminoethoxy)ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline- 1,3-dione (ref. J. Med. Chem. 2019, 62, 448 - 466) instead of 4-((2-(2-(2- aminoethoxy)ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione in Step 11. MS (ES, m/z): [M+H]+ =1009.4.
Example 4
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (8-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-8-
Figure imgf000213_0001
Step 1: tert-butyl (8-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-8- oxooctyl)carbamate
Figure imgf000213_0002
A mixture of 5-amino-2-(2,6-dioxopiperidin-3-yl)isoindoline-l, 3-dione (200 mg, 0.73 mmol) and 8-((tert-butoxycarbonyl)amino)octanoic acid (227.79 mg, 0.88 mmol) in anhydrous N,N- Dimethylformamide (4 mL) was added 1 -methylimidazole (420.64 mg, 5.12 mmol), followed by chloro-N,N,N',N'-tetramethylformamidinium hexafluorophosphate (616.1 mg, 2.2 mmol) at 25 °C for 3 h under argon atmosphere and stirred. The mixture was dissolved in water and extracted with ethyl acetate. The combined organic layers were washed with water, brine, dried over anhydrous sodium sulphate. After filtration, the filtrate was concentrated under reduced pressure. The residue obtained was purified by Prep-HPLC (0~70% acetonitrile in water with 0.05% formic acid) to afford title compound (200 mg, 0.3887 mmol, 53.102% yield) as an off-white solid.
Step 2: 8-amino-N-(2-(2,6-dioxopiperidin-3-yl)-l ,3-dioxoisoindolin-5-yl)octanamide hydrochloride
Figure imgf000213_0003
To a stirred solution of tert-butyl (8-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5- yl)amino)-8-oxooctyl)carbamate (50 mg, 0.0972 mmol) in anhydrous dichloromethane (1 mL) was added 4M hydrogen chloride in dioxane (0.5 mL) at 0 °C and the mixture was stirred for 1 h. The solvent was removed under reduced pressure to afford title compound (40 mg, 0.0887 mmol, 91.29% yield) as a white solid, which was used to next step directly without further purification.
Step 3: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy) methyl)-6-methylene-hexahydro- lH-pyrrolizin-3-yl)methyl (8-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-8- oxooctyl)carbamate
Figure imgf000214_0001
The title compound was prepared by proceeding analogously as described in Example 1 , Steps
11 and 12, using 8-amino-N-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)octanamide hydrochloride instead of 4-((2-(2-(2-aminoethoxy)ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline- 1,3-dione in Step 11. LCMS (ESI) m/z [M+H]+ = 1065.
Example 5
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (8-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)amino)-8- oxooctyl)carbamate
Figure imgf000214_0002
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 8-amino-N-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)octanamide hydrochloride (ref: WO2021/201577) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)-ethyl)amino)-2- (2, 6-dioxopiperidin-3-yl)isoindoline-l, 3-dione in Step 11. LCMS (ESI) m/z [M+H]+ = 1065.
Example 6
Synthesis of ((3R,7aR)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (2-(2-(2-(3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)- 3-oxopropoxy)ethoxy)ethoxy)ethyl)carbamate
Figure imgf000215_0001
The title compound was prepared by proceeding analogously as described in Example 4, Steps 1-3, using 2,2-dimethyl-4-oxo-3,8,l l,14-tetraoxa-5-azaheptadecan- 17-oic acid instead of 8-((tert- butoxycarbonyl)amino)octanoic acid in Step 1. LCMS (ESI) m/z [M+H]+ = 1126.3.
Example 7
Synthesis of ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen- 1 -yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy) methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (4-((2-(2,6-dioxo piperidin-3-yl)-l,3-dioxoisoindolin-5- yl)amino)butyl)carbamate
Figure imgf000215_0002
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 5-((4-aminobutyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l, 3-dione (ref: WO2021/155321) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline- 1,3-dione in Step 1. LCMS (ESI) m/z [M+H]+ = 995.3.
Example 8
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-3- oxopropyl)(methyl) carbamate
Figure imgf000216_0001
Step 1 : tert-butyl (3-((2-(2,6-dioxopiperidin-3-yl)-l ,3-dioxoisoindolin-5-yl)amino)-3-oxopropyl)- (methyl)carbamate
Figure imgf000216_0002
To a solution of 3- [tert-butoxycarbonyl (methyl)amino]propanoic acid (89.25 mg, 0.44 mmol) and 5-amino-2-(2,6-dioxo-3-piperidyl)isoindoline-l, 3-dione (100 mg, 0.37 mmol) in anhydrous DMF (2 mL) was added NMI (210.32 mg, 2.56 mmol), followed by TCFH (308.05 mg, 1.1 mmol) at 25 °C under argon atmosphere and the reaction mixture was stirred for 2 h at rt. The reaction mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with water, brine, dried over anhydrous sodium sulphate and concentrated under reduced pressure. The residue obtained was purified by Prep-HPLC (0~50% acetonitrile in water with 0.05% formic acid) to afford the title compound (100 mg).
Step 2: N-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)-3-(methylamino)propanamide
Figure imgf000216_0003
To a stirred solution of tert-butyl N-[3-[[2-(2,6-dioxo-3-piperidyl)-l,3-dioxo-isoindolin-5- yl]amino]-3-oxo-propyl]-N-methylcarbamate (50 mg, 0.11 mmol) in DCM (1 mL) was added 4M HC1 in dioxane (0.5mL) at 0 °C and the reaction mixture was stirred for 1 h. The solvent was removed under reduced pressure to afford the title compound (43 mg).
Step 3: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-3- oxopropyl)(methyl)carbamate
Figure imgf000217_0001
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using N-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)-3-(methylamino) propenamide instead of 4-((2-(2-(2-aminoethoxy)ethoxy)-ethyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline- 1,3-dione in Step 11. MS (ES, m/z): [M+H]+ = 1009.3.
Example 9
Synthesis ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-3- oxopropyl)carbamate
Figure imgf000217_0002
The title compound was prepared by proceeding analogously as described in Example 8, Steps 1- 3, using 3-(tert-butoxycarbonylamino)propanoic acid instead of 3-[tert-butoxycarbonyl(methyl)- amino]propanoic acid in Step 1. MS (ES, m/z): [M+H]+ = 995.3.
Example 10
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)ethyl)(methyl)carbamate
Figure imgf000217_0003
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 2-(2,6-dioxopiperidin-3-yl)-4-((2-(methylamino)ethyl)amino)isoindoline-l, 3-dione (ref: WO2022/147465) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)-ethyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline- 1,3-dione in Step 11. MS (ES, m/z): [M+H]+ = 981.3.
Example 11
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-
Figure imgf000218_0001
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 4-((2-aminoethyl)amino)-2-(2,6-dioxopiperidin-3-yl) isoindoline- 1,3-dione (ref: WO2022/147465A1) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)-ethyl)amino)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l, 3-dione in Step 11. MS (ES, m/z): [M+H]+ = 967.3.
Example 12
Synthesis of ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)- amino)ethyl)(methyl) carbamate
Figure imgf000218_0002
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 2-(2,6-dioxopiperidin-3-yl)-5-((2-(methylamino)ethyl)amino)isoindoline-l, 3-dione (ref: WO2022/99117) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)-ethyl)amino)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l, 3-dione in Step 11. MS (ES, m/z): [M+H]+ = 981.3.
Example 13
Synthesis of ((3R,7aR)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (2-(2-(2-(3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)amino)- 3-oxopropoxy)ethoxy)ethoxy)ethyl)carbamate
Figure imgf000219_0001
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 3-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)-N-(2-(2,6-dioxo piperidin-3-yl)-l,3- dioxoisoindolin-4-yl)propanamide (ref: J. Med. Chem. 2019, 62, 2508 - 2520) instead of 4-((2-(2-(2- aminoethoxy)ethoxy)-ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione in Step 11. MS (ES, m/z): [M+H]+ = 1127.3.
Example 14
Synthesis of ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (6-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5- yl)amino)hexyl)carbamate
Figure imgf000219_0002
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 5-((6-aminohexyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l, 3-dione (ref: WO2020/38415) instead of 4-((2-(2-(2-aminoethoxy) ethoxy)-ethyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline- 1,3-dione in Step 11. MS (ES, m/z): [M+H]+ = 1023.3. Example 15
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen- 1 -yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl) oxy)methyl)-6-methylenehexahydro-
1 H-pyrrolizin-3 -yl)methyl ( 1 -((3 -(3 -(4-(3 -(2,4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H- indazol-6-yl)piperidin-l-yl)-2-methylpropyl)phenyl)sulfonyl)piperidin-4-yl)carbamate
Figure imgf000220_0001
Step 1: tert-butyl (l-((3-(2-methyl-3-oxopropyl)phenyl)sulfonyl)piperidin-4-yl)carbamate
Figure imgf000220_0002
A mixture of palladium(II) acetate (53.54 mg, 0.24 mmol) and tetrabutylammonium bromide (4000 mg, 12.41 mmol) was heated to 130 °C under argon atmosphere, then tert-butyl (l-((3- bromophenyl)sulfonyl)piperidin-4-yl)carbamate (1.0 g, 2.38mmol), 2-methylprop-2-en-l-ol (515.89 mg, 7.15 mmol) and sodium bicarbonate (400.69 mg, 4.77 mmol) were added. The resulting mixture was stirred at 130 °C for 4 h. After cooling to rt, water and EtOAc were added, and the mixture was filtered. The phases were separated, and the aqueous layer was extracted with EtOAc. The combined organic layers were washed with water, brine, dried over anhydrous sodium sulphate and concentrated under reduced pressure, The residue was purified by silica gel chromatography, eluted with EtOAc/petroleum ester (0—21% with 5% dichloromethane) to afford the title compound (830 mg).
Step 2 : tert-butyl ( 1 -((3 -(3 -(4-(3 -(2,4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-6- yl)piperidin-l-yl)-2-methylpropyl)phenyl)sulfonyl)piperidin-4-yl)carbamate
Figure imgf000221_0001
A mixture of tert-butyl (l-((3-(2-methyl-3-oxopropyl)phenyl)sulfonyl)piperidin-4- yl)carbamate hydrochloride (100 mg, 0.24 mmol) and l-(l-methyl-6-(piperidin-4-yl)-lH-indazol-3- yl)dihydropyrimidine-2,4(lH,3H)-dione hydrochloride (88.63 mg, 0.24 mmol) in anhydrous dichloromethane (2 mL) was stirred at 25 °C for 3 h. Sodium triacetoxyborohydride (154.88 mg, 0.73 mmol) was added and the resulting mixture was stirred for another 16 h. The mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with water, brine, dried over anhydrous sodium sulphate and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluted with methanol/dichloromethane (0~5%) to afford the title compound (62 mg).
Step 3: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl) oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (1 -((3-(3-(4-(3-(2,4-dioxotetrahydropyrimidin-l(2H)-yl)-l -methyl- 1H- indazol-6-yl)piperidin-l-yl)-2-methylpropyl)phenyl)sulfonyl)piperidin-4-yl)carbamate
Figure imgf000221_0002
The title compound was prepared by proceeding analogously as described in Example 8, Steps 2 and 3, using tert-butyl (l-((3-(3-(4-(3-(2,4-dioxotetrahydropyrimidin-l(2H)-yl)-l-methyl-lH- indazol-6-yl)piperidin- 1 -yl)-2-methylpropyl)phenyl)sulfonyl)piperidin-4-yl)carbamate instead of tertbutyl N-[3-[[2-(2,6-dioxo-3-piperidyl)-l,3-dioxo-isoindolin-5-yl]amino]-3-oxo-propyl]-N- methylcarbamate in Step 2. MS (ES, m/z): [M+H]+ = 1272.6.
Example 16
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (5-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-5-
Figure imgf000222_0001
The title compound was prepared by proceeding analogously as described in Example 8, Steps 1-3, using 5-(tert-butoxycarbonylamino)pentanoic acid instead of 3-[tert-butoxycarbonyl(methyl)- amino]propanoic acid in Step 1. MS (ES, m/z): [M+H]+ = 1023.3.
Example 17
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (5-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)amino)-5-
Figure imgf000222_0002
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 5-amino-N-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)pentanamide hydrochloride (ref: CN114853735,2022, A) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)- ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione in Step 11. MS (ES, m/z): [M+H]+ = 1023.4
Example 18
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (7-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)hept-6-yn-l- yl)carbamate
Figure imgf000223_0001
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 4-(7-aminohept-l-yn-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l, 3-dione hydrochloride (ref: WO2021/201577) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)-ethyl)amino)-2- (2, 6-dioxopiperidin-3-yl)isoindoline-l, 3-dione in Step 11. MS (ES, m/z): [M+H]+ = 1018.4.
Example 19
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (7-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)heptyl)carbamate
Figure imgf000223_0002
Step 1: tert-butyl (7-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)heptyl)carbamate
Figure imgf000223_0003
To a solution of tert-butyl N-[7-[2-(2,6-dioxo-3-piperidyl)-l,3-dioxo-isoindolin-4-yl]hept-6- ynyl] carbamate (50 mg, 0.11 mmol) in ethanol (0.5 mL) and THF (0.5 mL) was added Pd/C (10 mg) and the resulting mixture was stirred for 40 h under H2 atmosphere. The mixture was filtered and the filtrate was concentrated under reduced pressure to afford the title compound (50 mg).
Step 2. ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (7-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)heptyl)carbamate
Figure imgf000224_0001
The title compound was prepared by proceeding analogously as described in Example 8, Steps 2 and 3, using tert-butyl (7-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)heptyl)carbamate instead of tert-butyl (3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-3- oxopropyl)(methyl)carbamate in Step 2. MS (ES, m/z): [M+H]+ = 1022.4.
Example 20
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (7-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)heptyl)carbamate
Figure imgf000224_0002
Step 1 : tert-butyl (7-(2-(2,6-dioxopiperidin-3-yl)-l ,3-dioxoisoindolin-5-yl)hept-6-yn-l-yl)carbamate
Figure imgf000224_0003
A mixture of 5-bromo-2-(2,6-dioxo-3-piperidyl)isoindoline-l, 3-dione (100 mg, 0.3 mmol), tert-butyl N-hept-6-ynylcarbamate (94.01 mg, 0.44 mmol), Pd(pph3)2C12 (20.82 mg, 0.03 mmol), Cui (11.3 mg, 0.06 mmol) and triethylamine (1.0 mL, 7.17 mmol) in anhydrous DMF (2 mL) was stirred at 70 °C for 3 h. After cooling to rt, the reaction mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with water, brine, dried over anhydrous sodium sulphate and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluted with methanol/dichloromethane (0~4%) to afford the title compound (100 mg).
Step 2: tert-butyl (7-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)heptyl)carbamate BocHN
Figure imgf000225_0001
To a mixture of tert-butyl N-[7-[2-(2,6-dioxo-3-piperidyl)-l,3-dioxo-isoindolin-5-yl]hept-6- ynyl] carbamate (50 mg, 0.11 mmol) in ethanol (1 mL) was added Pd/C (10 mg) and the resulting mixture was stirred for 18 h under H2 atmosphere. The mixture was filtered, and the filtrate was concentrated under reduced pressure to afford the title compound (50 mg).
Step 3: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (7-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)heptyl)carbamate
Figure imgf000225_0002
The title compound was prepared by proceeding analogously as described in Example 8, Steps 2 and 3, using tert-butyl (7-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)heptyl) carbamate instead of tert-butyl (3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-3- oxopropyl)(methyl)carbamate in Step 2. MS (ES, m/z): [M+H]+ = 1022.3.
Example 21
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-
1 H-pyrrolizin-3 -yl)methyl 4-(3 -(2 ,4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-6-yl)-
Figure imgf000225_0003
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using l-(l-methyl-6-(piperidin-4-yl)-lH-indazol-3-yl)dihydropyrimidine-2,4(lH,3H)- dione hydrochloride (ref: WO2021/127561) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)- ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione in Step 11. MS (ES, m/z): [M+H]+ = 979.3.
Example 22
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)- amino)propyl)(methyl)carbamate
Figure imgf000226_0001
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 2-(2,6-dioxopiperidin-3-yl)-4-((3-(methylamino)propyl)amino)isoindoline-l, 3-dione (ref: WO2022/147465) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)ethyl)amino)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l, 3-dione in Step 11. MS (ES, m/z): [M+H]+ = 995.3.
Example 23
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl) amino)propyl) carbamate
Figure imgf000226_0002
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 4-((3-aminopropyl)amino)-2-(2,6-dioxopiperidin-3-yl) isoindoline- 1,3-dione (ref: J. Med. Chem, 2019, 62, 448 - 466) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)ethyl)amino)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l, 3-dione in Step 11. MS (ES, m/z): [M+H]+ = 981.3.
Example 24
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-
1 H-pyrrolizin-3 -yl)methyl 4-(4-(2,4-dioxotetrahydropyrimidin- 1 (2H)-yl)phenyl)piperazine- 1 - carboxylate
Figure imgf000227_0001
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using l-(4-(piperazin-l-yl)phenyl)dihydropyrimidine-2,4(lH,3H)-dione (ref: WO2022/147465) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)-ethyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline- 1,3-dione in Step 11. MS (ES, m/z): [M+H]+ = 926.3
Example 25
Synthesis of ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5- yl)amino)propyl)piperidine- 1 -carboxylate
Figure imgf000227_0002
Step 1: tert-butyl 4-(3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)propyl)- piperidine- 1 -carboxylate
Figure imgf000228_0001
A mixture of 2-(2,6-dioxo-3-piperidyl)-5-fluoro-isoindoline-l, 3-dione (138 mg, 0.5 mmol), tertbutyl 4-(3-aminopropyl)piperidine-l -carboxylate (121 mg, 0.5 mmol) and N,N-diisopropyl ethylamine (194 mg, 1.5 mmol) in 1 mL of DMF was stirred at 105 °C for 12 h. After cooling to rt, the reaction mixture was diluted with water and precipitates were isolated by filtration. The precipitates was dissolved in DCM, dried over Na2SO4 and concentrated in vacuum. The residue was purified by column chromatography, eluted with EtOAc/PE = 0-50% to afford the title compound (130 mg). Step 2: ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5- yl)amino)propyl)piperidine- 1 -carboxylate
Figure imgf000228_0002
The title compound was prepared by proceeding analogously as described in Example 8, Steps 2 and 3, using tert-butyl 4-(3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)- propyl)piperidine-l -carboxylate instead of tert-butyl (3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo- isoindolin-5-yl)amino)-3-oxopropyl)(methyl)carbamate in Step 2. MS (ES, m/z): [M+H]+ = 1049.3.
Example 26
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(3-(2,4-dioxotetrahydropyrimidin-l(2H)-yl)-l -methyl- lH-indazol-6- yl)piperazine- 1 -carboxylate
Figure imgf000229_0001
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using l-(l-methyl-6-(piperazin-l-yl)-lH-indazol-3-yl)dihydropyrimidine-2,4(lH,3H)- dione (ref: WO2021/127561) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)-ethyl)amino)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l, 3-dione in Step 11. MS (ES, m/z): [M+H]+ = 980.4.
Example 27
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)ethyl)piperazine- 1 -carboxylate
Figure imgf000229_0002
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 2-(2,6-dioxopiperidin-3-yl)-4-((2-(piperazin-l-yl)ethyl)amino)isoindoline- 1,3-dione (ref: J. Med. Chem, 2021, 64, 1626 - 1648) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)-ethyl)amino)- 2-(2,6-dioxopiperidin-3-yl) isoindoline- 1,3-dione in Step 11. MS (ES, m/z): [M+H]+ = 1020.4.
Example 28
Synthesis of ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)- amino)propyl)piperidine- 1 -carboxylate
Figure imgf000230_0001
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 2-(2,6-dioxopiperidin-3-yl)-4-((3-(piperidin-4-yl)propyl) amino)isoindoline- 1,3- dione (ref: J. Med. Chem, 2021, 64, 1626 - 1648) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)- ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione in Step 11. MS (ES, m/z): [M+H]+ = 1049.3.
Example 29
Synthesis of ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 3-(((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)methyl)pyrrolidine- 1 -carboxylate
Figure imgf000230_0002
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 2-(2,6-dioxopiperidin-3-yl)-4-((pyrrolidin-3-ylmethyl)amino)isoindoline-l, 3-dione (ref: W02017/197051) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)-ethyl)amino)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l, 3-dione in Step 11. MS (ES, m/z): [M+H]+ = 1007.3.
Example 30
Synthesis of ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen- 1 -yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy) methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 3-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)- amino)ethyl)azetidine- 1 -carboxylate
Figure imgf000231_0001
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 4-((2-(azetidin-3-yl)ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l, 3-dione, (ref: W02021/127190) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)-ethyl)amino)-2-(2,6-dioxo- piperidin-3-yl)isoindoline-l,3-dione in Step 11. MS (ES, m/z): [M+H]+ = 1007.3.
Example 31
Synthesis of ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen- 1 -yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methyl enehexahydro- lH-pyrrolizin-3-yl)methyl 3-(((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5- yl)amino)methyl)pyrrolidine- 1 -carboxylate
Figure imgf000231_0002
Step 1: tert-butyl 3-(((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)methyl)- pyrrolidine- 1 -carboxylate
Figure imgf000231_0003
A solution of 2-(2,6-dioxo-3-piperidyl)-5-fluoroisoindoline-l, 3-dione (138 mg, 0.5 mmol), tert-butyl 3-(aminomethyl)pyrrolidine-l -carboxylate (100 mg, 0.5 mmol), N,N-diisopropyl ethylamine (194 mg, 1.5 mmol) in 1 mL of DMF was stirred at 105 °C for 12 h. After cooling to rt, the reaction mixture was diluted with water, extracted by DCM, dried over Na2SO4 and concentrated. The residue was purified by silica gel column chromatography, eluted with EtOAc/PE = 0-60% to afford the title compound (100 mg).
Step 2: ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methyl enehexahydro- lH-pyrrolizin-3-yl)methyl 3-(((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)- methyl)pyrrolidine- 1 -carboxylate
Figure imgf000232_0001
The title compound was prepared by proceeding analogously as described in Example 8, Steps 2 and 3, using tert-butyl 3-(((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)methyl)- pyrro li dine- 1 -carboxylate instead of tert-butyl (3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5- yl)amino)-3-oxopropyl)(methyl)carbamate in Step 2. MS (ES, m/z): [M+H]+ = 1007.3.
Example 32
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)amino)-2- oxoethyl)piperidine- 1 -carboxylate
Figure imgf000232_0002
Step 1 : tert-butyl 4-(2-((2-(2,6-dioxopiperidin-3-yl)-l ,3-dioxoisoindolin-4-yl)amino)-2-oxoethyl)- piperidine- 1 -carboxylate
Figure imgf000233_0001
To a solution of 4-amino-2-(2,6-dioxo-3-piperidyl)isoindoline- 1,3 -dione (200 mg, 0.73 mmol) and 2-( 1 -tert-butoxycarbonyl-4-piperidyl)acetic acid (213.69 mg, 0.88 mmol) in anhydrous DMF (3 ml) was added N,N-dimethylpyridin-2-amine (268.26 mg, 2.2 mmol), followed by l-chloro-N,N,2- trimethylprop- 1 -en- 1 -amine (195.6 mg, 1.46 mmol) and the resulting mixture was stirred at 25 °C for 1 h. The reaction mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with water, brine, dried over anhydrous sodium sulphate and concentrated under reduced pressure. The residue was purified by Prep-HPLC (0-70% acetonitrile in water with 0.05% formic acid) to afford the title compound (100 mg).
Step 2: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(2-((2-(2,6-dioxopiperi din-3 -yl)- 1,3 -di oxoisoindolin-4-yl)amino)-2- oxoethyl)piperidine- 1 -carboxylate
Figure imgf000233_0002
The title compound was prepared by proceeding analogously as described in Example 8, Steps 2 and 3, using tert-butyl 4-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)amino)-2- oxoethyl)piperidine-l -carboxylate instead of tert-butyl (3-((2-(2,6-dioxopiperidin-3-yl)-l,3- dioxoisoindolin-5-yl)amino)-3-oxopropyl)(methyl)carbamate in Step 2. MS (ES, m/z): [M+H]+ = 1049.4
Example 33
Synthesis of ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen- 1 -yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy) methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)carbamoyl)morpholine-4-carboxylate
Figure imgf000234_0001
The title compound was prepared by proceeding analogously as described in Example 32, Steps 1 and 2, using 4-tert-butoxycarbonylmorpholine-2-carboxylic acid instead of 2-(l-tert- butoxycarbonyl-4-piperidyl)acetic acid in Step 1. MS (ES, m/z): [M+H]+ = 1023.3.
Example 34
Synthesis of ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl (4-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)oxy)butyl)carbamate
Figure imgf000234_0002
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 3 -(4-(4-aminobutoxy)-l-oxoisoindolin-2-yl)piperidine-2, 6-dione (ref: WO2022/148459) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)ethyl)amino)-2-(2,6-dioxopiperi din-3 - yl)isoindoline- 1,3-dione in Step 11. MS (ES, m/z): [M+H]+ = 982.3.
Example 35
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-
1 H-pyrrolizin-3 -yl)methyl 4-(2-(2 ,6-dioxopiperidin-3 -yl)- 1 -oxoisoindolin-5-yl)piperidine- 1 - carboxylate
Figure imgf000235_0001
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 3-(l-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2, 6-dione (ref: WO2021/87093) instead of 4-((2-(2-(2-aminoethoxy)ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline- 1,3-dione in Step 11. MS (ES, m/z): [M+H]+ = 978.4.
Example 36
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-
1 H-pyrrolizin-3 -yl)methyl 4-(2-(2 ,6-dioxopiperidin-3 -yl)- 1 -oxoisoindolin-4-yl)piperidine- 1 - carboxylate
Figure imgf000235_0002
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using 3-(l-oxo-4-(piperidin-4-yl)isoindolin-2-yl)piperidine-2, 6-dione (ref: WO2017/197056) instead of 4-((2-(2-(2-aminoethoxy) ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline- 1,3-dione in Step 11. MS (ES, m/z): [M+H]+ = 978.4.
Example 37
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-
1 H-pyrrolizin-3 -yl)methyl 4-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-2- oxoethyl)piperidine- 1 -carboxylate
Figure imgf000236_0001
The title compound was prepared by proceeding analogously as described in Example 8, Steps 1-3, using 2-(l-tert-butoxycarbonyl-4-piperidyl)acetic acid instead of 3-[tert-butoxycarbonyl(methyl)- amino]propanoic acid in Step 1. MS (ES, m/z): [M+H]+ = 1049.4.
Example 38
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl(4-((2-(2,6-dioxopiperidin-3-yl)-l -oxoisoindo lin-5-yl)oxy)butyl)carbamate
Figure imgf000236_0002
Step 1: tert-butyl (4-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-5-yl)oxy)butyl)carbamate
Figure imgf000236_0003
A mixture of 3-(5-hydroxy-l-oxoisoindolin-2-yl)piperidine-2, 6-dione (260 mg, 1 mmol), tertbutyl (4-bromobutyl)carbamate (252 mg, 1 mmol) and K2CO3 (207 mg, 1.5 mmol) in ACN (2 mL) was stirred at 85 °C for 12 h under argon atmosphere. After cooling to rt, the reaction mixture was filtered and the filtrate was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with MeOH/DCM = 0-8% to yield the title compound (80 mg).
Step 2: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl(4-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-5-yl)oxy)butyl)carbamate
Figure imgf000237_0001
The title compound was prepared by proceeding analogously as described in Example 8, Steps 2 and 3, using tert-butyl (4-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-5-yl)oxy)butyl)carbamate instead of tert-butyl (3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-3- oxopropyl)(methyl)carbamate in Step 2. MS (ES, m/z): [M+H]+ = 982.4.
Example 39
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl(2-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-5-yl)oxy)ethyl)carbamate
Figure imgf000237_0002
Step 1: tert-butyl (2-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-5-yl)oxy)ethyl)carbamate
Figure imgf000237_0003
A mixture of 3-(5-hydroxy-l-oxoisoindolin-2-yl)piperidine-2, 6-dione (65 mg, 0.25 mmol), tert-butyl (2-bromoethyl)carbamate (63 mg, 0.25 mmol) and K2CO3 (52 mg, 0.37 mmol) in ACN (2 mL) was stirred at 85 °C for 12 h under argon atmosphere. The reaction mixture was filtered, and the filtrate was concentrated in vacuum. The residue was purified by silica gel chromatography, eluted with MeOH/DCM = 0-8% to afford the title compound (60 mg).
Step 2: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl(2-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-5-yl)oxy)ethyl)carbamate
Figure imgf000238_0001
The title compound was prepared by proceeding analogously as described in Example 8, Steps 2 and 3, using tert-butyl (2-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-5-yl)oxy)ethyl)carbamate instead of tert-butyl (3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-3- oxopropyl)(methyl)carbamate in Step 2. MS (ES, m/z): [M+H]+ = 954.3.
Example 40
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen- 1 -yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy) methyl)-6-methylenehexahydro-
1 H-pyrrolizin-3 -yl)methyl( 1 -((4-((4-(3-(2,4-dioxo tetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H- indazol-6-yl)piperidin- 1 -yl)methyl) phenyl)sulfonyl)piperidin-4-yl)carbamate
Figure imgf000238_0002
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using l-(6-(l-(3-((4-aminopiperidin-l-yl)sulfonyl)benzyl) piperidin-4-yl)- 1 -methyl- 1 H- indazol-3-yl)dihydropyrimidine-2,4(lH,3H)-dione instead of 4-((2-(2-(2- aminoethoxy)ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione in Step 11. MS (ES, m/z): [M+H]+ = 1230.5.
Example 41
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen- l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)oxy)ethyl)piperidine- 1 -carboxylate
Figure imgf000239_0001
Step 1: tert-butyl 4-(2-(tosyloxy)ethyl)piperidine-l -carboxylate
Figure imgf000239_0002
To a solution of tert-butyl 4-(2 -hydroxyethyl)piperi dine- 1 -carboxylate (500 mg, 2.18 mmol) and
4-methylbenzenesulfonyl chloride (623.5 mg, 3.27 mmol) in anhydrous DCM (10 mL) was added 4- dimethylaminopyridine (266.37 mg, 2.18 mmol) at rt and the resulting mixture was stirred at this temperature for 18 h. The reaction mixture was quenched with 1.0 M hydrogen chloride aq. solution and then extracted with EtOAc. The combined organic layers were washed with 10% sodium hydroxide, water, brine, dried over anhydrous sodium sulphate and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluted with EtOAc/petroleum (0~20%) to afford the title compound (490 mg).
Step 2: tert-butyl 4-(2-((2-(2,6-dioxopiperi din-3 -yl)- 1,3 -di oxoisoindolin-4-yl)oxy) ethyl)piperidine-l- carboxylate
Figure imgf000239_0003
To a stirred solution of 2-(2,6-dioxopiperidin-3-yl)-4-hydroxyisoindoline-l, 3-dione (178.77 mg, 0.65 mmol) and tert-butyl 4-(2-(tosyloxy)ethyl)piperi dine- 1 -carboxylate (250 mg, 0.65 mmol) in anhydrous DMF (3 mL) was added sodium carbonate (138.19 mg, 1.3 mmol) and the resulting mixture was stirred at 80 °C for 16 h under argon atmosphere. After cooling to rt, the reaction mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with water, brine, dried over anhydrous sodium sulphate and concentrated. The residue was purified by silica gel chromatography, eluted with methanol/di chloromethane (0~3%) to afford the title compound (250 mg).
Step 3: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(2-((2-(2,6-dioxopiperi din-3 -yl)- 1,3 -di oxoisoindolin-4-yl)oxy)- ethyl)piperidine- 1 -carboxylate
Figure imgf000240_0001
The title compound was prepared by proceeding analogously as described in Example 8, Steps 2 and 3, using tert-butyl 4-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)oxy) ethyl)piperidine-l -carboxylate instead of tert-butyl (3-((2-(2,6-dioxopiperidin-3-yl)-l,3- dioxoisoindolin-5-yl)amino)-3-oxopropyl)(methyl)carbamate in Step 2. MS (ES, m/z): [M+H]+ = 1036.4.
Example 42
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(2-((2-(2,6-dioxo piperidin-3-yl)-l ,3-dioxoisoindolin-5- yl)oxy)ethyl)piperidine- 1 -carboxylate
Figure imgf000240_0002
The title compound was prepared by proceeding analogously as described in Example 41, Steps 2-3, using 2-(2,6-dioxopiperidin-3-yl)-5-hydroxyisoindoline-l, 3-dione instead of 2-(2,6- dioxopiperidin-3-yl)-4-hydroxyisoindoline-l, 3-dione in step 2. MS (ES, m/z): [M+H]+ = 1036.5. Example 43
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen- 1 -yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy) methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 2-(((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)oxy)methyl)morpholine-4-carboxylate
Figure imgf000241_0001
Step 1: tert-butyl 2-(((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)oxy) methyl)morpholine- 4-carboxylate
Figure imgf000241_0002
To a solution of tert-butyl 2-(hydroxymethyl)morpholine-4-carboxylate (109 mg, 0.5 mmol), 2-(2,6-dioxopiperidin-3-yl)-4-hydroxyisoindoline- 1,3-dione (165 mg, 0.6 mmol) and PPI13 (158 mg, 0.6 mmol) in anhydrous THF (2 mL), was added DIAD (122 mg, 0.6 mmol) dropwise at 0 °C. The resulting mixture was stirred for 12 h at rt. The reaction mixture was quenched by water, extracted by DCM, dried over Na2SO4, and concentrated in vacuum. The residue was purified by flash, eluted with MeOH/DCM = 0-6% to yield the title compound (60 mg, 25%) as a yellow oil.
Step 2: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy) methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 2-(((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)oxy)methyl)morpholine-4-carboxylate
Figure imgf000242_0001
The title compound was prepared by proceeding analogously as described in Example 8, Steps 2 and 3, using tert-butyl 2-(((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)oxy) methyl)morpholine-4-carboxylate instead of tert-butyl (3-((2-(2,6-dioxopiperidin-3-yl)-l,3- dioxoisoindolin-5-yl)amino)-3-oxopropyl)(methyl)carbamate in Step 2. MS (ES, m/z): [M+H]+ = 1024.3.
Example 44
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-((2-(2,6-dioxopiperidin-3-yl)-3-oxoisoindolin-4-yl)ethynyl)piperidine-l- carboxylate
Figure imgf000242_0002
Step 1: tert-butyl 4-((2-(2,6-dioxopiperidin-3-yl)-3 -oxoisoindo lin-4-yl)ethynyl)piperi dine- 1- carboxylate
Figure imgf000242_0003
A mixture of 3-(7-bromo-l-oxoisoindolin-2-yl)piperidine-2, 6-dione (323 mg, 1 mmol), tertbutyl 4-ethynylpiperidine-l -carboxylate (418 mg, 2 mmol), Cui (38 mg, 0.2 mmol), Pd(PPh3)3C12 (70.1 mg, 0.1 mmol) and TEA (1 mL) in DMF (2 mL) was stirred at 85 °C for 3 h under argon atmosphere. After cooling to rt, the reaction mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2SO4, and concentrated in vacuum. The residue was purified by flash, eluted with MeOH/DCM = 0-6% to yield the title compound (260 mg). Step 2: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8- ethynyl-7-fluoro-3-hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6- methylenehexahydro-lH-pyrrolizin-3-yl)methyl 4-((2-(2,6-dioxopiperidin-3-yl)-3-oxoisoindolin-4- yl)ethynyl)piperidine- 1 -carboxylate
Figure imgf000243_0001
The title compound was prepared by proceeding analogously as described in Example 8, Steps 2 and 3, using tert-butyl 4-((2-(2,6-dioxopiperidin-3-yl)-3-oxoisoindolin-4-yl)ethynyl)piperidine-l- carboxylate instead of tert-butyl (3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-3- oxopropyl)(methyl)carbamate in Step 2. MS (ES, m/z): [M+H]+ = 1002.4.
Example 45
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(4-((2-(2,6-dioxopiperidin-3-yl)-3-oxoisoindolin-4-yl)carbamoyl)-lH- 1 ,2,3-triazol- 1 -yl) piperidine- 1 -carboxylate
Figure imgf000243_0002
The title compound was prepared by proceeding analogously as described in Example 32, Steps 1 and 2, using l-(l-(tert-butoxycarbonyl)piperidin-4-yl)-lH-l,2,3-triazole-4-carboxylic acid instead of 2-(l-tert-butoxycarbonyl-4-piperidyl)acetic acid and 3-(7-amino-l-oxoisoindolin-2- yl)piperidine-2, 6-dione instead of 4-amino-2-(2,6-dioxo-3-piperidyl)isoindoline-l,3-dione in Step 1. MS (ES, m/z): [M+H]+ = 1088.3. Example 46
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(2-(2-(2,6-dioxopiperidin-3-yl)-3-oxoisoindolin-4-yl)ethyl)piperidine-l-
Figure imgf000244_0001
Step 1: tert-butyl 4-(2-(2-(2,6-dioxopiperidin-3-yl)-3-oxoisoindolin-4-yl)ethyl)piperidine-l- carboxylate
Figure imgf000244_0002
To a solution of tert-butyl 4-((2-(2,6-dioxopiperidin-3-yl)-3-oxoisoindolin-4-yl)ethynyl)- piperidine- 1 -carboxylate (200 mg, 0.44 mmol) in EtOAc (2 mL) was added ammonium formate (140 mg, 2.2 mmol) and Pd/C (40 mg). The reaction was stirred for 3 h at 25 °C under hydrogen atmosphere. The reaction mixture was diluted with EtOAc and filtered. The filtrate was washed with water, brine, dried over Na2SO4 and concentrated. The residue was purified by silica gel column chromatography, eluted with MeOH/DCM = 0-6%, to afford the title compound (160 mg). Step 2: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(2-(2-(2,6-dioxopiperidin-3-yl)-3-oxoisoindolin-4-yl)ethyl)piperidine-l- carboxylate
Figure imgf000244_0003
The title compound was prepared by proceeding analogously as described in Example 8, Steps 2 and 3, using tert-butyl 4-(2-(2-(2,6-dioxopiperidin-3-yl)-3-oxoisoindolin-4-yl)ethyl)piperidine-l- carboxylate instead of tert-butyl (3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-3- oxopropyl)(methyl)carbamate in Step 2 MS (ES, m/z): [M+H]+ = 1006.3.
Example 47
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-((l-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)piperidin-4- yl)oxy)piperidine- 1 -carboxylate
Figure imgf000245_0001
Step 1 : tert-butyl 4- [ [ 1 - [2-(2,6-dioxo-3 -piperidyl)- 1 ,3 -dioxo-isoindolin-5-yl] -4-piperidyl] oxy] - piperidine- 1 -carboxylate
Figure imgf000245_0002
To a solution of tert-butyl 4-(4-piperidyloxy)piperidine- 1 -carboxylate (340 mg, 1.19 mmol, 1.1 eq) and 2-(2,6-dioxo-3-piperidyl)-5-fluoro-isoindoline- 1,3-dione (300 mg, 1.09 mmol, 1.0 eq) in anhydrous NMP (10 mL), was added N,N-diisopropyl ethylamine (0.28 mL, 1.63 mmol, 1.5 eq) and the resulting mixture was stirred at 130 °C for 3 h. After cooling to rt, the reaction mixture was diluted with water, then extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with MeOH/DCM (0-3%) to afford the title compound (400 mg).
Step 2: 2-(2,6-dioxo-3-piperidyl)-5-[4-(4-piperidyloxy)-l-piperidyl]isoindoline- 1,3-dione hydrochloride salt
Figure imgf000245_0003
To a solution of tert-butyl 4-[[l-[2-(2,6-dioxo-3-piperidyl)-l,3-dioxo-isoindolin-5-yl]-4- piperidyl]oxy]piperidine-l -carboxylate (400 mg, 0.74 mmol, 1.0 eq) in anhydrous DCM (20 mL) at 25 °C was added HC1 solution in 1,4-dioxane (4.0 M, 3.7 mL, 14.8 mmol, 20.0 eq) and the resulting mixture was stirred at 25 °C for 2 h. The reaction mixture was concentrated under reduced pressure and the residue was recrystallized from EtOAc to afford the title compound as HC1 salt (300 mg). Step 3: 8-fluoro-7-(7-fluoro-8-((triisopropylsilyl)ethynyl)naphthalen-l-yl)pyrido[4,3-d]-pyrimidine-
2,4-diol
Figure imgf000246_0001
To a solution of 7-chloro-8-fluoropyrido[4,3-d]pyrimidine-2,4-diol (4.0 g, 18.6 mmol, 1.0 eq.) and ((2-fluoro-8-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)naphthalen-l-yl)ethynyl)- triisopropylsilane (10.9 g, 24.1 mmol, 1.3 eq.) in EtOH (150 mL) and H2O (50 mL) was added CATACXIUM A Pd G3 (2.4 g, 3.3 mmol, 0.18 eq.), K3PO4 (11.7 g, 55.1 mmol, 3.0 eq.) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 80 °C under nitrogen atmosphere. After cooling to rt, the reaction mixture was diluted with water, extracted with EtOAc. The combined organic layers were washed with water, brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by silica gel column chromatography, eluted with MeOH/DCM (0-10%), to afford the title compound (5.5 g).
Step 4: 7-(8-ethynyl-7-fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidine-2,4-diol
Figure imgf000246_0002
A mixture of 8-fluoro-7-(7-fluoro-8-((triisopropylsilyl)ethynyl)naphthalen-l-yl)pyrido [4,3- d]pyrimidine-2,4-diol (10.0 g, 19.8 mmol, 1.0 eq.) and CsF (12.0 g, 79.0 mmol, 4.0 eq.) in DMF (100 mL) was stirred for 1 h at 50 °C under nitrogen atmosphere. After cooling to rt, the reaction mixture was concentrated and the residue was purified by silica gel column chromatography, eluted with MeOH/DCM (0-10%), to afford the title compound (6.0 g).
Step 5: 2,4-dichloro-7-(8-ethynyl-7-fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidine
Figure imgf000247_0001
To a solution of POCh (15.8 g, 103.0 mmol, 30.3 eq.) and DIPEA (13.4 g, 103.7 mmol, 30.5 eq.) was added 7-(8-ethynyl-7-fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]-pyrimidine-2,4-diol (1.2 g, 3.4 mmol, 1.0 eq.) in portions at 0-5 °C and the resulting mixture was stirred at 50 °C for 1 h. After cooling to rt, the reaction mixture was concentrated and the residue was diluted with ice water and extracted with EtOAc. The combined organic layers were washed with water, brine, dried over anhydrous Na2SO4 and concentrated to afford the title compound (1.5 g).
Step 6: tert-butyl (lR,5S)-3-(2-chloro-7-(8-ethynyl-7-fluoronaphthalen-l-yl)-8-fluoropyrido[4,3- d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate
Figure imgf000247_0002
To a solution of 2,4-dichloro-7-(8-ethynyl-7-fluoronaphthalen-l-yl)-8-fluoropyrido-[4,3-d]- pyrimidine (1.5 g, 3.9 mmol, 1.0 eq., crude) in DCM (30 mL) was added DIEA (1.1 g, 8.5 mmol, 2.2 eq.) dropwise at -40 °C and the resulting solution was stirred at -40 °C for 5 mins. A solution of tertbutyl (lR,5S)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate (870 mg, 4.1 mmol, 1.1 eq.) in THF (5 mL) was added to the above reaction mixture dropwise and the resulting mixture was stirred for additional 15 mins at -40 °C. The reaction mixture was concentrated and the residue was purified by silica gel column chromatography, eluted with EA/PE (0-50%), to afford the title compound (770 mg).
Step 7: tert-butyl (lR,5S)-3-(2-(((5S,7aS)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2-methylene- tetrahydro-lH-pyrrolizin-7a(5H)-yl)methoxy)-7-(8-ethynyl-7-fluoro naphthalen-l-yl)-8- fluoropyrido[4,3-d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate
Figure imgf000247_0003
To a solution of ((5R,7aR)-5-(((tert-butyldiphenylsilyl)oxy)methyl)-2-methylenetetrahydro- lH-pyrrolizin-7a(5H)-yl)methanol (1 -.65 g, 3.9 mmol, 1.1 eq) in THF (20 mL) was added NaH (427 mg, 10.7 mmol, 60% purity) at 0 °C and the resulting mixture was stirred at 0 °C for 30 min. Tertbutyl (lR,5S)-3-(2-chloro-7-(8-ethynyl-7-fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-4- yl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate (2.0 g, 3.6 mmol, 1.0 eq) was added to the above mixture at 0 °C and the the mixture was stirred at 25 °C for 3 h. The reaction mixture was quenched by NH4CI (sat aq.) at 0 °C, and then extracted with EtOAc. The combined organic layers were dried over Na2SO4 and concentrated. The residue was purified by silica gel column chromatography, eluted with MeOH/DCM (0-5%), to give the title compound (3.0 g).
Step 8: tert-butyl (lR,5S)-3-(7-(8-ethynyl-7-fluoronaphthalen-l-yl)-8-fluoro-2-(((5S,7aS)-5- (hydroxymethyl)-2-methylenetetrahydro-lH-pyrrolizin-7a(5H)-yl) methoxy)pyrido[4,3-d]pyrimidin- 4-yl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate
Figure imgf000248_0001
To a solution of tert-butyl (lR,5S)-3-(2-(((5S,7aS)-5-(((tert-butyldiphenylsilyl) oxy)methyl)- 2 -methylenetetrahydro- 1 H-pyrrolizin-7a(5H)-yl)methoxy)-7-(8-ethynyl-7-fluoronaphthalen- 1 -yl)-8- fluoropyrido[4,3-d] pyramidin-4-yl)-3,8-diazabicyclo [3.2.1]octane-8-carboxylate (3.0 g, 3.2 mmol, 1.0 eq) in THF (10 mL) was added TBAF THF solution (1.0 M, 12.7 mL, 4.0 eq) and the resulting mixture was stirred at 25 °C for 2 hours. The mixture was then concentrated, and the residue was purified by silica gel column chromatography, eluted with MeOH/DCM (0-5%) to give the title compound (2.0 g).
Step 9: tert-butyl (lR,5S)-3-(7-(8-ethynyl-7-fluoronaphthalen-l-yl)-8-fluoro-2-(((5S,7aS)-2- methylene-5-((((4-nitrophenoxy)carbonyl)oxy)methyl)tetrahydro-lH-pyrrolizin-7a(5H)- yl)methoxy)pyrido[4,3-d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1] octane-8-carboxylate
Figure imgf000248_0002
To a solution of TEA (0.31 mL, 2.26 mmol) and tert-butyl (lR,5S)-3-(7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoro-2-(((5S,7aS)-5-(hydroxymethyl)-2-methylene tetrahydro-lH- pyrrolizin-7a(5H)-yl)methoxy)pyrido[4,3-d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1]octane-8- carboxylate (200 mg, 0.28 mmol) in anhydrous THF (1 mL) was added (4-nitrophenyl) carbonochloridate (199 mg, 0.99 mmol) at 25 °C and the resulting mixture was stirred at 25 °C for 16 h under Ar atmosphere. The reaction mixture was diluted with water, extracted with EtOAc, washed with brine, dried over anhydrous Na2SO4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with EA/PE (0-50%) to afford the title compound (130 mg).
Step 10: tert-butyl (lR,5S)-3-(2-(((5S,7aS)-5-(((4-((l-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo- isoindolin-5-yl)piperidin-4-yl)oxy)piperidine-l-carbonyl)oxy)methyl)-2-methylenetetrahydro-lH- pyrrolizin-7a(5H)-yl)methoxy)-7-(8-ethynyl-7-fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]- pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1] octane-8-carboxylate
Figure imgf000249_0001
To a solution of 2-(2,6-dioxo-3-piperidyl)-5-[4-(4-piperidyloxy)-l-piperidyl]isoindoline-l,3- dione (HC1 salt, 142 mg, 0.3 mmol), TEA (151 mg, 1.49 mmol) in anhydrous THF (4.5 mL) and NMP (1.5 mL) was added tert-butyl (lR,5S)-3-(7-(8-ethynyl-7-fluoronaphthalen-l-yl)-8-fluoro-2- (((5S,7aS)-2-methylene-5-((((4-nitrophenoxy)carbonyl)oxy)methyl)tetrahydro-lH-pyrrolizin-7a (5H)-yl)methoxy)pyrido[4,3-d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate (130 mg, 0.1488 mmol) at 25 °C and the resulting mixture was stirred at 25 °C for 16 h. To the reaction mixture was added water and saturated NaHC'CF, at 25 °C. The resulting mixture was stirred at 25 °C for 10 min. The mixture was extracted with EtOAc, washed with brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by silica gel column chromatography, eluted with MeOH/DCM (0-5%) to afford the title compound (122 mg).
Step 11: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen- 1 -yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- 1 H- pyrrolizin-3-yl)methyl 4-((l-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)piperidin-4-yl)- oxy)piperidine- 1 -carboxylate
Figure imgf000250_0001
To a solution of tert-butyl (lR,5S)-3-(2-(((5S,7aS)-5-(((4-((l-(2-(2,6-dioxopiperidin-3-yl)-l,3- dioxoisoindolin-5-yl)piperidin-4-yl)oxy)piperidine-l-carbonyl)oxy)methyl)-2-methylenetetrahydro- lH-pyrrolizin-7a(5H)-yl)methoxy)-7-(8-ethynyl-7-fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]- pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate (122 mg, 0.1038 mmol) in anhydrous DCM (10 mL) at -10 °C was added HCl/l,4-di oxane (0.52 mL, 2.08 mmol) dropwise and the resulting mixture was stirred at 25 °C for 2 h. The reaction mixture was then concentrated under reduced pressure. The residue was diluted with DCM (10 mL) and basified with NHs/MeOH (0.5 mL) at -78 °C. The resulting mixture was concentrated under reduced pressure and the residue was purified by Cl 8 column chromatography, eluted with MeCN/water (0-60%, 0.05% NH4HCO3) to afford the title compound (70.5 mg). MS (ES, m/z): [M+H]+ = 1075.4.
Example 48
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-(l-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)piperidine-4- carbonyl)piperazine- 1 -carboxylate
Figure imgf000250_0002
Step 1: tert-butyl 4-[l-[2-(2,6-dioxo-3-piperidyl)-l,3-dioxo-isoindolin-5-yl]piperidine-4- carbonyl]piperazine- 1 -carboxylate
Figure imgf000251_0001
To a solution of tert-butyl 4-(piperidine-4-carbonyl)piperazine- 1 -carboxylate (592 mg, 1.99 mmol) and 2-(2,6-dioxo-3-piperidyl)-5-fluoro-isoindoline-l,3-dione (500 mg, 1.81 mmol) in anhydrous NMP (10 mL) was added N,N-diisopropylethylamine (0.47 mL, 2.72 mmol) at 25 °C and the resulting mixture was stirred at 130 °C for 3 h under Ar atmoshpere. After cooling to rt, the reaction mixture was diluted with water, extracted with EtOAc, washed with brine, dried over anhydrous Na2SO4, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with EA/PE (0-80%) to afford the title compound (800 mg).
Step 2: 2-(2,6-dioxo-3-piperidyl)-5-[4-(piperazine-l -carbonyl)-! -piperidyl] isoindoline- 1,3-dione
Figure imgf000251_0002
To a solution of tert-butyl 4-[l-[2-(2,6-dioxo-3-piperidyl)-l,3-dioxo-isoindolin-5-yl]- piperidine-4-carbonyl]piperazine- 1 -carboxylate (800 mg, 1.45 mmol) in anhydrous DCM (20 mL) was added HC1 solution in 1,4-dioxane (7.23 mL, 28.9 mmol, 20.0 eq) at 25 °C dropwise and the resulting mixture was stirred at 25 °C for 2 h. The reaction mixture was then concentrated under reduced pressure and the residue was recrystallized from EtOAc to afford the title compound as HC1 salt (650 mg).
Step 3: tert-butyl (lR,5S)-3-(2-(((5S,7aS)-5-(((4-(l-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin- 5 -yl)piperidine-4-carbonyl)piperazine- 1 -carbonyl)oxy)methyl)-2-methylenetetrahydro- 1 H-pyrrolizin- 7a(5H)-yl)methoxy)-7-(8-ethynyl-7-fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-4-yl)-3,8- diazabicyclo[3.2.1] octane-8-carboxylate
Figure imgf000251_0003
To a solution of 2-(2,6-dioxo-3-piperidyl)-5-[4-(piperazine-l-carbonyl)-l-piperidyl]- isoindoline- 1,3-dione (HC1 salt, 146 mg, 0.3 mmol), TEA (151 mg, 1.49 mmol) in anhydrous NMP (1.5 mL) was added a solution of tert-butyl (lR,5S)-3-(7-(8-ethynyl-7-fluoronaphthalen-l-yl)-8- fluoro-2-(((5S,7aS)-2-methylene-5-((((4-nitrophenoxy)carbonyl)oxy) methyl)tetrahydro-lH- pyrrolizin-7a(5H)-yl)methoxy)pyrido[4,3-d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1]octane-8- carboxylate (130 mg, 0.1488 mmol) in anhydrous THF (4.5 mL) at 25 °C and the resulting mixture was stirred at 25 °C for 16 h. The reaction mixture was then added water and saturated NaHCCh aq. solution at 25 °C. The resulting mixture was stirred at 25 °C for 10 min. The mixture was extracted with EtoAc, washed with brine, dried over anhydrous NA2SO4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with MeOH/DCM (0- 3%) to afford the title compound (110 mg).
Step 4: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen- 1 -yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- 1 H- pyrrolizin-3-yl)methyl 4-(l-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)piperidine-4- carbonyl)piperazine- 1 -carboxylate
Figure imgf000252_0001
To a solution of tert-butyl (lR,5S)-3-(2-(((5S,7aS)-5-(((4-(l-(2-(2,6-dioxopiperidin-3-yl)-l,3- dioxoisoindolin-5 -yl)piperidine-4-carbonyl)piperazine- 1 -carbonyl)oxy) methyl)-2-methylenetetra- hydro- 1 H-pyrrolizin-7 a(5H)-yl)methoxy)-7 -(8-ethynyl-7 -fluoronaphthalen- 1 -yl)-8 -fluoropyri do- 14, 3-d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1] octane-8-carboxylate (110 mg, 0.092 mmol) in anhydrous DCM (10 mL) was added HC1 solution in 1,4-dioxane (4.0 M, 1.5 mL, 6.0 mmol) at -10 °C and the resulting mixture was stirred at 25 °C for 1 h. The reaction mixture was then concentrated under reduced pressure and the residue was dissolved in DCM (10 mL). The mixture was basified by adding NH3/McOH (0.5 mL) at -78 °C, and the resulting mixture was concentrated under reduced pressure. The residue was purified by Cl 8 column chromatography, eluted with MeCN/water (0-60%, 0.05% NH4HCO3) to afford the title compound (60.1 mg). MS (ES, m/z): [M+H]+ = 1088.4. Example 49
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl (8-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-8- oxooctyl)carbamate
Figure imgf000253_0001
Step 1: tert-butyl (8-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl) amino)-8-oxooctyl)- carbamate
Figure imgf000253_0002
To a solution of 5-amino-2-(2,6-dioxo-3-piperidyl) isoindoline- 1,3 -di one (200 mg, 0.73 mmol) and 8-(tert-butoxycarbonylamino)octanoic acid (227.79 mg, 0.88 mmol) in anhydrous DMF (4 mL) was added NMI (420.64 mg, 5.12 mmol), followed by TCFH (616.1 mg, 2.2 mmol) at 25 °C under argon atmosphere and stirred for 3 h. The reaction mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with water, brine, dried over anhydrous sodium sulphate and concentrated under reduced pressure. The residue was purified by Prep-HPLC (0~70% acetonitrile in water with 0.05% formic acid) to afford the title compound (200 mg). Step 2 : ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen- 1 -yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylene hexahydro- lH-pyrrolizin-3-yl)methyl(8-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoiso indolin-5-yl)amino)-8- oxooctyl)carbamate
Figure imgf000253_0003
The title compound was prepared by proceeding analogously as described in Example 48, Steps 2-4, using tert-butyl (8-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-8- oxooctyl)carbamate instead of tert-butyl 4-[l-[2-(2,6-dioxo-3-piperidyl)-l,3-dioxo-isoindolin-5- yl]piperidine-4-carbonyl]piperazine- 1 -carboxylate in Step 2. MS (ES, m/z): [M+H]+ =1049.4.
Example 50
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3 -yl)methyl ( 1 -((4-((4-(3 -(2, 4-di oxotetrahydro pyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-
6-yl)piperidin- 1 -yl)methyl)phenyl)sulfonyl) piperidin-4-yl)carbamate
Figure imgf000254_0001
Step 1 : tert-butyl ( 1 -((3 -((4-(3 -(2,4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-6-yl)- piperidin-l-yl)methyl)phenyl)sulfonyl)piperidin-4-yl)carbamate
Figure imgf000254_0002
To a solution of l-(l-methyl-6-(piperidin-4-yl)-lH-indazol-3-yl)dihydro pyrimidine- 2,4(lH,3H)-dione (50 mg, 0.15 mmol, 1.00 eq.) and tert-butyl (l-((3-(bromomethyl)phenyl) sulfonyl)piperidin-4-yl)carbamate (99 mg, 0.23 mmol, 1.50 eq.) in THF (5.0 mL) was added TEA (45 mg, 0.45 mmol, 3.00 eq.) and the resulting mixture was stirred at 55 °C overnight. After cooling to rt, the reaction mixture was diluted with water and extracted with DCM. The combined organic layers were washed with brine, dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by flash chromatography (DCM:MeOH=20: 1) to give the title compound.
Step 2 : 1 -(6-( 1 -(3-((4-aminopiperidin- 1 -yl)sulfonyl)benzyl)piperidin-4-yl)- 1 -methyl- 1 H-indazol-3 - yl)dihydropyrimidine-2,4(lH,3H)-dione
Figure imgf000255_0001
A mixture of tert-butyl (l-((3-((4-(3-(2,4-dioxotetrahydropyrimidin-l(2H)-yl)-l-methyl-lH- indazol-6-yl)piperidin-l-yl)methyl)phenyl)sulfonyl)piperidin-4-yl)carbamate (50 mg, 0.07 mmol, 1.00 eq.) in TFA/DCM (0.5 mL/2.0 mL) was stirred at rt for 2 h. The mixture was concentrated to give the title compound (40 mg, crude).
Step 3: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro- naphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl ( 1 -((4-((4-(3 -(2,4-dioxotetrahydro pyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-
6-yl)piperidin- 1 -yl)methyl)phenyl)sulfonyl) piperidin-4-yl)carbamate
Figure imgf000255_0002
The title compound was prepared by proceeding analogously as described in Example 47, Steps 10 and 11, using l-(6-(l-(3-((4-aminopiperidin-l-yl)sulfonyl)benzyl)piperidin-4-yl)-l-methyl- lH-indazol-3-yl)dihydropyrimidine-2,4(lH,3H)-dione instead of 2-(2,6-dioxo-3-piperidyl)-5-[4-(4- piperidyl oxy)- l-piperidyl]isoindo line- 1,3-dione in Step 10. MS (ES, m/z): [M+H]+ = 1214.5.
Example 51
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-((4-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)piperazin-l- yl)methyl)piperidine- 1 -carboxylate
Figure imgf000255_0003
The title compound was prepared by proceeding analogously as described in Example 47, Steps 10 and 11, using 2-(2,6-dioxo-3-piperidyl)-5-[4-(4-piperidylmethyl)piperazin-l-yl]isoindoline- 1, 3-dione (ref: WO2021/155321) instead of 2-(2,6-dioxo-3-piperidyl)-5-[4-(4-piperidyloxy)-l- piperidyl] isoindoline- 1,3-dione in Step 10. MS (ES, m/z): [M+H]+ = 1074.5.
Example 52
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-(3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5- yl)amino)propyl)piperidine- 1 -carboxylate
Figure imgf000256_0001
Step 1: tert-butyl 4-(3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino) propyl)piperidine- 1 -carboxylate
Figure imgf000256_0002
A solution of tert-butyl 4-(3-aminopropyl)piperidine-l -carboxylate (242.17 mg, 1 mmol), 2- (2, 6-dioxopiperidin-3-yl)-5-fluoroisoindoline-l, 3-dione (276 mg, 1 mmol) and N,N-diisopropyl- ethylamine (0.52 mL, 3 mmol) in DMF (1 mL) was stirred for 12 h at 105 °C. After cooling to rt, water was added to the reaction mixture and the precipitates were isolated by filtration. The isolated solid was dissolved in DCM, dried over Na2SO4, and concentrated in vacuum. The residue was purified by silica gel column chromatography, eluted with EtOAc/PE = 0-50% to afford the title compound (120 mg).
Step 2: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen- 1 -yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- 1 H-
Figure imgf000257_0001
yl)methyl 4-((4-(3 -(2, 4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- lH-indazol-6- yl)phenyl)sulfonyl)piperidine- 1 -carboxylate
Figure imgf000258_0001
Step 1: tert-butyl 4-((4-(3-(2,4-dioxotetrahydropyrimidin-l(2H)-yl)-l-methyl-lH-indazol-6- yl)phenyl)sulfonyl)piperidine- 1 -carboxylate
Figure imgf000258_0002
To a solution of l-(6-bromo-l-methyl-indazol-3-yl)hexahydropyrimidine-2, 4-dione (162 mg, 0.5 mmol) and tert-butyl 4-[4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl]sulfonyl- piperazine- 1 -carboxylate (249.46 mg, 0.55 mmol) in 1,4-dioxane (5 mL) and water (1 mL) was added CsF (114.22 mg, 0.75 mmol) and Pd(dppf)C12 (40.61 mg, 0.05 mmol) and the resulting mixture was stirred for 5 h at 85 °C. After coolin tot rt, the reaction was quenched by water, extracted with DCM, washed with brine, dried over Na2SO4, and concentrated in vacuum. The residue was purified by silica gel column chromatography, eluted with MeOH/DCM (0-6%) to afford the title compound (270 mg). Step 2: ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoronaphthalen-l-yl)- 8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH-pyrrolizin-3-yl)methyl 4-((4-(3-(2,4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-6-yl)phenyl)sulfonyl)- piperidine- 1 -carboxylate
Figure imgf000258_0003
The title compound was prepared by proceeding analogously as described in Example 48, Steps 2-4, using : tert-butyl 4-((4-(3-(2,4-dioxotetrahydropyrimidin-l(2H)-yl)-l-methyl-lH-indazol- 6-yl)phenyl)sulfonyl)piperidine-l -carboxylate instead of tert-butyl 4-[l-[2-(2,6-dioxo-3-piperidyl)- 1 ,3 -dioxo-isoindolin-5-yl]piperidine-4-carbonyl]piperazine-l -carboxylate in Step 2. MS (ES, m/z): [M+H]+ = 1102.4.
Example 55
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-(3-(2,4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-6-yl)-
Figure imgf000259_0001
The title compound was prepared by proceeding analogously as described in Example 47, Steps 10 and 11, using l-(l-methyl-6-(piperidin-4-yl)-lH-indazol-3-yl)dihydropyrimidine- 2,4(lH,3H)-dione (ref: WO2021/127561) instead of 2-(2,6-dioxo-3-piperidyl)-5-[4-(4-piperidyloxy)- l-piperidyl]isoindoline- 1,3-dione in Step 10. MS (ES, m/z): [M+H]+ = 962.4.
Example 56
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl (4-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-5-yl)oxy)butyl)carbamate
Figure imgf000259_0002
The title compound was prepared by proceeding analogously as described in Example 47, Steps 10 and 11, using 3-(5-(4-aminobutoxy)-l-oxoisoindolin-2-yl)piperidine-2, 6-dione instead of 2- (2, 6-dioxo-3-piperidyl)-5-[4-(4-piperidyloxy)-l-piperidyl]isoindoline-l, 3-dione in Step 10. MS (ES, m/z): [M+H]+ = 966.4.
Example 57
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-(2-(2,6-dioxopiperidin-3-yl)- 1 -oxoisoindolin-4-yl)piperidine- 1 -carboxylate
Figure imgf000260_0001
The title compound was prepared by proceeding analogously as described in Example 47, Steps 10 and 11, using 3 -(l-oxo-4-(piperidin-4-yl)isoindolin-2-yl)piperidine-2, 6-dione (ref:
WO20 17/197056) instead of 2-(2,6-dioxo-3-piperidyl)-5-[4-(4-piperidyloxy)-l-piperidyl]isoindoline- 1, 3-dione in Step 10. MS (ES, m/z): [M+H]+ = 962.4.
Example 58
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-(2-(2,6-dioxopiperidin-3-yl)- 1 -oxoisoindolin-5-yl)piperidine- 1 -carboxylate
Figure imgf000260_0002
The title compound was prepared by proceeding analogously as described in Example 47, Steps 10 and 11, using 3-(l-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2, 6-dione (ref: WO2020/165833) instead of 2-(2,6-dioxo-3-piperidyl)-5-[4-(4-piperidyloxy)-l-piperidyl]isoindoline- 1, 3-dione in Step 10. MS (ES, m/z): [M+H]+ = 962.4. Example 59
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-(3-(2,4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-6-yl)- piperazine- 1 -carboxylate
Figure imgf000261_0001
The title compound was prepared by proceeding analogously as described in Example 47, Steps 10 and 11, using l-(l-methyl-6-(piperazin-l-yl)-lH-indazol-3-yl)dihydro pyrimidine- 2,4(lH,3H)-dione (ref: WO2021/127561) instead of 2-(2,6-dioxo-3-piperidyl)-5-[4-(4-piperidyloxy)- l-piperidyl]isoindoline- 1,3-dione in Step 10. MS (ES, m/z): [M+H]+ = 963.4.
Example 60
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl (2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)ethyl)(methyl)carbamate
Figure imgf000261_0002
The title compound was prepared by proceeding analogously as described in Example 47, Steps 10 and 11, using 2-(2,6-dioxo-3-piperidyl)-4-[2-(methylamino)ethylamino] isoindoline- 1,3- dione (ref: W02021/18018) instead of 2-(2,6-dioxo-3-piperidyl)-5-[4-(4-piperidyloxy)-l- piperidyl] isoindoline- 1,3-dione in Step 10. MS (ES, m/z): [M+H]+ = 965.4. Example 61
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl (4-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)amino)-2- oxoethyl)cyclohexyl)carbamate
Figure imgf000262_0001
The title compound was prepared by proceeding analogously as described in Example 49, Steps 1-2, using 2-(4-((tert-butoxycarbonyl)amino)cyclohexyl)acetic acid instead of 8-(tert- butoxycarbonylamino)octanoic acid in Step 1.. MS (ES, m/z): [M+H]+ = 1047.4.
Example 62
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 9-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)carbamoyl)-3- azaspiro[5.5]undecane-3-carboxylate
Figure imgf000262_0002
The title compound was prepared by proceeding analogously as described in Example 49, Steps 1-2, using 3-(tert-butoxycarbonyl)-3-azaspiro[5.5]undecane-9-carboxylic acid instead of 8-(tert- butoxycarbonylamino)octanoic acid in Step 1. MS (ES, m/z): [M+H]+ = 1087.3.
Example 63
Synthesis of ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoronaphthalen- l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH-pyrrolizin-3- yl)methyl 4-((3-(2,4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-6-yl)- ethynyl)piperidine- 1 -carboxylate
Figure imgf000263_0001
Step 1 : tert-butyl 4-((3 -(2,4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-6-yl)ethynyl)- piperidine- 1 -carboxylate
Figure imgf000263_0002
To a mixture of l-(5-bromo-l-methyl-indazol-3-yl)hexahydropyrimidine-2, 4-dione (323 mg, 1.0 mmol), tert-butyl 4-ethynylpiperidine- 1 -carboxylate (418.39 mg, 2.mmol), Pd(PPh3)2C12 (70.16 mg, 0.1 mmol), and Cui (38.07 mg, 0.2 mmol) in DMF (2 mL), was added TEA (0.7 mL, 5.0 mmol) and the resulting mixture was stirred at 105 °C for 3 h under Ar atmosphere. After cooling to rt, the reaction mixture was quenched by water, extracted by DCM, washed with brine, dried over anhydrous Na2SO4, and concentrated in vacuum to afford the title compound (420 mg).
Step 2: ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoronaphthalen-l-yl)- 8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH-pyrrolizin-3-yl)methyl 4-((3-(2,4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-6-yl)ethynyl)piperidine- 1 - carboxylate
Figure imgf000263_0003
The title compound was prepared by proceeding analogously as described in Example 48, Steps 2-4, using tert-butyl 4-((3-(2,4-dioxotetrahydropyrimidin-l(2H)-yl)-l -methyl- lH-indazol-6- yl)ethynyl)piperidine-l -carboxylate instead of tert-butyl 4-[l-[2-(2,6-dioxo-3-piperidyl)-l,3-dioxo- isoindolin-5-yl]piperidine-4-carbonyl]piperazine-l -carboxylate in Step 2. MS (ES, m/z): [M+H]+ = 986.4.
Example 64
Synthesis of ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoronaphthalen- l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH-pyrrolizin-3- yl)methyl 4-(2-(3-(2,4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-6- yl)ethyl)piperidine- 1 -carboxylate
Figure imgf000264_0001
Step 1 : tert-butyl 4-(2-(3-(2,4-dioxotetrahydropyrimidin-l(2H)-yl)-l-methyl-lH-indazol-6-yl)ethyl)- piperidine- 1 -carboxylate
Figure imgf000264_0002
To a solution of tert-butyl 4-[2-[3-(2,4-dioxohexahydropyrimidin-l-yl)-l-methyl-indazol-6- yl]-ethynyl]piperidine-l -carboxylate (120 mg, 0.27 mmol) in ethanol (2 mL) was added 10% Pd/C (20 mg) and ammonium formate (16.76 mg, 0.27 mmol) and the resulting mixture was stirred at 25 °C for 12 h. The reaction mixture was filtered and the filtrate was evaporated in vacuum. The residue was purified by silica gel column chromatography, eluted with MeOH/DCM (0-6%) to afford the title compound (120 mg).
Step 2: ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoronaphthalen-l-yl)- 8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH-pyrrolizin-3-yl)methyl 4-(2-(3 -(2,4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-6-yl)ethyl)piperidine- 1 - carboxylate
Figure imgf000265_0001
The title compound was prepared by proceeding analogously as described in Example 48, Steps 2-4, using tert-butyl 4-(2-(3 -(2, 4-dioxotetrahydropyrimi din- l(2H)-yl)-l -methyl- lH-indazol-6- yl)ethyl)piperidine-l -carboxylate instead of tert-butyl 4-[l-[2-(2,6-dioxo-3-piperidyl)-l,3-dioxo- isoindolin-5-yl]piperidine-4-carbonyl]piperazine-l -carboxylate in Step 2. MS (ES, m/z): [M+H]+ = 990.4.
Example 65
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-((l-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)piperidin-4-yl)-
Figure imgf000265_0002
The title compound was prepared by proceeding analogously as described in Example 47, Steps 10 and 11, using 2-(2,6-dioxo-3-piperidyl)-5-[4-(4-piperidylmethyl)-l-piperidyl]isoindoline- 1, 3-dione (ref: WO2022/120355) instead of 2-(2,6-dioxo-3-piperidyl)-5-[4-(4-piperidyloxy)-l- piperidyl] isoindoline- 1,3-dione in Step 10. MS (ES, m/z): [M+H]+ = 1073.5.
Example 66
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl (2-(l-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)piperidin-4- yl)ethyl)carbamate
Figure imgf000266_0001
The title compound was prepared by proceeding analogously as described in Example 47, Steps 10 and 11, using 5-[4-(2-aminoethyl)-l-piperidyl]-2-(2,6-dioxo-3-piperidyl) isoindoline- 1,3- dione (ref: WO2021/155321) instead of 2-(2,6-dioxo-3-piperidyl)-5-[4-(4-piperidyloxy)-l- piperidyl] isoindoline- 1,3-dione in Step 10. MS (ES, m/z): [M+H]+ = 1019.4.
Example 67
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-((2-(2,6-dioxopiperidin-3-yl)- 1 ,3-dioxoisoindolin-5- yl)carbamoyl)piperidine- 1 -carboxylate
Figure imgf000266_0002
The title compound was prepared by proceeding analogously as described in Example 49, Steps 1-2, using l-tert-butoxycarbonylpiperidine-4-carboxylic acid instead of 8-(tert- butoxycarbonylamino)octanoic acid in Step 1. MS (ES, m/z): [M+H]+ = 1019.4.
Example 68
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl (4-(4-(3 -(2,4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-6- yl)piperidine- 1 -carbonyl)cyclohexyl)carbamate
Figure imgf000267_0001
Step 1: tert-butyl (4-(4-(3-(2,4-dioxotetrahydropyrimidin-l(2H)-yl)-l-methyl-lH-indazol-6-yl)- piperidine- 1 -carbonyl)cyclohexyl)carbamate
Figure imgf000267_0002
A solution of HATU (116.14 mg, 0.31 mmol), N,N-diisopropylethylamine (0.08 mL, 0.46 mmol), 4-(tert-butoxycarbonylamino)cyclohexane-l -carboxylic acid (44.77 mg, 0.18 mmol), 1-[1- methyl-6-(4-piperidyl)indazol-3-yl]hexahydropyrimidine-2, 4-dione (50 mg, 0.15 mmol) in DMF (1 mL) was stirred at rt for 12 h. The reaction was quenched with water, extracted by EtOAc, washed with brine, dried overNa2 SO4, and concentrated in vacuum. The residue was purified by column chromatography, eluted with MeOH/DCM (0-10%) to afford the title compound (76 mg).
Step 2: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen- 1 -yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- 1 H- pyrrolizin-3-yl)methyl (4-(4-(3 -(2,4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-6- yl)piperidine- 1 -carbonyl)cyclohexyl)carbamate
Figure imgf000267_0003
The title compound was prepared by proceeding analogously as described in Example 48, Steps 2-4, using tert-butyl (4-(4-(3 -(2, 4-dioxotetrahydropyrimi din- l(2H)-yl)-l -methyl- lH-indazol-6- yl)piperidine- 1 -carbonyl)cyclohexyl)carbamate instead of tert-butyl 4-[l-[2-(2,6-dioxo-3-piperidyl)- 1,3 -di oxo-isoindolin-5-yl]piperidine-4-carbonyl]piperazine-l -carboxylate in Step 2. MS (ES, m/z): [M+H]+ = 1087.4. Example 69
Synthesis of ((3R,7aR)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-(4-(3-(2,4-dioxotetrahydropyrimidin-l(2H)-yl)-l -methyl- lH-indazol-6-yl)- piperidine- 1 -carbonyl)piperidine- 1 -carboxylate
Figure imgf000268_0001
The title compound was prepared by proceeding analogously as described in Example 68, Steps 1-2, using l-tert-butoxycarbonylpiperidine-4-carboxylic acid instead of 4-(tert- butoxycarbonylamino)cyclohexane- 1 -carboxylic acid in Step 1. MS (ES, m/z): [M+H]+ = 1074.2 Example 70
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-(4-((3 -(2,4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-6- yl)amino)piperidine- 1 -carbonyl)piperidine- 1 -carboxylate
Figure imgf000268_0002
Step 1: tert-butyl 4-((3 -(2, 4-dioxotetrahydropyrimi din- l(2H)-yl)-l -methyl- 1 H-indazol-6-yl)amino)- piperidine- 1 -carboxylate
Figure imgf000268_0003
A mixture of l-(6-bromo-l-methyl-lH-indazol-3-yl)dihydropyrimidine-2,4(lH,3H)-dione (50 mg, 0.15 mmol, 1.0 eq), tert-butyl 4-aminopiperi dine- 1 -carboxylate (37.19 mg, 0.19 mmol, 1.2 eq), CS2CO3 (60.53 mg, 0.19 mmol, 1.2 eq), Brettphos (16.61 mg, 0.03 mmol, 0.03 eq) and Pd2(dba)3 (14.17 mg, 0.02 mmol, 0.1 eq) in 1,4-dioxane (1.0 mL) was stirred at 90 °C for 3 h under Ar2 atmosphere. After cooling to rt, the mixture was filtrated and the filtrate was concentrated under reduced pressure. The residue obtained was purified by silica gel column chromatography, eluted with DCM/MeOH (0-5%) to afford the title compound (40 mg).
Step 2: l-(l-methyl-6-(piperidin-4-ylamino)-lH-indazol-3-yl)dihydropyrimidine-2,4(lH,3H)-dione
Figure imgf000269_0001
To a solution of tert-butyl 4-((3-(2,4-dioxotetrahydropyrimidin- l(2H)-yl)-l -methyl- 1H- indazol-6-yl)amino)piperi dine- 1 -carboxylate (40 mg, 0.09 mmol, 1.0 eq) in EtOAc (1 mL) was added HC1 solution in EtOAc (2.0 M, 1 mL) at 25°C and the resulting mixture was stirred at 25°C for 1 h. The mixture was concentrated in reduced pressure to afford the title compound (30 mg). Step 3: tert-butyl 4-(4-((3-(2,4-dioxotetrahydropyrimidin-l(2H)-yl)-l-methyl-lH-indazol-6- yl)amino)piperidine- 1 -carbonyl)piperidine- 1 -carboxylate
Figure imgf000269_0002
A mixture of l-(l-methyl-6-(piperidin-4-ylamino)-lH-indazol-3-yl)dihydro pyrimidine - 2,4(lH,3H)-dione (30 mg, 0.09 mmol , 1.0 eq), 1 -(tert-butoxycarbonyl) piperidine-4-carboxylic acid (22.10 mg, 0.10 mmol, 1.1 eq), 1 -methylimidazole (28.78 mg, 0.35 mmol, 4.0 eq) and TCFH (49.17 mg, 0.18 mmol, 2.0 eq) in DMF (1 mL) was stirred at 25 °C for 5 h. The mixture was quenched with water, extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2SO4, and concentrated under reduced pressure. The residue obtained was purified by column chromatography, eluted with DCM/MeOH (0-5%) to afford the title compound (35 mg). Step 4. ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro- naphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-(4-((3 -(2, 4-dioxotetrahydropyrimi din- l(2H)-yl)-l -methyl- lH-indazol-6- yl)amino)piperidine- 1 -carbonyl)piperidine- 1 -carboxylate
Figure imgf000270_0001
The title compound was prepared by proceeding analogously as described in Example 48, Steps 2-4, using tert-butyl 4-(4-((3 -(2, 4-dioxotetrahydropyrimi din- l(2H)-yl)-l -methyl- lH-indazol-6- yl)amino)piperi dine- 1 -carbonyl)piperidine- 1 -carboxylate instead of tert-butyl 4-[l-[2-(2,6-dioxo-3- piperidyl)-l,3-dioxo-isoindolin-5-yl]piperidine-4-carbonyl]piperazine-l-carboxylate in Step 2. MS (ES, m/z): [M+H]+ = 1088.4
Example 71
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-(4-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)oxy)piperidine-l- carbonyl)piperidine- 1 -carboxylate
Figure imgf000270_0002
Step 1: tert-butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)oxy)piperidine-l- carboxylate
Figure imgf000270_0003
A mixture of PPI13 (393.25 mg, 1.5 mmol) tert-butyl 4-hydroxypiperi dine- 1 -carboxylate (0.38 mL, 2 mmol), DIAD (0.58 mL, 3 mmol) and 2-(2,6-dioxo-3-piperidyl)-5-hydroxy-isoindoline-l,3- dione (274.1 mg, 1 mmol) in THF (2 mL) was stirred at 25 °C for 12 h. The reaction mixture was diluted with water, extracted with DCM. The combined organic layers were washed with brine, dried over Na2SO4 and concentrated. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (0-6%) to afford the title compound (230 mg).
Step 2: 2-(2,6-dioxopiperidin-3-yl)-5-(piperidin-4-yloxy)isoindoline- 1,3-dione
Figure imgf000271_0001
To a solution of tert-butyl 4-[2-(2,6-dioxo-3-piperidyl)-l,3-dioxo-isoindolin-5- yl] oxypiperidine- 1 -carboxylate (230 mg, 0.50 mmol) in EtOAc (ImL), was added HC1 solution in EA (4.0 M, 1 mL) and the resulting mixture was stirred at 25 °C for 1 h. The reaction mixture was concentrated in vacuum and the residue obtained was washed with EtOAc to afford the title compound (120 mg).
Step 3: tert-butyl 4-(4-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)oxy) piperidine- 1 - carbonyl)piperidine- 1 -carboxylate
Figure imgf000271_0002
A mixture of TEA (0.05 mL, 0.34 mmol), HATU (63.84 mg, 0.17 mmol), 2-(2,6-dioxo-3- piperidyl)-5-(4-piperidyloxy)isoindoline- 1,3-dione (40 mg, 0.11 mmol), 1 -tert-butoxycarbonyl piperidine-4-carboxylic acid (0.03 mL, 0.13 mmol) in DMF (1 mL) was stirred at 25 °C for 12 h. The reaction was quenched by water, extracted by EtOAc, dried over Na2SO4 and concentrated in vacuum. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (0-6%) to afford the title compound (35 mg).
Step 4: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro- naphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-(4-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)oxy)piperidine-l- carbonyl)piperidine- 1 -carboxylate
Figure imgf000271_0003
The title compound was prepared by proceeding analogously as described in Example 48, Steps 2-4, using tert-butyl 4-[4-[2-(2,6-dioxo-3-piperidyl)-l,3-dioxo-isoindolin-5-yl]oxypiperidine-l- carbonyl]piperidine-l -carboxylate instead of tert-butyl 4-[l-[2-(2,6-dioxo-3-piperidyl)-l,3-dioxo- isoindolin-5-yl]piperidine-4-carbonyl]piperazine-l -carboxylate in Step 2. MS (ES, m/z): [M+H]+ = 1104.4
Example 72
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3 -yl)methyl (3 -(4-(3 -(2,4-dioxotetrahydro pyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-6- yl)piperidine- 1 -carbonyl)cyclopentyl) carbamate
Figure imgf000272_0001
The title compound was prepared by proceeding analogously as described in Example 68, Steps 1-2, using 3-(tert-butoxycarbonylamino)cyclopentanecarboxylic acid instead of 4-(tert- butoxycarbonylamino)piperidine- 1 -carboxylic acid in Step 1. MS (ES, m/z): [M+H]+ = 1073.5.
Example 73
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 3-(4-(3-(2,4-dioxotetrahydropyrimidin- l(2H)-yl)-l -methyl- lH-indazol-6- yl)piperidine- 1 -carbonyl)pyrrolidine- 1 -carboxylate
Figure imgf000272_0002
The title compound was prepared by proceeding analogously as described in Example 68, Steps 1-2, using 1 -tert-butoxycarbonylpyrrolidine-3 -carboxylic acid instead of 4-(tert- butoxycarbonylamino)piperidine- 1 -carboxylic acid in Step 1. MS (ES, m/z): [M+H]+ = 1060.2
Example 74
Synthesis of ((3R,7aR)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 3-(4-(3-(2,4-dioxotetrahydropyrimidin- l(2H)-yl)-l -methyl- lH-indazol-6- yl)piperidine- 1 -carbonyl)piperidine- 1 -carboxylate
Figure imgf000273_0001
The title compound was prepared by proceeding analogously as described in Example 68, Steps 1-2, using l-tert-butoxycarbonylpiperidine-3 -carboxylic acid instead of 4-(tert- butoxycarbonylamino)piperidine- 1 -carboxylic acid in Step 1. MS (ES, m/z): [M+H]+ = 1073.5
Example 75
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl (2-(l-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)piperidine-4-
Figure imgf000273_0002
Step 1: methyl l-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)piperidine-4-carboxylate
Figure imgf000274_0001
A solution of methyl piperidine-4-carboxylate (0.27 mL, 2 mmol), TEA (0.42 mL, 3.0 mmol) and 2-(2,6-dioxo-3-piperidyl)-5-fluoro-isoindoline- 1,3-dione (276.09 mg, 1.0 mmol) in DMF (2mL) was heated at 120 °C for 1 h with microwave. After cooling to rt, the reaction mixture was diluted with water, extracted by EtOAc, dried over Na2SO4 and concentrated. The residue was purified by column chromatography, eluted with DCM/MeOH (0-10%) to afford the title compound (210 mg).
Step 2: l-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)piperidine-4-carboxylic acid
Figure imgf000274_0002
A mixture of methyl 1-[2-(2,6-dioxo-3-piperidyl)-l,3-dioxo-isoindolin-5-yl]piperidine-4- carboxylate (100 mg, 0.25 mmol) and sulfuric acid (0.04 mL, 0.75 mmol) in water (2 mL) was stirred at 90 °C for 5 h. The reaction mixture was extracted by EtOAc, dried over Na2SO4 and concentrated to afford the title compound (96 mg).
Step 3: tert-butyl (2-(l-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)piperidine-4- carboxamido)ethyl)carbamate
Figure imgf000274_0003
A solution of TEA (0.04 mL, 0.26 mmol), tert-butyl N-(2-aminoethyl)carbamate (0.07 mL, 0.78 mmol), HATU (98.67 mg, 0.26 mmol) and l-[2-(2,6-dioxo-3-piperidyl)-l,3-dioxo-isoindolin-5- yl]piperidine-4-carboxylic acid (96 mg, 0.2491 mmol) in DMF ( ImL) was stirred at 25 °C for 12 h. The reaction was quenched by water, extracted by EtOAc, washed with brine, dried over Na2SO4 and concentrated in vacuum. The residue was purified by column chromatography, eluted with DCM/MeOH (0-6%) to afford the title compound (50 mg).
Step 4: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro- naphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl (2-(l-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)piperidine-4- carboxamido)ethyl)carbamate
Figure imgf000275_0001
The title compound was prepared by proceeding analogously as described in Example 48, Steps 2-4, using tert-butyl (2-(l-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl) piperidine-4- carboxamido)ethyl)carbamate instead of tert-butyl 4-[l-[2-(2,6-dioxo-3-piperidyl)-l,3-dioxo- isoindolin-5-yl]piperidine-4-carbonyl]piperazine-l -carboxylate in Step 2. MS (ES, m/z): [M+H]+ = 1063.5.
Example 76
Synthesis of ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7- fluoronaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylene hexahydro-
1 H-pyrrolizin-3 -yl)methyl 4-((4-(3 -(2 ,4-dioxotetrahydropyrimidin- 1 (2H)-yl)- 1 -methyl- 1 H-indazol-6- yl)piperidin- 1 -yl)sulfonyl)piperidine- 1 -carboxylate
Figure imgf000275_0002
Step 1: tert-butyl 4-((4-(3-(2,4-dioxotetrahydropyrimidin-l(2H)-yl)-l-methyl-lH-indazol-6- yl)piperidin- 1 -yl)sulfonyl)piperidine- 1 -carboxylate
Figure imgf000275_0003
A solution of l-[l-methyl-6-(4-piperidyl)indazol-3-yl]hexahydropyrimidine-2, 4-dione (30 mg, 0.09 mmol), TEA (0.04 mL, 0.27 mmol) and tert-butyl 4-chlorosulfonylpiperidine- 1 -carboxylate (39 mg, 0.14 mmol) in DMSO (ImL) and THF (ImL) was stirred at 90 °C for 5 h. After cooling to rt, the reaction mixture was diluted with water, extracted by EtOAc, washed with brine, and evaporated in vacuum. The residue was purified by column chromatography, eluted with DCM/MeOH (0-8%) to afford the title compound (40 mg).
Step 2: ((3S,7aS)-7a-(((4-((lR,5S)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro- naphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylene hexahydro- 1H- pyrrolizin-3-yl)methyl 4-((4-(3 -(2, 4-dioxotetrahydropyrimi din- l(2H)-yl)-l -methyl- lH-indazol-6- yl)piperidin- 1 -yl)sulfonyl)piperidine- 1 -carboxylate
Figure imgf000276_0001
The title compound was prepared by proceeding analogously as described in Example 48, Steps 2-4, using tert-butyl 4-((4-(3 -(2, 4-dioxotetrahydropyrimi din- l(2H)-yl)-l -methyl- lH-indazol-6- yl)piperidin-l-yl)sulfonyl)piperidine-l -carboxylate instead of tert-butyl 4-[l-[2-(2,6-dioxo-3- piperidyl)-l,3-dioxo-isoindolin-5-yl]piperidine-4-carbonyl]piperazine-l-carboxylate in Step 2. MS (ES, m/z): [M+H]+ = 1110.4.
Example 501
Synthesis of ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(4-(((S)-l-((2S,4R)-4-hydroxy-2-(((S)-l-(4-(4-methylthiazol-5- yl)phenyl)ethyl)carbamoyl)pyrrolidin- 1 -yl)-3 ,3-dimethyl- 1 -oxobutan-2-yl)carbamoyl)- 1 H- 1 ,2,3- triazol- 1 -yl)piperidine- 1 -carboxylate
Figure imgf000276_0002
Step 1: tert-butyl 4-(4-(((S)-l-((2S,4R)-4-hydroxy-2-(((S)-l-(4-(4-methylthiazol-5-yl)phenyl)- ethyl)carbamoyl)pyrrolidin- 1 -yl)-3 ,3 -dimethyl- 1 -oxobutan-2-yl)carbamoyl)- 1 H- 1 ,2,3-triazol- 1 -yl)- piperidine- 1 -carboxylate
Figure imgf000277_0001
A mixture of (2S,4R)-l-[(2S)-2-amino-3,3-dimethylbutanoyl]-4-hydroxy-N-[(lS)-l-(4-(4- methyl-l,3-thiazol-5-yl)phenyl)ethyl]pyrrolidine-2-carboxamide (0.1 g, 0.22 mmol), l-(l-[(tert- butoxy)carbonyl]piperidin-4-yl)-lH-l,2,3-triazole-4-carboxylic acid (0.098 g, 0.33 mmol), HATU (0.10 g, 0.26 mmol) and DIPEA (0.085 g, 0.66 mmol) in DMF (3 mL) was stirred at rt for 16 h. The reaction mixture was purified by reverse phase C18 column chromatography (10-100% MeOH/H2O with 0.1% NH4HCO3] to give the title compound (0.11 g).
Step 2: N-((S)-l-((2S,4R)-4-hydroxy-2-(((S)-l-(4-(4-methylthiazol-5-yl)phenyl)ethyl)- carbamoyl)pyrro lidin- 1 -y 1) - 3 ,3 -dimethyl- 1 -oxobutan-2-yl)- 1 -(piperidin-4-yl)- 1 H- 1 ,2 ,3 -triazole-4- carboxamide
Figure imgf000277_0002
To a solution of tert-butyl 4-(4-{[(2S)-l-[(2S,4R)-4-hydroxy-2-{[(lS)-l-(4-(4-methyl-l,3- thiazol-5 -yl)phenyl)ethyl] carbamoyl [pyrrolidin- 1 -yl] -3 ,3 -dimethyl- 1 -oxobutan-2-yl] carbamoyl} - 1 H- 1, 2, 3 -triazol- l-yl)piperidine-l -carboxylate (0.03 g, 0.041 mmol) in DCM (3 mL) was added a solution of HC1 in dioxane (4.0 M , 1.0 mL). The reaction mixture was stirred at rt for 3 h and then concentrated to give the title compound (0.025 g).
Step 3: ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3-hydroxy- naphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-(4-(((S)-l-((2S,4R)-4-hydroxy-2-(((S)-l-(4-(4-methylthiazol-5- yl)phenyl)ethyl)carbamoyl)pyrrolidin- 1 -yl)-3 ,3-dimethyl- 1 -oxobutan-2-yl)carbamoyl)- 1 H- 1 ,2,3- triazol- 1 -yl)piperidine- 1 -carboxylate
Figure imgf000278_0001
The title compound was prepared by proceeding analogously as described in Example 1 , Steps 11 and 12, using N-((S)-l-((2S,4R)-4-hydroxy-2-(((S)-l-(4-(4-methylthiazol-5-yl)phenyl)ethyl)- carbamoyl)pyrro lidin- 1 -y 1) - 3 ,3 -dimethyl- 1 -oxobutan-2-yl)- 1 -(piperidin-4-yl)- 1 H- 1 ,2 ,3 -triazole-4- carboxamide instead of 4-((2-(2-(2-aminoethoxy)ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline- 1,3-dione in Step 11. MS (ESI) m/z = 637.1 [(M+2)/2]+.
Example 502
Synthesis ((3S,7aS)-7a-(((4-((lR,5R)-3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen- 1 -yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-
1 H-pyrrolizin-3 -yl)methyl 4-((l-((S)-3 -((2 S,4R)- l-((S)-2-(l -fluorocyclopropane- 1 -carboxamido)-3 ,3 - dimethylbutanoyl)-4-hydroxypyrrolidine-2-carboxamido)-3-(4-(4-methylthiazol-5- yl)phenyl)propanoyl)piperidin-4-yl)methyl)piperidine-l -carboxylate
Figure imgf000278_0002
Step 1: tert-butyl 4-((l-((S)-3-((2S,4R)-l-((S)-2-(l-fluorocyclopropane-l-carboxamido)-3,3- dimethylbutanoyl)-4-hydroxypyrrolidine-2-carboxamido)-3-(4-(4-methylthiazol-5-yl)phenyl)- propanoyl)piperidin-4-yl)methyl)piperidine- 1 -carboxylate
Figure imgf000279_0001
To a solution of (S)-3-((2S,4R)-l-((S)-2-(l-fluorocyclopropane-l-carboxamido)-3,3- dimethylbutanoyl)-4-hydroxypyrrolidine-2-carboxamido)-3-(4-(4-methylthiazol-5-yl)phenyl)- propanoic acid (synthesized as described in WO2021222114A1) (176 mg, 626 pmol) and tert-butyl 4- (piperidin-4-ylmethyl)piperi dine- 1 -carboxylate in DCM (5 mL) was added TEA (158 mg, 1.57 mmol) and HATU (238 mg, 626 pmol). The resulting mixture was stirred at 20 °C for 2 h. The reaction mixture was diluted with DCM, washed with brine, dried over Na2SO4 and concentrated. The residue was purified by flash chromatography to give the title compound (430 mg).
Step 2: ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3-hydroxy- naphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-((l -((S)-3-((2S,4R)-l-((S)-2-(l -fluorocyclopropane- l-carboxamido)-3, 3- dimethylbutanoyl)-4-hydroxypyrrolidine-2-carboxamido)-3-(4-(4-methylthiazol-5-yl)phenyl)- propanoyl)piperidin-4-yl)methyl)piperidine- 1 -carboxylate
Figure imgf000279_0002
The title compound was prepared by proceeding analogously as described in Example 501, Steps 2 and 3, using tert-butyl 4-((l-((S)-3-((2S,4R)-l-((S)-2-(l-fluorocyclopropane-l-carboxamido)- 3,3-dimethylbutanoyl)-4-hydroxypyrrolidine-2-carboxamido)-3-(4-(4-methylthiazol-5-yl)phenyl)- propanoyl)piperidin-4-yl)methyl)piperidine-l -carboxylate instead of tert-butyl 4-(4-(((S)-l-((2S,4R)- 4-hydroxy-2-(((S)- 1 -(4-(4-methylthiazol-5-yl)phenyl)ethyl)carbamoyl)pyrrolidin- 1 -yl)-3,3-dimethyl- l-oxobutan-2-yl)carbamoyl)-lH-l, 2, 3 -triazol- l-yl)piperi dine- 1 -carboxylate in Step 2. MS (ESI) m/z = 695.6 [(M+2)/2]+. Example 503
Synthesis of ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3- hydroxynaphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro- lH-pyrrolizin-3-yl)methyl 4-(l-((S)-l-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5- yl)benzyl)carbamoyl)pyrrolidin- 1 -yl)-3-methyl- 1 -oxobutan-2-yl)- 1 H- 1 ,2,3-triazol-4-yl)piperidine- 1 -
Figure imgf000280_0001
Step 1 : (2S,4R)- 1 -(L-valyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide
Figure imgf000280_0002
Tert-butyl ((S)-l-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5-yl)benzyl)carbamoyl)- pyrrolidin- 1 -yl)-3 -methyl- l-oxobutan-2-yl)carbamate (900 mg, 1.74 mmol) was added to a solution of HC1 in dioxane (4.0 M, 10.0 mL) and the resulting solution stirred at 20 °C for 0.5 h. The reaction mixture was concentrated to give the title compound (780 mg).
Step 2: (2S,4R)-l-((S)-2-azido-3-methylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)- pyrrolidine-2-carboxamide
Figure imgf000280_0003
To a solution of (2S,4R)-l-(L-valyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)- pyrrolidine-2-carboxamide (820 mg, 1.81 mmol) in MeOH (10 mL) was added K2CO3 (1.13 g, 8.15 mmol) and CUSO4.5H2O (45.2 mg, 181 pmol). IH-imidazole-l -sulfonylazide hydrochloride (759 mg, 3.62 mmol) was then added dropwise to the above mixture and the resulting mixture was stirred at 20 °C for 12 h. The reaction mixture was diluted with H2O, extracted with EA, washed with brine, dried over Na2SO4 and concentrated. The residue was purified by flash chromatography (PE/EA = 1/0 to
0/1) to give the title compound (500 mg).
Step 2: tert-butyl 4-(l-((S)-l-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5-yl)benzyl)carbamoyl)- pyrro lidin- 1 -yl)-3 -methyl- 1 -oxobutan-2-yl)- 1 H- 1 ,2,3-triazol-4-yl)piperidine- 1 -carboxylate
Figure imgf000281_0001
A mixture of (2S,4R)-l-((S)-2-azido-3-methylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5- yl)benzyl)pyrrolidine-2-carboxamide (100 mg, 226 pmol), tert-butyl 4-ethynylpiperidine- 1 - carboxylate, CUSO4.5H2O (10.3 mg, 56.5 pmol), sodium ascorbate (17.9 mg, 90.4 pmol) in t-BuOH (0.5 mL) and H2O (0.5 mL) was stirred at 20 °C for 12 h under N2. The reaction mixture was diluted with H2O, extracted with EA, washed with brine, dried over Na2SO4 and concentrated. The residue was purified by prep-HPLC to give the title compound (115 mg).
Step 3: ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoro-3-hydroxy- naphthalen-l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH- pyrrolizin-3-yl)methyl 4-(l-((S)-l-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5-yl)benzyl)- carbamoyl)pyrrolidin- 1 -yl)-3 -methyl- 1 -oxobutan-2-yl)- 1 H- 1 ,2,3-triazol-4-yl)piperidine- 1 -carboxylate
Figure imgf000281_0002
The title compound was prepared by proceeding analogously as described in Example 501, Steps 2 and 3, using tert-butyl 4-(l-((S)-l-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5- yl)benzyl)carbamoyl)pyrrolidin- 1 -yl)-3-methyl- 1 -oxobutan-2-yl)- 1H- 1 ,2,3-triazol-4-yl)piperidine- 1 - carboxylate instead of tert-butyl 4-(4-(((S)-l-((2S,4R)-4-hydroxy-2-(((S)-l-(4-(4-methylthiazol-5- yl)phenyl)ethyl)carbamoyl)pyrrolidin- 1 -yl)-3 ,3-dimethyl- 1 -oxobutan-2-yl)carbamoyl)- 1 H- 1 ,2,3- triazol- 1 -yl)piperi dine- 1 -carboxylate in Step 2. MS (ESI) m/z =601.7 [(M+2)/2]+.
Example 504 Synthesis of ((3S,7aS)-7a-(((4-(3,8-diazabicyclo[3.2.1]octan-3-yl)-7-(8-ethynyl-7-fluoronaphthalen- l-yl)-8-fluoropyrido[4,3-d]pyrimidin-2-yl)oxy)methyl)-6-methylenehexahydro-lH-pyrrolizin-3- yl)methyl 4-(( 1 -((S)-3-((2S,4R)- 1 -((S)-2-( 1 -fluorocyclopropane- 1 -carboxamido)-3 ,3- dimethylbutanoyl)-4-hydroxypyrrolidine-2-carboxamido)-3-(4-(4-methylthiazol-5- yl)phenyl)propanoyl)piperidin-4-yl)methyl)piperidine-l -carboxylate
Figure imgf000282_0001
The title compound was prepared by proceeding analogously as described in Example 47, Steps 10 and 11, using (2S,4R)-l-((S)-2-(l-fluorocyclopropane-l-carboxamido)-3,3-dimethyl- butanoyl)-4-hydroxy-N-((S)-l-(4-(4-methylthiazol-5-yl)phenyl)-3-oxo-3-(4-(piperidin-4-yl- methyl)piperidin-l-yl)propyl)pyrrolidine-2-carboxamide instead of 2-(2,6-dioxo-3-piperidyl)-5-[4-(4- piperidyloxy)- l-piperidyl]isoindoline- 1,3-dione in Step 10. MS (ESI) m/z = 723.3 [(M+2)/2]+.
Compounds 505-533 in Compound Table 2 were prepared by proceeding analogously as described in
Example 501, using acids in table below instead of l-(l-[(tert-butoxy)carbonyl]piperidin-4-yl)-lH- l,2,3-triazole-4-carboxylic acid in Example 501, Step 1.
Figure imgf000282_0002
Figure imgf000283_0001
Figure imgf000284_0001
Figure imgf000285_0001
Figure imgf000286_0001
Compounds 505, 515, 534 in Compound Table 2 were prepared by proceeding analogously as described in Example 501 Step 1-3, using acids in table below instead of l-(l-[(tert- butoxy)carbonyl]piperidin-4-yl)-lH-l,2,3-triazole-4-carboxylic acid in Example 501, step 1 and tert- butyl 3-(7-(8-ethynyl-7-fluoronaphthalen-l-yl)-8-fluoro-2-(((5S,7aS)-2-methylene-5-((((4- nitrophenoxy)carbonyl)-oxy)methyl)tetrahydro-lH-pyrrolizin-7a(5H)-yl)methoxy)pyrido[4,3- d]pyrimidin-4-yl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate instead of tert-butyl 3-(7-(8-ethynyl-7- fluoro-3-(methoxymethoxy)naphthalen- 1 -yl)-8-fluoro-2- { [(2S,4R)- 1 -methyl-4- {[(4- nitrophenoxy)carbonyl]oxy}pyrrolidin-2-yl]methoxy}pyrido[4,3-d]pyrimidin-4-yl)-3,8- diazabicyclo[3.2.1]octane-8-carboxylate in Example 501, step 3.
Figure imgf000287_0001
Compounds 514, 525, 535 in Compound Table 2 were prepared by proceeding analogously as described in Example 502, step 1 and 2, using amines in table below instead of tert-butyl 4-(piperidin- 4-ylmethyl)-piperidine- 1 -carboxylate in step 1.
Figure imgf000287_0002
Figure imgf000288_0001
Biological Examples
Example 1 AGS 3D Spheroid Cell Proliferation Assays
AGS cells were seeded into 96-well round black/clear bottom, ultra-low attachment surface plate in 100 pl cell culture medium (RPMI1640 with 10% FBS). After 3 days incubation at 37 °C and 5% CO2, compounds solubilized in DMSO were added by Tecan D300e dispenser (0.5% DMSO final). The cells were incubated for 4 days at 37 °C and 5% CO2. Cell proliferation was quantitated by addition of 50 pl/well of CellTiter-Glo® 3D reagent (Promega). The solutions were well mixed by shaking the plate for 10 minutes using an orbital plate shaker and then incubated at room temperature for a total of 30 minutes. After incubation, luminescence was then measured on a EnVision multimode plate reader (PerkinElmer). The results were normalized to percentage inhibition with DMSO control as 0% inhibition. The normalized luminescence results were plotted against compound concentration, and the data fit to 4-Parameter Logistic Model to calculate the EC50 by XLfit 5.5.0.
Figure imgf000288_0002
Figure imgf000289_0001
AspC-1 3D Spheroid Cell Proliferation Assays
AspC- 1 cells were seeded into 96-well round black/clear bottom, ultra-low attachment surface plate in 100 pl cell culture medium (RPMI1640 with 10% FBS). After 3 days incubation at 37 °C and 5% CO2, compounds solubilized in DMSO were added by Tecan D300e dispenser (0.5% DMSO final). The cells were incubated for 4 days at 37 °C and 5% CO2. Cell proliferation was quantitated by addition of 50 pl/well of CellTiter-Glo® 3D reagent (Promega). The solutions were well mixed by shaking the plate for 10 minutes using an orbital plate shaker and then incubated at room temperature for a total of 30 minutes. After incubation, luminescence was then measured on a EnVision multimode plate reader (PerkinElmer). The results were normalized to percentage inhibition with DMSO control as 0% inhibition. The normalized luminescence results were plotted against compound concentration, and the data fit to 4-Parameter Logistic Model to calculate the EC50 by XLfit 5.5.0.
Table 5. Inhibitory activity in AspC-1 3D spheroid cell proliferation assay.
A: Less than 0.1 uM
B: 0.1 uM ~ 1.0 uM
C: >1.0 uM ~ 10 uM
D: > 10 uM
Figure imgf000289_0002
Figure imgf000290_0001
Figure imgf000291_0001
Formulation Examples
The following are representative pharmaceutical formulations containing a compound of the present disclosure.
Tablet Formulation
The following ingredients are mixed intimately and pressed into single scored tablets.
Ingredient Quantity per tablet (mg) compound Formula (IA) or (I) 400 cornstarch 50 croscarmellose sodium 25 lactose 120 magnesium stearate 5 Capsule Formulation
The following ingredients are mixed intimately and loaded into a hard-shell gelatin capsule.
Ingredient Quantity per capsule (mg) compound Formula (IA) or (I) 200 lactose spray dried 148 magnesium stearate 2
Injectable Formulation
Compound of the disclosure (e.g., compound 1) in 2% HPMC, 1% Tween 80 in DI water, pH 2.2 with MSA, q.s. to at least 20 mg/mL
Inhalation Composition
To prepare a pharmaceutical composition for inhalation delivery, 20 mg of a compound disclosed herein is mixed with 50 mg of anhydrous citric acid and 100 mL of 0.9% sodium chloride solution. The mixture is incorporated into an inhalation delivery unit, such as a nebulizer, which is suitable for inhalation administration.
Topical Gel Composition
To prepare a pharmaceutical topical gel composition, 100 mg of a compound disclosed herein is mixed with 1.75 g of hydroxypropyl cellulose, 10 mL of propylene glycol, 10 mL of isopropyl myristate and 100 mL of purified alcohol USP. The resulting gel mixture is then incorporated into containers, such as tubes, which are suitable for topical administration.
Ophthalmic Solution Composition
To prepare a pharmaceutical ophthalmic solution composition, 100 mg of a compound disclosed herein is mixed with 0.9 g of NaCl in 100 mL of purified water and filtered using a 0.2 micron filter. The resulting isotonic solution is then incorporated into ophthalmic delivery units, such as eye drop containers, which are suitable for ophthalmic administration. Nasal spray solution
To prepare a pharmaceutical nasal spray solution, 10 g of a compound disclosed herein is mixed with 30 mL of a 0.05M phosphate buffer solution (pH 4.4). The solution is placed in a nasal administrator designed to deliver 100 ul of spray for each application.

Claims

What is Claimed:
1. A compound of Formula (lb):
Figure imgf000294_0001
wherein: m is 1 or 2, and n is 1, 2, or 3;
R1 is hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, hydroxy, hydroxylalkyl, alkoxyalkyl, cyano, or cyanomethyl, provided R1 is not attached to the ring -NH-;
R1a is hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, hydroxy, hydroxylalkyl, or alkoxyalkyl, provided R1a is not attached to the ring -NH-; or when R1 and R1a are attached to the carbon atoms of the ring that are opposite or diagonal to each other, then R1 and R1a can combine to form -(CH2)z- where (z is 1, 2, or 3), or -CH=CH-;
R1b is hydrogen, deuterium, alkyl, alkylidenyl, alkoxy, halo, haloalkyl, hydroxy, hydroxyalkyl, alkoxyalkyl, cyano, cyanomethyl, cyanoethyl, or 2-cyanovinyl, provided R1b is not attached to the ring -NH-;
R2 is hydrogen;
R3 is fluoro or cyclopropyl;
Rd is =CR9R10;
R9 is hydrogen, deuterium, alkyl, fluoro, or haloalkyl;
R10 is hydrogen, deuterium, alkyl, halo, haloalkyl, cyano, alkyloxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, phenyl, or heteroaryl; or R9 andR10 together with the carbon atom to which they are attached form cycloalkylene optionally substituted with alkyl, halo, alkoxy, or hydroxy; Rf is hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, or hydroxy;
R5 is -Q-R14 where Q is a bond and R14 is phenyl or naphthyl substituted with Rt Ru, Rv, and Rw wherein Rt and Ruare independently selected from hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, hydroxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, and cyano, Rv is hydrogen, alkenyl, alkynyl, cyanoalkenyl, cyanoalkynyl, or halo, and Rw is hydrogen, alkyl, alkylthio, cycloalkyl, halo, haloalkyl, haloalkoxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, cyano, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, or optionally substituted heterocyclylalkyl;
Degron is an E3 ligase ligand selected from:
(a) a group of formula (i);
Figure imgf000295_0001
(iii)
Figure imgf000296_0001
(e) a group of formula (v):
Figure imgf000296_0002
(f) a group of formula (vi):
Figure imgf000296_0003
(g) a group of formula (vii):
Figure imgf000297_0001
(h) a group of formula (viii):
Figure imgf000297_0002
(viii),
(i) a group of formula (ix):
Figure imgf000297_0003
where:
R15 and R16 are hydrogen, alkyl, cycloalkyl, or alkylcarbonyloxy;
Ya is CH or N;
Za is a bond, -CH2-, -NH-, O, or -NHC(O)- where NH of -NHC(O)- is attached to Ya; ring A is a ring of formula (a), (b), or (c):
Figure imgf000298_0001
where:
X2, X3’ and X4 are independently a bond, -alkylene-, alkynylene, -O-, -(O- alkylene)-, -(alkylene-O)-, -(NRgg-alkylene)-, -(alkylene-NR1111)- -NH-, -N(alkyl)-,
Figure imgf000298_0002
-C(=O)-, -(alkylene)-heterocyclylene-, cycloalkylene, NRjjC(=O)-, or C(=O)NRkk -, where Rgg, Rhh, Rh, and Rkk are independently hydrogen, alkyl, or cycloalkyl and each alkylene is optionally substituted with one or two fluoro; Raa, Rbb, Rcc, and Rdd are independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano;
R20 and R21 are independently hydrogen or alkyl; or R20 and R21 together with the carbon to which they are attached form >C=O; and
R22 is hydrogen or alkyl; ring B is phenylene, cyclylaminylene, a 5- or 6-membered monocyclic heteroarylene, or a 9- or 10-membered fused bicyclic heteroarylene, wherein each heteroarylene ring contains one to three nitrogen ring atoms and further wherein the phenylene, cyclylaminylene, and heteroarylene are independently substituted with Ree and Rff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano;
X1 is a bond, -alkylene-, alkynylene, -O-, -(O-alkylene)-, -(alkylene-O)-, -
(NRgg-alkylene)-, -(alkylene -NRhh)-, -NH-, -N(alkyl)-, C(=O)-, -(alkylene)-
Figure imgf000298_0003
heterocyclylene-, cycloalkylene, NRjjC(=O)-, or C(=O)NRkk-, where Rgg, Rhh, Rh, and Rkk are independently hydrogen, alkyl, or cycloalkyl and each alkylene is optionally substituted with one or two fluoro; Raa, Rbb, Rcc, and Rdd are independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano; and
R17, R18, and R19 are alkyl, hydroxyalkyl, cycloalkyl or heterocyclyl wherein cycloalkyl and heterocyclyl are substituted with Rmm selected from hydrogen, alkyl, halo, haloalkyl, alkoxyl, haloalkoxyl, cyano, alkylcarbonyl, alkylcarbonylamino, or -COR23 where R23 is alkyl, hydroxalkyl, cycloalkyl or heterocyclyl, wherein cycloalkyl and heterocyclyl are substituted with Rnn selected from hydrogen, alkyl, halo, haloalkyl, alkoxyl, haloalkoxyl, cyano, alkylcarbonyl, and alkylcarbonylamino; and
Wa is bond, O, S, or alkylene; and
L is -Z1-Z2-Z3-Z4-Z5-Z6 -where:
Z1 is -(alkylene-O)-;
Z2 is -C(O)- or -C(O)NR-;
Z3 is a bond, alkylene, alkynylene, -(alkylene -NR”)-, -(NR”-alkylene)-, -O-, -C(O)-, - NR”-, -(O-alkylene)d-, -(alkylene-O)d-, cycloalkylene, spiro cyclolalkylene, phenylene, heteroarylene, heterocyclylene, fused heterocyclylene, bridged heterocyclylene, or spiro heterocyclylene, where each ring is substituted with Ruu and Rvv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, cyanoalkyl, hydroxy, hydroxyalkyl, alkoxyalkyl, amino, alkylamino, and dialkylamino;
Z4 is a bond, alkylene, alkynylene, -C(O)NR-, -NR’(CO)-, -O-, -NR”-, -(O-alkylene)c-, -(alkylene-O)c-, cycloalkylene, spiro cyclolalkylene, phenylene, monocyclic heteroarylene, heterocyclylene, bicyclic heterocyclylene, bridged heterocyclylene, fused heterocyclylene, spiro heterocyclylene, or 11 to 13 membered spiro heterocyclylene, where each ring is substituted with Rss and Rt independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
Z5 is a bond, alkylene, alkynylene, -SO2-, -SO2NR-, -NR’SO2-, -C(O)-, -C(O)N(R)-, - NR’(CO)-, -(O-alkylene)b-, -(alkylene-O)b-,-O(CH2)7-, -O(CH2)8-, cycloalkylene, or heterocyclylene, where each ring is substituted with Rqq and Rrr independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
Z6 is a bond, alkylene, -C(O)NR-, -NR’(CO)-, -S(O)2NR-, -NR’S(O)2-, -(O-alkylene)a- , -(alkylene-O)a-, phenylene, monocyclic heteroarylene, heterocyclylene, bicyclic heterocyclylene, spiro heterocyclylene, -O-heterocyclylene-, -heterocyclylene-C(O)-, where each ring is substituted with R00 and Rpp independently selected from hydrogen, deuterium, alkyl, alkoxy, cyano, hydroxy, carboxy, halo, haloalkyl, and haloalkoxy; and where each alkylene of -Z1-Z2-Z3-Z4-Z5-Z6- is independently one to eight carbon atoms, R, R’ and R” is independently hydrogen or alkyl, each a, b, c, and d is independently an integer selected from 1 to 6, and each alkylene of -Z1-, -Z3-, -Z4-, -Z5-, and -Z6- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano; wherein when several adjacent groups of Z3 to Z6 are a bond, the adjacent groups represent one bond; or a pharmaceutically acceptable salt thereof.
2. A compound of F ormula (lb) :
Figure imgf000300_0001
wherein: m is 1 or 2, and n is 1, 2, or 3;
R1 is hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, hydroxy, hydroxylalkyl, alkoxyalkyl, cyano, or cyanomethyl, provided R1 is not attached to the ring -NH-;
R1a is hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, hydroxy, hydroxylalkyl, or alkoxyalkyl, provided R1a is not attached to the ring -NH-; or when R1 and R1a are attached to the carbon atoms of the ring that are opposite or diagonal to each other, then R1 and R1a can combine to form -(CH2)z- where (z is 1, 2, or 3), or -CH=CH-;
R1b is hydrogen, deuterium, alkyl, alkylidenyl, alkoxy, halo, haloalkyl, hydroxy, hydroxyalkyl, alkoxyalkyl, cyano, cyanomethyl, cyanoethyl, or 2-cyanovinyl, provided R1b is not attached to the ring -NH-;
R2 is hydrogen;
R3 is fluoro or cyclopropyl;
Rd is =CR9R10;
R9 is hydrogen, deuterium, alkyl, fluoro, or haloalkyl; R10 is hydrogen, deuterium, alkyl, halo, haloalkyl, cyano, alkyloxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, phenyl, or heteroaryl; or R9 andR10 together with the carbon atom to which they are attached form cycloalkylene optionally substituted with alkyl, halo, alkoxy, or hydroxy; Rf is hydrogen, alkyl, halo, alkoxy, alkoxyalkyl, or hydroxy;
R5 is -Q-R14 where Q is a bond and R14 is phenyl or naphthyl substituted with Rt Ru, Rv, and Rw wherein Rt and Ruare independently selected from hydrogen, alkyl, cycloalkyl, halo, haloalkyl, haloalkoxy, hydroxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, and cyano, Rv is hydrogen, alkenyl, alkynyl, cyanoalkenyl, cyanoalkynyl, or halo, and Rw is hydrogen, alkyl, alkylthio, cycloalkyl, halo, haloalkyl, haloalkoxy, alkoxy, heteroalkyl, hydroxyalkyl, amino, cyano, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, or optionally substituted heterocyclylalkyl;
Degron is an E3 ligase ligand selected from:
(a) a group of formula (i);
Figure imgf000301_0001
(b) a group of formula (ii);
Figure imgf000301_0002
(c) a group of formula (iii):
5
Figure imgf000302_0001
(f)
Figure imgf000303_0001
(g) a group of formula (vii):
Figure imgf000303_0002
where:
R15 and R16 are hydrogen, alkyl, cycloalkyl, or alkylcarbonyloxy;
Ya is CH or N;
Za is a bond, -CH2-, -NH-, O, or -NHC(O)- where NH of -NHC(O)- is attached to Ya; ring A is a ring of formula (a), (b), or (c):
Figure imgf000303_0003
where:
X2, X3’ and X4 are independently a bond, -alkylene-, -O-, -(O-alkylene)-, -
(alkylene-O)-, -(NRgg-alkylene)-, -(alkylene -NRhh)-, -NH-, -N(alkyl)-,
-C(=O)-, NRjjC(=O)-, or C(=O)NRkk-, where Rgg, Rhh, Rjj, and Rkk are independently hydrogen, alkyl, or cycloalkyl and each alkylene is optionally substituted with one or two fluoro; Raa, Rbb, Rcc, and Rdd are independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano;
R20 and R21 are independently hydrogen or alkyl; or R20 and R21 together with the carbon to which they are attached form >C=O; and
R22 is hydrogen or alkyl; ring B is phenylene, cyclylaminylene, a 5- or 6-membered monocyclic heteroarylene, or a 9- or 10-membered fused bicyclic heteroarylene, wherein each heteroarylene ring contains one to three nitrogen ring atoms and further wherein the phenylene, cyclylaminylene, and heteroarylene are independently substituted with Ree and Rff independently selected from hydrogen, alkyl, cycloalkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano;
X1 is a bond, alkynylene, -O-, -(O-alkylene)-, -(alkylene-O)-, -(NRgg-alkylene)-
, -(alkylene -
Figure imgf000304_0001
(alkyl)-,
-C(=O)-, NRjjC(=O)-, or C(=O)NRkk-, where Rgg, Rhh, Rjj, and Rkk are independently hydrogen, alkyl, or cycloalkyl and each alkylene is optionally substituted with one or two fluoro; Raa, Rbb, Rcc, and Rdd are independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano; and
R17, R18, and R19 are alkyl, hydroxyalkyl, cycloalkyl or heterocyclyl wherein cycloalkyl and heterocyclyl are substituted with Rmm selected from hydrogen, alkyl, halo, haloalkyl, alkoxyl, haloalkoxyl, cyano, alkylcarbonyl, alkylcarbonylamino, or -COR23 where R23 is alkyl, hydroxalkyl, cycloalkyl or heterocyclyl, wherein cycloalkyl and heterocyclyl are substituted with Rnn selected from hydrogen, alkyl, halo, haloalkyl, alkoxyl, haloalkoxyl, cyano, alkylcarbonyl, and alkylcarbonylamino; and
Wa is bond, O, S, or alkylene; and
L is -Z1-Z2-Z3-Z4-Z5-Z6- where:
Z1 is -(alkylene-O)-;
Z2 is a bond, -alkylene, -C(O)-, -C(O)NR-, phenylene, monocyclic heteroarylene, or heterocycylene, where each ring is substituted with R'™ and Rxx independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
Z3 is a bond, alkylene, alkynylene, -(alkylene -NR”)-, -(NR”-alkylene)-, -O-, -C(O)-, - NR”-, -(O-alkylene)a-, -(alkylene-O)d-, cycloalkylene, spiro cyclolalkylene, phenylene, heteroarylene, heterocyclylene, fused heterocyclylene, bridged heterocyclylene, or spiro heterocyclylene, where each ring is substituted with Ruu and Rvv independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, cyano, hydroxy, amino, alkylamino, and dialkylamino;
Z4 is a bond, alkylene, alkynylene, -C(O)NR-, -NR’(CO)-, -O-, -NR”-, -(O-alkylene)c-, -(alkylene-O)c-, cycloalkylene, spiro cyclolalkylene, phenylene, monocyclic heteroarylene, heterocyclylene, bicyclic heterocyclylene, bridged heterocyclylene, fused heterocyclylene, spiro heterocyclylene, or 11 to 13 membered spiro heterocyclylene, where each ring is substituted with Rss and Rt independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
Z5 is a bond, alkylene, alkynylene, -C(O)-, -C(O)N(R)-, -NR’(CO)-, -(O-alkylene)b-, - (alkylene-O)b-,-O(CH2)7-, -O(CH2)8-, cycloalkylene, or heterocyclylene, where each ring is substituted with Rqq and Rrr independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy;
Z6 is a bond, alkylene, -C(O)NR-, -NR’(CO)-, -S(O)2NR-, -NR’S(O)2-, -(O-alkylene)a- , -(alkylene-O)a-, phenylene, monocyclic heteroarylene, heterocyclylene, where each ring is substituted with R00 and Rpp independently selected from hydrogen, deuterium, alkyl, alkoxy, halo, haloalkyl, and haloalkoxy; and where each alkylene of -Z1-Z2-Z3-Z4-Z5-Z6- is independently one to eight carbon atoms, R, R’ and R” is independently hydrogen or alkyl, each a, b, c, and d is independently an integer selected from 1 to 6, and each alkylene of -Z1-, -Z3-, -Z4-, -Z5-, and -Z6- is substituted with Ryy and Rzz where Ryy is hydrogen or deuterium and Rzz is hydrogen, deuterium, haloalkyl, hydroxy, alkoxy, cyano, cycloalkyl, heterocyclyl, aryl, or monocyclic heteroaryl, wherein cycloalkyl, heterocyclyl, aryl, and monocyclic heteroaryl are substituted with one or two substituents independently selected from hydrogen, alkyl, alkoxy, halo, haloalkyl, haloalkoxy, and cyano; wherein when several adjacent groups of Z3 to Z6 are a bond, the adjacent groups represent one bond; or a pharmaceutically acceptable salt thereof.
3. The compound of claim 1 or 2, wherein the compound is of Fomula (lb’):
Figure imgf000306_0001
4. The compound of any one of claims 1-3, wherein m and n are each 1.
5. The compound of claim 1 or 2, wherein the compound is of Fomula (Ib-1):
Figure imgf000306_0002
(Ib-1).
6. The compound of any one of claims 1-3, wherein the compound is of Fomula (lb- 1 ’):
Figure imgf000306_0003
7. The compound of any one of claims 1 to 6, wherein R14 is:
Figure imgf000307_0001
substituted with Rt Ru, and Rw where Rv is alkynyl.
8. The compound of any one of claims 1 to 7, wherein R14 is:
Figure imgf000307_0002
9. The compound of any one of claims 1 to 8, wherein R14 is:
Figure imgf000307_0003
10. The compound of any one of claims 1 to 9, wherein R9 is hydrogen.
11. The compound of any one of claims 1 to 10, wherein R10 is hydrogen.
12. The compound of any one of claims 1 to 11, wherein Rf is hydrogen.
13. The compound of any one of claims 1 and 3 to 12, wherein:
Z2 is -C(O);
Z3 is a heterocyclylene;
Z4 is alkylene, -(O-alkylene)c, -O-, or heteroarylene;
Z6 is -C(O)NR-, heterocyclylene, phenylene, -O-heterocyclylene, or -heterocyclylene-CO-; and
Z5, X1, X2, X3, and X4 are a bond.
14. The compound of any one of claims 1 and 3 to 12, wherein:
Z2 is a -C(O);
Z3 is a heterocyclylene, spiro heterocyclylene, bridged heterocycylene, or heterocyclylene substituted with cyano, methoxy, methoxymethyl, hydroxymethyl, cyanomethyl, or 1 or 2 methyl;
Z5 is -C(O)- or -(alkylene-O)i-3;
Z6 is heterocyclylene, bicyclic heterocycylene, spiro heterocycylene, -O- heterocyclylene, or heterocyclylene substituted with carboxy, cyano, hydroxy, methyl, methoxy, or fluoro; and
Z4, X1, X2, X3, and X4 are a bond.
15. The compound of any one of claims 1 and 3 to 12, wherein:
Z2 is -C(O)-;
Z3 is heterocyclylene;
Z4 is alkylene or -O-alkylene-;
Z5 is -C(O)-;
Z6 is heterocyclylene; and
X1 , X2, X3, and X4 are a bond.
16. The compound of any one of claims 1 and 3 to 12, wherein:
Z2 is -C(O)NR-;
Z3 is a heterocyclylene;
Z5 is -C(O)- or -SO2-;
Z6 is phenylene;
X1, X2, X3, and X4 are -alkylene -heterocycylene-; and
Z4 is a bond.
17. The compound of any one of claims 1-12, wherein Z2 is -C(O)-.
18. The compound of any one of claims 1-12, wherein Z3 is -NR”-, or heterocyclene substituted with Rww and Rxx.
19. The compound of any one of claims 1-12, wherein wherein Z4 is a bond, alkylene, -(O- alkylene)c-, or -(alkylene-O)c-.
20. The compound of any one of claims 1 to 19, wherein Degron is an E3 ligase ligand selected from:
Figure imgf000309_0001
21. A pharmaceutical composition comprising a compound of any one claims 1 to 20, or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient.
22. A method of treating cancer in a patient comprising administering to the patient, a therapeutically effective amount of a compound of any one claims 1 to 20, or a pharmaceutically acceptable salt thereof; or a pharmaceutical composition of claim 21.
23. The method of claim 22, wherein the compound of any one of claims 1 to 20, or a pharmaceutically acceptable salt thereof; or the pharmaceutical composition of claim 21 is administered in combination with at least one additional anticancer agent.
PCT/US2023/032284 2022-09-08 2023-09-08 Bifunctional compounds for degrading kras g12d via ubiquitin proteasome pathway WO2024054625A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CNPCT/CN22/117697 2022-09-08
PCT/CN2022/117697 WO2024050742A1 (en) 2022-09-08 2022-09-08 Bifunctional compounds for degrading kras g12d via ubiquitin proteasome pathway
CN2023072897 2023-01-18
CNPCT/CN23/072897 2023-01-18

Publications (2)

Publication Number Publication Date
WO2024054625A2 true WO2024054625A2 (en) 2024-03-14
WO2024054625A3 WO2024054625A3 (en) 2024-04-18

Family

ID=90191765

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/032284 WO2024054625A2 (en) 2022-09-08 2023-09-08 Bifunctional compounds for degrading kras g12d via ubiquitin proteasome pathway

Country Status (1)

Country Link
WO (1) WO2024054625A2 (en)

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2021521112A (en) * 2018-04-04 2021-08-26 アルビナス・オペレーションズ・インコーポレイテッドArvinas Operations, Inc. Regulators of proteolysis and related uses
AU2019418416A1 (en) * 2019-01-03 2021-07-22 The Regents Of The University Of Michigan Estrogen receptor protein degraders
US11453683B1 (en) * 2019-08-29 2022-09-27 Mirati Therapeutics, Inc. KRas G12D inhibitors
WO2022173870A1 (en) * 2021-02-09 2022-08-18 Kumquat Biosciences Inc. Heterocyclic compounds and uses thereof

Also Published As

Publication number Publication date
WO2024054625A3 (en) 2024-04-18

Similar Documents

Publication Publication Date Title
US11518772B2 (en) Fused tricyclic ring derivatives as Src homology-2 phosphate inhibitors
WO2022187528A1 (en) Quinazoline amine derivatives as kras inhibitors
WO2022140472A1 (en) Compounds for degrading cyclin-dependent kinase 2 via ubiquitin proteosome pathway
EP4370522A1 (en) Alkylidene derivatives as kras inhibitors
WO2022187527A1 (en) Quinazoline nitrile derivatives as kras inhibitors
WO2022236578A1 (en) Exocyclic amino quinazoline derivatives as kras inhibitors
WO2024054625A2 (en) Bifunctional compounds for degrading kras g12d via ubiquitin proteasome pathway
WO2024091409A1 (en) Tricyclic derivatives as kras inhibitors
WO2024050742A1 (en) Bifunctional compounds for degrading kras g12d via ubiquitin proteasome pathway
WO2023249968A1 (en) Bifunctional compounds containing pyrido[2,3-djpyrimidin-7(8h)-one derivatives for degrading cyclin-dependent kinase 2 via ubiquitin proteasome pathway
WO2023250029A1 (en) Bifunctional compounds containing substituted pyrimidine derivatives for degrading cyclin-dependent kinase 2 via ubiquitin proteasome pathway
WO2024045066A1 (en) Alkylidene carbamate as kras inhibitors
WO2023249970A1 (en) Bifunctional compounds containing pyrimidine derivatives for degrading cyclin-dependent kinase 2 via ubiquitin proteasome pathway
WO2024102849A1 (en) Bifunctional compounds containing 2,5-substituted pyrimidine derivatives for degrading cyclin-dependent kinase 2 via ubiquitin proteasome pathway
EA044940B1 (en) DERIVATIVES OF CONDENSED TRICYCLIC RING AS PHOSPHATASE INHIBITORS WITH SRC2 HOMOLOGY DOMAIN
WO2023240024A1 (en) Sulfamide derivatives as cyclin-dependent kinase 2 inhibitors
US20230303509A1 (en) Sulfonamido derivatives as cyclin-dependent kinase 2 inhibitors

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: 23863825

Country of ref document: EP

Kind code of ref document: A2