[go: up one dir, main page]

WO2025215536A1 - Macrocyclic panras inhibitors for the treatment of cancer - Google Patents

Macrocyclic panras inhibitors for the treatment of cancer

Info

Publication number
WO2025215536A1
WO2025215536A1 PCT/IB2025/053708 IB2025053708W WO2025215536A1 WO 2025215536 A1 WO2025215536 A1 WO 2025215536A1 IB 2025053708 W IB2025053708 W IB 2025053708W WO 2025215536 A1 WO2025215536 A1 WO 2025215536A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
group
ring
formula
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
PCT/IB2025/053708
Other languages
French (fr)
Inventor
Zhuoliang Chen
Yingling CHIANG
William Francis CONNORS
Markus Reto FUREGATI
Xin Gao
Zachary Zephenia GULLEDGE
Aaron Bret HINDS
Toshio Kawanami
Marc LAFRANCE
Eric Andrew Mcneill
Henrik Moebitz
Katsumasa Nakajima
Christopher Joseph SCHWALEN
Yiping Shen
Ranny Mathew Thomas
Joseph S. WZOREK JR
Frédéric Jean ZECRI
Qiang Zhang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novartis AG
Original Assignee
Novartis AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis AG filed Critical Novartis AG
Publication of WO2025215536A1 publication Critical patent/WO2025215536A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/18Bridged systems

Definitions

  • the present disclosure relates to panRAS inhibitors.
  • the disclosure further relates to methods and compositions useful in the treatment and/or diagnosis of cancers that express 10 RAS and/or are amenable to treatment by modulating panRAS expression and/or activity, as well as methods of making those compositions.
  • Ras proteins K-Ras, H-Ras and N-Ras
  • Ras proteins play an essential role in various human cancers 15 and are therefore appropriate targets for anticancer therapy. Indeed, mutations in Ras proteins account for approximately 30% of all human cancers in the United States, many of which are fatal.
  • Ras proteins Dysregulation of Ras proteins by activating mutations, overexpression or upstream activation is common in human tumors, and activating mutations in Ras are frequently found in human cancer.
  • activating mutations at codon 12 in Ras proteins function by 20 inhibiting both GTPase-activating protein (GAP)-dependent and intrinsic hydrolysis rates of GTP, significantly skewing the population of Ras mutant proteins to the “on” (GTP-bound) state (Ras(ON)), leading to oncogenic MAPK signaling.
  • GAP GTPase-activating protein
  • Ras exhibits a picomolar affinity for GTP, enabling Ras to be activated even in the presence of low concentrations of this nucleotide.
  • the present disclosure provides, 1 PAT059646-WO-PCT 5 A compound of formula (I*) or formula (XX*), wherein 10 ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 15 N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and
  • PAT059646-WO-PCT 5 A compound of formula (I*) or formula (XX*), wherein 10 ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 15 N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-f
  • PAT059646-WO-PCT 5 A compound of formula (I*) or formula (XX*), wherein 10 ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 15 N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-f
  • the disclosure provides a pharmaceutical composition comprising a compound of the disclosure and a pharmaceutically acceptable carrier.
  • the disclosure provides a method of treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective 35 amount of a compound of the disclosure, or a pharmaceutical composition comprising a compound of the disclosure.
  • the disclosure provides methods of synthesis of a compound of the disclosure.
  • Fig.1 is a depiction of the 3-dimensional structure of Compound 101 derived from an X- 10 ray diffraction crystal study.
  • Fig.2 is a depiction of the PDB coordinates for Compound 101 derived from an X-ray diffraction crystal study.
  • DETAILED DESCRIPTION 15 [11] The disclosed compositions and methods may be understood more readily by reference to the following detailed description.
  • the present disclosure provides. 20 A compound of formula (I*) or formula (XX*), or in some embodiments a compound of formula (I) or formula (XX), PAT059646-WO-PCT 5 wherein 10 ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 15 N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of
  • a compound of formula (I*) or formula (XX*), or in some embodiments a compound of formula (I) or formula (XX), 10 PAT059646-WO-PCT 5 wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 10 N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- 15 membered heteroary
  • PAT059646-WO-PCT 5 A compound of formula (I*) or formula (XX*), wherein 10 ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 15 N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-f
  • a compound of formula (I) PAT059646-WO-PCT 5 ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 10 N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge
  • the present disclosure provides, A compound of formula (I) wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- 10 membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; 15 ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B
  • the present disclosure provides a compound of formula (I) 20 wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- 25 membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and 30 ring B is edge-fused to ring C
  • the present disclosure provides a compound of formula (I) 30 PAT059646-WO-PCT 5
  • ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted
  • ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 10 N, O, and S, and ring B is optionally substituted
  • ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted
  • ring B is edge
  • the compound is a compound of formula (II) wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 10 N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- 15 membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring C is optionally substituted; and ring A is 6-membered aryl
  • the compound is a compound of compound of formula (I) 15 wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; 20 ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 25 N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form
  • the compound is a compound of formula (II) wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 30 N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; PAT059646-WO-PCT 5 ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B
  • the compound is a compound of formula (II) PAT059646-WO-PCT 5 wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; 10 ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 15 N, O, and S, and ring C is optionally substituted; and ring C is optionally substituted; and
  • the compound is a compound of formula (Iw) or formula (XXw) wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 15 N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- 20 membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-
  • the compound is a compound of formula (Iw) 20 wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; 25 ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 30 N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to
  • ring A is phenyl, pyridine, thiazole, triazole, oxazole, or oxadiazole, and ring A is substituted with 0-3 R 10 ; wherein R 10 is selected from the group consisting of H, halo, OH, CN, C 1-6 alkyl, C 1-6 heteroalkyl, C 1-6 haloalkyl, NH 2 , and COR 6 .
  • R 10 is selected from the group consisting of H, halo, OH, CN, C 1-6 alkyl, C 1-6 heteroalkyl, C 1-6 haloalkyl, NH 2 , and COR 6 .
  • ring A is 25 [27]
  • ring A is [28]
  • R 10 is not H.
  • ring B is optionally substituted pyrrole.
  • ring C is optionally substituted phenyl.
  • the BC ring system is indole, benzothiophene, benzoxazole, or indolizine, each of which optionally has 1-3 additional N ring atoms in its 6-membered ring, wherein each BC ring system is optionally substituted, for example wherein the BC ring system is optionally substituted with 1-3 R 11 , wherein each R 11 is independently selected from the group consisting of H, halo, OH, CN, C 1-6 alkyl, C 1-6 10 heteroalkyl, C 1-6 haloalkyl, NH 2 , and COR 6 .
  • each R 11 is independently selected from the group consisting of H and halo. In some embodiments there are no R 11 substituents.
  • the BC ring system is , wherein 15 X 1 is N, S, or O; X 2 is N or CR 11 ; for example wherein either 0 or 1 of X 2 are N, and the remaining X 2 are CR 11 , for example wherein the BC ring system is , 20 wherein each R 11 is independently selected from the group consisting of H, halo, OH, CN, C 1-6 alkyl, C 1-6 heteroalkyl, C 1-6 haloalkyl, NH 2 , and COR 6 .
  • each R 11 is independently selected from the group consisting of H and halo. In some embodiments there are no R 11 substituents. In some embodiments, R 11 is not H. [33] In some embodiments, each R 1 is independently selected from the group consisting of 25 H, halo, C 1-6 alkyl, or C 1-6 haloalkyl; for example wherein each R 1 is H. [34] In some embodiments, R 2 is substituted pyridine. In some embodiments, R 2 is substituted 3-pyridyl.
  • R 2 is 1 PAT059646-WO-PCT 5 , wherein R 2a is C 1-10 alkyl, C 3-6 cycloalkyl, or C 1-10 heteroalkyl, and R 2a is optionally substituted by 1-3 substituents independently selected from the group consisting of halo, OH, CN, C 1-6 alkyl, C 1-6 heteroalkyl, -(CH 2 ) 0-2 phenyl, -(CH 2 ) 0-2 -C 3-6 cycloalkyl, C 1-6 haloalkyl, NH 2 , and COR 6 ; for example, wherein R 2a is C 1-10 optionally substituted heteroalkyl; 10 R 2b is 5-6-membered heterocycloalkyl, with 1-3 ring atoms selected from the group consisting of N, O, P(O) xx , and S(O) xx , wherein xx is 0, 1, or 2, and R 2b is optionally substitute
  • R 2 is , wherein R 2a is C 1-10 alkyl, C 3-6 cycloalkyl, or C 1-10 heteroalkyl, and R 2a is optionally substituted by 1-3 substituents independently selected from the group consisting of halo, OH, CN, C 1-6 alkyl, C 1-6 heteroalkyl, -(CH 2 ) 0-2 phenyl, -(CH 2 ) 0-2 -C 3-6 cycloalkyl, C 1-6 haloalkyl, NH 2 , NR x COR 6 , and COR 6 ; 25 for example, wherein R 2a is C 1-10 optionally substituted heteroalkyl; R 2b is 4-10-membered heterocycloalkyl, with 1-4 ring atoms selected from the group consisting of N, O, P(O) xx , and S(O) xx , wherein xx is 0, 1, or 2, and R 2b is optionally substituted,
  • R 2b is 10 wherein X 3 is C, CR x , N, or P(O); is a single or double bond; and each R x is independently selected from H, C 1-6 alkyl, C 3-6 cycloalkyl, and C 1-6 haloalkyl; for example, wherein R 2ba is H, C 1-6 alkyl, -(CH 2 ) 0-2 phenyl, C 1-6 heteroalkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, COR 6 ,or heterocycloalkyl wherein the heterocycloalkyl is 4-7 membered with 1-3 ring 15 atoms selected from the group consisting of N, O, and S, and each phenyl, cycloalkyl, or heterocycloalkyl is optionally substituted.
  • R 2b is wherein X 3 is CH, N, or P(O); and 20 is a single or double bond; for example, wherein R 2ba is H, C 1-6 alkyl, -(CH 2 ) 0-2 phenyl, C 1-6 heteroalkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, COR 6 ., or heterocycloalkyl wherein the heterocycloalkyl is 4-7 membered with 1-3 ring atoms selected from the group consisting of N, O, and S.
  • R 2b is PAT059646-WO-PCT 5 for example, wherein R 2ba is H, C1-6 alkyl, -(CH2)0-2 phenyl, C1-6 heteroalkyl, C1-6 haloalkyl, C3-6 cycloalkyl, COR 6 ., or heterocycloalkyl wherein the heterocycloalkyl is 4-7 membered with 1-3 ring atoms selected from the group consisting of N, O, and S.
  • R 2a is 10 , wherein X 4 is O or N, and X 5 is CH; or X 5 is O or N, and X 4 is CH; and R 2aa is C 1-3 alkyl or C 3-6 cycloalkyl, each optionally substituted with 1-3 halo, or R 2aa is absent or H; 15 R 2ab is C 1-3 alkyl or C 3-6 cycloalkyl, each optionally substituted with 1-3 halo, or R 2ab is absent or H; and R 2ac is C 1-3 alkyl or C 3-6 cycloalkyl, each optionally substituted with 1-3 halo or one OH, or R 2ac is absent or H.
  • R 2a is 20 , wherein X 4 is O or N, and X 5 is CH; or X 5 is O or N, and X 4 is CH; and R 2aa is C 1-3 alkyl optionally substituted with 1-3 halo, or R 2aa is absent or H; R 2ab is C 1-3 alkyl optionally substituted with 1-3 halo, or R 2ab is absent or H; and 25 R 2ac is C 1-3 alkyl optionally substituted with 1-3 halo or one OH.
  • R 2 is PAT059646-WO-PCT 5
  • X 3 is C, CR x , N, or P(O);
  • X 4 is O or N, and X 5 is CH; or
  • X 5 is O or N, and X 4 is CH; and is a single or double bond
  • R 2aa is C 1-3 alkyl or C 3-6 cycloalkyl, each optionally substituted with 1-3 halo, or R 2aa is absent or 10 H
  • R 2ab is C 1-3 alkyl or C 3-6 cycloalkyl, each optionally substituted with 1-3 halo, or R 2ab is absent or H
  • R 2ac is C 1-3 alkyl or C 3-6 cycloalkyl, each optionally substituted with 1-3 halo or one OH
  • R 2ba is H, C 1-6 alkyl, -(CH 2 ) 0-2 phenyl, C 1-6 heteroalkyl, C 1-6 haloal
  • R 2 is 20 , wherein X 3 is CH, N, or P(O); X 4 is O or N, and X 5 is CH; or X 5 is O or N, and X 4 is CH; and is a single or double bond 25
  • R 2aa is C 1-3 alkyl optionally substituted with 1-3 halo, or R 2aa is absent or H
  • R 2ab is C 1-3 alkyl optionally substituted with 1-3 halo, or R 2ab is absent or H
  • R 2ac is C1-3 alkyl optionally substituted with 1-3 halo or one OH
  • R 2ba is H, C 1-6 alkyl, -(CH 2 ) 0-2 phenyl, C 1-6 heteroalkyl, C 1-6 haloalkyl, or COR 6 .
  • R 2 is PAT059646-WO-PCT 5
  • R 2aa is C 1-3 alkyl optionally substituted with 1-3 halo, or R 2aa is absent;
  • R 2ab is C 1-3 alkyl optionally substituted with 1-3 halo, or R 2aa is absent;
  • R 2ac is C 1-3 alkyl optionally substituted with 1-3 halo;
  • R 2ba is H, C1-6 alkyl, -(CH2)0-2 phenyl, C1-6 heteroalkyl, C1-6 haloalkyl, C3-6 cycloalkyl, COR 6 ., or heterocycloalkyl wherein the heterocycloalkyl is 4-7 membered with 1-3 ring atoms selected from the group consisting of N, O, and S, and each phenyl, cycloalkyl, or heterocycloalkyl is 15 optionally substituted.
  • R 2a is [46] In some embodiments, R 2ba is C1-6 alkyl. 20 [47] In some embodiments, R 3 is selected from the group consisting of H, C 1-6 alkyl, C 1-6 heteroalkyl, and C 1-6 haloalkyl, or R 3 is absent; for example, R 3 is C 1-6 alkyl. In some embodiments, R 3 is methyl or ethyl. In some embodiments, R 3 is ethyl. [48] In some embodiments, each R 4 is independently selected from the group consisting of H, C 1-6 alkyl, and C 1-6 haloalkyl; for example, each R 4 is H.
  • each R 5 is independently selected from H and C 1-6 alkyl; or two R 5 groups are taken together with the carbon atom to which they are connected to form a C 3-6 cycloalkyl. In some embodiments, each R 5 is independently selected from H and C 1-6 alkyl; for example both R 5 are CH 3 .
  • L is absent, or L is -COCHR 7 N(R 8 )-*, wherein * indicates the 30 point of attachment to W;
  • R 7 is C 1-6 alkyl, C 2-6 alkenyl, or C 2-6 alkynyl, each optionally substituted by C 3-6 cycloalkyl; or R 7 is C 3-6 cycloalkyl;
  • PAT059646-WO-PCT 5 wherein R 7 is optionally further substituted by 1-3 substituents selected from the group consisting of halo, OH, C 1-3 haloalkyl, C 1-3 heteroalkyl, and C 1-3 alkyl; and
  • R 8 is H or C 1-3 alkyl.
  • R 7 is C 1-6 alkyl, C 1-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, or C 1-6 alkyl substituted by C 3-6 cycloalkyl, wherein R 7 is optionally further substituted by 1-3 10 substituents selected from the group consisting of halo and C 1-3 alkyl.
  • R 7 is C1-6 alkyl, C2-6 alkenyl, or C3-6 cycloalkyl, wherein R 7 is optionally further substituted by 1-3 substituents selected from the group consisting of halo and C 1-3 alkyl.
  • R 7 is C 2-6 alkenyl, C 2-6 alkynyl, C 4-6 cycloalkenyl, or C 1-6 alkyl substituted by C 4-6 cycloalkenyl; wherein R 7 is optionally further substituted by 1-3 substituents 15 selected from the group consisting of halo and C 1-3 alkyl.
  • R 7 is 20 [54] In some embodiments, R 7 is 25 [55] In some embodiments, L is PAT059646-WO-PCT 5 r absent; for example, wherein R 8 is CH 3 .
  • L is 10 In some embodiments, L is [57] In some embodiments, 15 W is -COR 9 ; R 9 is C 3-7 cycloalkyl or 4-6 membered heterocycloalkyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, wherein each R 9 is optionally substituted by 1-3 R 9a , and optionally substituted with 1 R 9b on a ring nitrogen atom, if present; each R 9a is independently selected from the group consisting of halo, OH, CN, C 1-6 alkyl, C 1-6 20 heteroalkyl, -(CH 2 ) 0-2 phenyl, -(CH 2 ) 0-2 -C 3-6 cycloalkyl, C 1-6 haloalkyl, alkenyl, C 1-6 alkynyl, NH 2 , PAT059646-WO-PCT 5 COR 6 , NR x COR 6 , and -(CH
  • 10 W is -COR 9 ;
  • R 9 is C 3-7 cycloalkyl or 4-6 membered heterocycloalkyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, wherein each R 9 is optionally substituted by 1-3 R 9a ;
  • each R 9a is independently selected from the group consisting of halo, OH, CN, C 1-6 alkyl, C 1-6 15 heteroalkyl, -(CH 2 ) 0-2 phenyl, -(CH 2 ) 0-2 -C 3-6 cycloalkyl, C 1-6 haloalkyl, NH 2 , COR 6 , and -(CH 2 ) 0-2 heterocyclyl of 4-7 ring atoms with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and wherein each phenyl, heterocyclyl and cycloalkyl is optionally substituted, for example with 1-3
  • R 9 is oxetane or pyrrolidine, each optionally substituted. In some embodiments, R 9 is optionally substituted oxetane. In some embodiments, R 9 is oxetane optionally substituted with 1-3 R 9a . In some embodiments, R 9 is 2-oxetanyl optionally substituted with 1-3 R 9a . In some embodiments R 9a is optionally substituted phenyl.
  • W is PAT059646-WO-PCT 5 , wherein R 9b is H, C 1-6 alkyl, C 1-6 heteroalkyl, -(CH 2 ) 0-2 phenyl, -(CH 2 ) 0-2 -C 3-6 cycloalkyl, C 1-6 haloalkyl, NH 2 , COR 6 , NR x COR 6 , and -(CH 2 ) 0-2 heterocyclyl of 4-7 ring atoms with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and wherein each 10 phenyl, heterocyclyl and cycloalkyl is optionally substituted, for example with 1-3 substituents selected from the group consisting of halo, OH, CN, C 1-6 alkyl, C 1-6 haloalkyl, and C 1-6 heteroalkyl, for example wherein R 9b is H, C 1-3 alkyl, or
  • W is 15 wherein R 9b is H or C 1-3 alkyl.
  • W is PAT059646-WO-PCT 5 wherein R 9b is H or C 1-3 alkyl; for example wherein R 9a is optionally substituted phenyl.
  • each R9 a’ is independently selected from the group consisting of halo, OH, CN, C 1-6 alkyl, C 1-6 heteroalkyl, -(CH 2 ) 0-2 phenyl, -(CH 2 ) 0-2 -C 3-6 cycloalkyl, C 1-6 haloalkyl, NH 2 , COR 6 , and -(CH 2 ) 0-2 heterocyclyl of 4-7 ring atoms with 1-3 ring atoms independently selected from the group 15 consisting of N, O, and S, and wherein each phenyl, heterocyclyl and cycloalkyl is optionally substituted, for example with 1-3 substituents selected from the group consisting of halo, OH, CN, C 1-6 alkyl, C 1-6 haloalkyl, and C 1-6 heteroalkyl.
  • R 9a is optionally substituted phenyl.
  • W is 20 PAT059646-WO-PCT 5 In some embodiments, W is . 10 In some embodiments, W is , 15 In some embodiments, W is PAT059646-WO-PCT 5 [62]
  • the compound is a compound of formula (III), formula (XXIII), or formula (XXXIII) 10 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H, for example wherein n is 0 or 1; for example a compound of formula (IIIa) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H.
  • the compound is a compound of formula (IV), formula (XXIV), or formula (XXXIV) 10 PAT059646-WO-PCT 5 wherein n is 0 or 1; wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H; for example, wherein there are no R 11 substituents; 10 for example, wherein the compound is a compound of formula (IVa) wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H; for example, wherein there are no R 11 substituents.
  • the compound is a compound of formula (V), formula (VI), formula (XXV), formula (XXVI), formula (XXXV), or formula (XXXVI)
  • PAT059646-WO-PCT 5 10 wherein n is 0 or 1; wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H; for example, wherein all R 5 are CH 3 ; for example, wherein there are no R 11 substituents; 15 for example, a compound of formula (V), formula (XXV), or formula (XXXV) wherein there is one R 10 selected from the group consisting of OH, NH 2 , and halo, and the remaining R 10 are H; for example, a compound of formula (VI), formula (XXVI), or formula (XXXVI) wherein R 10 is H.
  • the compound is a compound of formula (Vw) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure, for example, wherein m is 0, and n is 0 or 1; for example, wherein all R 1 are H; for example, wherein all R 5 are CH 3 ; for example, wherein there are no R 11 substituents; 10 for example, a compound of formula (Vw) wherein there is one R 10 selected from the group consisting of OH, NH 2 , and halo, and the remaining R 10 are H.
  • the compound is a compound of formula (Va) or formula (VIa) 15 are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H; for example, wherein all R 5 are CH 3 ; for example, wherein there are no R 11 substituents; for example, a compound of formula (Va) wherein there is one R 10 selected from the group consisting of OH, NH 2 , and halo, and the remaining R 10 are H; 20 for example, a compound of formula (VIa) wherein R 10 is H.
  • the compound is a compound of formula (VII), formula (XXVII), or formula (XXXVII) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure; 10 for example, wherein all R 1 are H.
  • the compound is a compound of formula (Iy’) or formula (XXy’) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H, for example wherein m is 1 and n is 0 or 1.
  • the compound is a compound of formula (Iy) or formula (XXy) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H, for example wherein m is 1 and n is 0 or 1.
  • the compound is a compound of formula (Iyz) 10 wherein the variables are defined as in various embodiments of the disclosure, and R 9a is not H; for example, wherein all R 1 are H, for example wherein m is 1 and n is 0 or 1.
  • the compound is a compound of formula (IIIy’), formula (XXIIIy’), 15 or formula (XXXIIIy’) PAT059646-WO-PCT 5 10 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H, for example wherein n is 0 or 1; for example a compound of formula (IIIya’) 1 PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H.
  • the compound is a compound of formula (IIIy’), formula (XXIIIy’), 10 or formula (XXXIIIy’) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H, for example wherein n is 0 or 1; 10 for example a compound of formula (IIIya’) wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H.
  • the compound is a compound of formula (IIIy), formula (XXIIIy), or formula (XXXIIIy) PAT059646-WO-PCT 5 ); 10 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H, for example wherein n is 0 or 1; for example a compound of formula (IIIya) 15 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H.
  • the compound is a compound of formula (IIIyz), formula (XXIIIyz), or formula (XXXIIIyz) PAT059646-WO-PCT 5 10 wherein the variables are defined as in various embodiments of the disclosure, and R 9a is not H; for example, wherein all R 1 are H, for example wherein n is 0 or 1; for example a compound of formula (IIIyza) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure, and R 9a is not H; for example, wherein all R 1 are H.
  • the compound is a compound of formula (IVy’), formula (XXIVy’), 10 or formula (XXXIVy’) 15 PAT059646-WO-PCT 5 wherein n is 0 or 1; wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H; for example, wherein there are no R 11 substituents; for example, wherein the compound is a compound of formula (IVya’) 10 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H; for example, wherein there are no R 11 substituents.
  • the compound is a compound of formula (IVy), formula (XXIVy), or formula (XXXIVy) 15 PAT059646-WO-PCT 5 wherein n is 0 or 1; wherein the variables are defined as in various embodiments of the 10 disclosure; for example, wherein all R 1 are H; for example, wherein there are no R 11 substituents; for example, wherein the compound is a compound of formula (IVya) 15 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H; for example, wherein there are no R 11 substituents.
  • the compound is a compound of formula (IVyz), formula (XXIVyz), or formula (XXXIVyz) PAT059646-WO-PCT 5 10 wherein n is 0 or 1; wherein the variables are defined as in various embodiments of the disclosure, and R 9a is not H; for example, wherein all R 1 are H; for example, wherein there are no R 11 substituents; for example, wherein the compound is a compound of formula (IVyza) PAT059646-WO-PCT 5 (IVyza); wherein the variables are defined as in various embodiments of the disclosure, and R 9a is not H; for example, wherein all R 1 are H; for example, wherein there are no R 11 substituents.
  • the compound is a compound of formula (Vy’) or formula (VIy’), formula (XXVy’), formula (XXVIy’), formula (XXXVIy’) or formula (XXXVIy’) 10
  • PAT059646-WO-PCT 5 10 wherein n is 0 or 1; wherein the variables are defined as in various embodiments of the disclosure; 1 PAT059646-WO-PCT 5 for example, wherein all R 1 are H; for example, wherein all R 5 are CH3; for example, wherein there are no R 11 substituents; for example, a compound of formula (Vy’), formula (XXVy’), or formula (XXXVy’) wherein there is one R 10 selected from the group consisting of OH, NH 2 , and halo, and the remaining R 10 are H; 10 for example, a compound of formula (VIy’), formula (XXVIy’), or formula (XXXVIy’) wherein R 10 is H.
  • the compound is a compound of formula (Vya’) or formula (Viya’) 15 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H; for example, wherein all R 5 are CH 3 ; for example, wherein 20 there are no R 11 substituents; for example, a compound of formula (Vya’) wherein there is one R 10 selected from the group consisting of OH, NH 2 , and halo, and the remaining R 10 are H; for example, a compound of formula (Viya’) wherein R 10 is H.
  • the compound is a compound of formula (Vy) or formula (VIy), 25 formula (XXVy), formula (XXVIy), formula (XXXVIy) or formula (XXXVIy)
  • n is 0 or 1; wherein the variables are defined as in various embodiments of the 10 disclosure; for example, wherein all R 1 are H; for example, wherein all R 5 are CH 3 ; for example, wherein there are no R 11 substituents; for example, a compound of formula (Vy), formula (XXVy), or formula (XXXVy) wherein there is one R 10 selected from the group consisting of OH, NH 2 , and halo, and the remaining R 10 are H; 15 for example, a compound of formula (VIy), formula (XXVIy), or formula (XXXVIy) wherein R 10 is H.
  • the compound is a compound of formula (Vya) or formula (VIya)
  • PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R 1 are H; for example, wherein all R 5 are CH3; for example, wherein 10 there are no R 11 substituents; for example, a compound of formula (Vya) wherein there is one R 10 selected from the group consisting of OH, NH 2 , and halo, and the remaining R 10 are H; for example, a compound of formula (VIya) wherein R 10 is H.
  • the compound is a compound of formula (Vyz) or formula (VIyz), 15 formula (XXVyz), formula (XXVIyz), formula (XXXVyz), or formula (XXXVIyz) PAT059646-WO-PCT 5 10 wherein n is 0 or 1; wherein the variables are defined as in various embodiments of the disclosure, and R 9a is not H; for example, wherein all R 1 are H; for example, wherein all R 5 are CH3; for example, wherein there are no R 11 substituents; PAT059646-WO-PCT 5 for example, wherein R 9a is optionally subsituted phenyl; for example, a compound of formula (Vyz), formula (XXVyz), or formula (XXXVyz) wherein there is one R 10 selected from the group consisting of OH, NH 2 , and halo, and the remaining R 10 are H; for example, a compound of formula (VIyz), 15 formula (XX
  • the compound is a compound of formula (Vyz*) 15 for example, wherein m is 0 and n is 0 or 1; wherein the variables are defined as in various embodiments of the disclosure, and R 9a is not H; for example, wherein all R 1 are H; for example, wherein all R 5 are CH 3 ; for example, wherein there are no R 11 substituents; for example, wherein R 9a is optionally subsituted phenyl; 20 for example, wherein there is one R 10 selected from the group consisting of OH, NH 2 , and halo, and the remaining R 10 are H.
  • Vyz* formula
  • the compound is a compound of formula (Vyza) or formula (VIyza) PAT059646-WO-PCT 5 embodiments of the disclosure, and R 9a is not H; for example, wherein all R 1 are H; for example, wherein all R 5 are CH 3 ; for example, wherein there are no R 11 substituents; for example, wherein R 9a is optionally subsituted phenyl; 10 for example, a compound of formula (Vyza) wherein there is one R 10 selected from the group consisting of OH, NH 2 , and halo, and the remaining R 10 are H; for example, a compound of formula (VIyza) wherein R 10 is H.
  • the compound is a compound of formula (VIIy), formula (VIIIy), 15 formula (XXVIIy), formula (XXVIIIy), formula (XXXVIIy), or formula (XXXVIIIy)
  • the compound is a compound of formula (VIIyz) (VIIyz); wherein the variables are defined as in various embodiments of the disclosure, and R 9a is not H; for example, wherein R 9a is optionally subsituted phenyl; for example, wherein all R 1 are H. 15 [85] In some embodiments, the compound is a compound of formula (Iw) or formula (XXw) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure.
  • the compound is a compound of formula (Iw*) or formula (XXw*) 10 1 PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure.
  • L is -COCHR 7 N(R 8 )-*, wherein * indicates the point of attachment to W; 10 R 7 is C 2-6 alkenyl, C 2-6 alkynyl, C 4-6 cycloalkenyl, or C 1-6 alkyl substituted by C 4-6 cycloalkenyl; wherein R 7 is optionally further substituted; and R 8 is H, C 1-6 alkyl, C 3-6 cycloalkyl, or C 1-6 haloalkyl; for example wherein R 7 is 15 for example wherein R 7 is for example wherein R 7 is .
  • the compound is selected from the compounds disclosed in the 20 specification. In some embodiments, the compound is selected from the compounds in Table 20. In some embodiments, the compound is selected from the group consisting of the following compound numbers: 103, 106, 107, 109, 112, 116, 117, 122, 123, 124, 125, 126, 130, 132, 133, 134, 135, 136, 137, 138, 139, 140, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 157, 158, 159, 160, 161, and 162.
  • the compound is selected from the group 25 consisting of the following compound numbers: 103, 106, 107, 109, 112, 116, 117, 122, 123, PAT059646-WO-PCT 5 130, 132, 133, 134, 135, 136, 137, 138, 139, 140, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 157, 158, 159, 160, 161, and 162.
  • the compound is selected from the group consisting of the following compound numbers: 112, 116, 122, 123, 132, 133, 134, 135, 136, 137, 138, 139, 140, 147, 148, 149, 150, 151, 152, 153, 154, 155, 157, 159, 160, 161, and 162. 10 [89] In some embodiments, any of the compounds disclosed herein may be in the form of a pharmaceutically acceptable salt. [90] In another aspect, disclosed is a pharmaceutical composition comprising a compound of the disclosure and a pharmaceutically acceptable carrier.
  • the cancer is a tumor or a hematological cancer
  • the cancer is a breast cancer including ER positive breast cancer, multiple myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, 20 bladder cancer, brain cancer, bone marrow cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal cancer, hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant prostate cancer, bladder urothelial carcinoma, mel
  • the cancer is non-small cell lung cancer, pancreatic cancer, colorectal cancer, melanoma, head and neck cancer, acute myeloid leukemia, and bladder cancer.
  • the descriptions refer to compositions and methods of using the compositions. Where the disclosure describes or claims a feature or embodiment associated with a composition, such a feature or embodiment is equally applicable to the methods of using the composition. Likewise, where the disclosure describes or claims a feature or embodiment associated with a method of using a composition, such a feature or embodiment is equally 35 applicable to the composition. [95] When a range of values is expressed, it includes embodiments using any particular value within the range. Further, reference to values stated in ranges includes each and every value within that range.
  • compositions and methods that are, for brevity, described in the context of a single embodiment, may also be provided separately or in any sub-combination.
  • Any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds.
  • lsotopically labeled compounds have structures 15 depicted by the formulae given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number.
  • Isotopes that can be incorporated into compounds disclosed herein include, for example, isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine, and chlorine, such as 3 H, 11 C, 13 C, 14 C, 15 N, 18 F, and 36 Cl.
  • the present disclosure includes compounds that incorporate one or more of 20 any of the aforementioned isotopes, including for example, radioactive isotopes, such as 3 H and 1 4 C, or those into which non-radioactive isotopes, such as 2 H and 13 C are present.
  • isotopically labelled compounds are useful in metabolic studies (with 14 C), reaction kinetic studies (with, for example 2 H or 3 H), detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or 25 substrate tissue distribution assays, or in radioactive treatment of patients.
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • an 18 F or labeled compound may be particularly desirable for PET or SPECT studies.
  • Isotopically- labeled compounds can generally be prepared by conventional techniques known to those skilled in the art, e.g., using an appropriate isotopically-labeled reagents in place of the non- labeled reagent previously employed.
  • 30 Definitions [98] Various terms relating to aspects of the description are used throughout the specification and claims. Such terms are to be given their ordinary meaning in the art unless otherwise indicated. Other specifically defined terms are to be construed in a manner consistent with the definitions provided herein. 35 [99] As used herein, the singular forms “a,” “an,” and “the” include plural forms unless the context clearly dictates otherwise.
  • the term “about” refers to a range of values which are 15 1% more or less than the specified value.
  • agent is used herein to refer to a chemical compound, a mixture of chemical compounds, a biological macromolecule, an extract made from biological materials, or a combination of two or more thereof.
  • therapeutic agent or “drug” refers to an agent that is capable of modulating a biological process and/or has biological activity.
  • panRAS 20 inhibitors as described herein, are exemplary therapeutic agents.
  • chemotherapeutic agent or “anti-cancer agent” is used herein to refer to all agents that are effective in treating cancer (regardless of mechanism of action).
  • chemotherapeutic agents include antibodies, biological molecules, and small molecules, and 25 encompass the panRAS inhibitors, as described herein.
  • a chemotherapeutic agent may be a cytotoxic or cytostatic agent.
  • cytostatic agent refers to an agent that inhibits or suppresses cell growth and/or multiplication of cells.
  • cytotoxic agent refers to a substance that causes cell death primarily by interfering with a cell’s expression activity and/or functioning.
  • Ras Sarcoma Virus refers to any native form of the human Ras protein family (e.g., K-Ras (including splice variants KRAS4A and KRAS4B), H-Ras and N-Ras).
  • K-Ras including splice variants KRAS4A and KRAS4B
  • H-Ras and N-Ras.
  • the term encompasses full-length human K-Ras (Kristen Rat Sarcoma Virus), N-Ras (Neuroblastoma Rat Sarcoma Virus) as well as any form of human Ras that may result from cellular processing.
  • the term also encompasses functional variants or 35 fragments of human Ras proteins, including but not limited to splice variants, allelic variants, and isoforms that retain one or more biologic functions of human Ras proteins (i.e., variants and fragments are encompassed unless the context indicates that the term is used to refer to the wild-type protein only).
  • Ras proteins can be isolated from human, or may be produced recombinantly or by synthetic methods. 40 [104]
  • the term "inhibit” or “inhibition” or “inhibiting,” as used herein, means to reduce a biological activity or process by a measurable amount, and can include but does not require PAT059646-WO-PCT 5 complete prevention or inhibition.
  • panRAS inhibitor refers to an agent capable of reducing the expression and/or activity of panRAS (e.g., K-Ras (including splice variants KRAS4A and 10 KRAS4B), H-Ras and N-Ras) and/or one or more upstream modulators or downstream targets thereof.
  • K-Ras including splice variants KRAS4A and 10 KRAS4B
  • panRAS modulators including exemplary inhibitors of panRAS are described in WO2021/091956 or WO2022/060836.
  • cancer refers to the presence of cells possessing characteristics typical of cancer-causing cells, such as uncontrolled proliferation, immortality, 15 metastatic potential, rapid growth and proliferation rate, and/or certain morphological features. Often, cancer cells can be in the form of a tumor or mass, but such cells may exist alone within a subject, or may circulate in the blood stream as independent cells, such as leukemic or lymphoma cells.
  • cancer includes all types of cancers and cancer metastases, including hematological cancers, solid tumors, sarcomas, carcinomas and other solid and non- 20 solid tumor cancers.
  • Hematological cancers may include B-cell malignancies, cancers of the blood (leukemias), cancers of plasma cells (myelomas, e.g., multiple myeloma), or cancers of the lymph nodes (lymphomas).
  • B-cell malignancies include chronic lymphocytic leukemia (CLL), follicular lymphoma, mantle cell lymphoma, and diffuse large B-cell lymphoma.
  • Leukemias may include acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), 25 chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myelomonocytic leukemia (CMML), acute monocytic leukemia (AMoL), etc.
  • ALL acute lymphoblastic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • CML chronic myelogenous leukemia
  • CMML chronic myelomonocytic leukemia
  • AoL acute monocytic leukemia
  • Lymphomas may include Hodgkin's lymphoma, non-Hodgkin's lymphoma, etc.
  • Other hematologic cancers may include myelodysplasia syndrome (MDS).
  • Solid tumors may 30 include carcinomas such as adenocarcinoma, e.g., a breast cancer including ER positive breast cancer, multiple myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, bladder cancer, brain cancer, bone marrow cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal cancer, hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, 35 follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant prostate cancer, bladder urothelial carcinoma, melanoma, myelogenous leukemia, myeloma, oral cancer, ovarian cancer, non-small cell lung cancer, prostate cancer, small cell lung cancer, spleen cancer, or head and neck cancer.
  • carcinomas
  • the term “tumor” refers to any mass of tissue that results from excessive 40 cell growth or proliferation, either benign or malignant, including precancerous lesions.
  • the tumor is a breast cancer including ER positive breast cancer, multiple PAT059646-WO-PCT 5 myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, bladder cancer, brain cancer, bone marrow cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal cancer, hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant 10 prostate cancer, bladder urothelial carcinoma, melanoma, myelogenous leukemia, myeloma,
  • tumor cell and “cancer cell” may be used interchangeably herein and refer to individual cells or the total population of cells derived from a tumor or cancer, including both 15 non-tumorigenic cells and cancer stem cells.
  • tumor cell and “cancer cell” will be modified by the term “non-tumorigenic” when referring solely to those cells lacking the capacity to renew and differentiate to distinguish those cells from cancer stem cells.
  • subject and “patient” are used interchangeably herein to refer to any human or non-human animal in need of treatment. Non-human animals include all vertebrates (e.g., 20 mammals and non-mammals) such as any mammal.
  • Non-limiting examples of mammals include humans, chimpanzees, apes, monkeys, cattle, horses, sheep, goats, swine, rabbits, dogs, cats, rats, mice, and guinea pigs.
  • Non-limiting examples of non-mammals include birds and fish.
  • the subject is a human.
  • the term “a subject in need of treatment,” as used herein, refers to a subject that would 25 benefit biologically, medically, or in quality of life from a treatment (e.g., a treatment with any one or more of the exemplary compounds described herein).
  • treatment refers to any improvement of any consequence of disease, disorder, or condition, such as prolonged survival, less morbidity, and/or a lessening of side effects which result from an alternative therapeutic modality.
  • treatment comprises delaying or ameliorating a disease, disorder, or condition (i.e., slowing or arresting or reducing the development of a disease or at least one of the clinical symptoms thereof).
  • treatment comprises delaying, alleviating, or ameliorating at least one physical parameter of a disease, disorder, or condition, including those which may not be discernible by the patient.
  • treatment comprises 35 modulating a disease, disorder, or condition, either physically (e.g., stabilization of a discernible symptom), physiologically (e.g., stabilization of a physical parameter), or both.
  • treatment comprises administration of a described compound or composition to a subject, e.g., a patient, to obtain a treatment benefit enumerated herein.
  • the treatment can be to cure, heal, alleviate, delay, prevent, relieve, alter, remedy, ameliorate, palliate, improve, or 40 affect a disease, disorder, or condition (e.g., a cancer), the symptoms of a disease, disorder, or condition (e.g., a cancer), or a predisposition toward a disease, disorder, or condition (e.g., a PAT059646-WO-PCT 5 cancer).
  • a composition disclosed herein in addition to treating a subject having a disease, disorder, or condition, can also be provided prophylactically to prevent or reduce the likelihood of developing that disease, disorder, or condition.
  • a "pharmaceutical composition” refers to a preparation of a composition, e.g., a panRAS inhibitor compound or composition, in addition to at least one other (and optionally more than one other) component suitable for administration to a subject, such as a pharmaceutically acceptable carrier, stabilizer, diluent, dispersing agent, suspending agent, 15 thickening agent, and/or excipient.
  • compositions provided herein are in such form as to permit administration and subsequently provide the intended biological activity of the active ingredient(s) and/or to achieve a therapeutic effect.
  • the pharmaceutical compositions provided herein preferably contain no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
  • pharmaceutically acceptable carrier and “physiologically acceptable carrier,” which may be used interchangeably, refer to a carrier or a diluent that does not cause significant irritation to a subject and does not abrogate the biological activity and properties of the administered compound or composition and/or any additional therapeutic agent in the composition.
  • Pharmaceutically acceptable carriers may enhance or stabilize the 25 composition or can be used to facilitate preparation of the composition.
  • Pharmaceutically acceptable carriers can include solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drug stabilizers, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, and the like and combinations thereof, as 30 would be known to those skilled in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, pp.1289- 1329). Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the therapeutic or pharmaceutical compositions is contemplated.
  • preservatives e.g., antibacterial agents, antifungal agents
  • isotonic agents e.g., absorption delaying agents, salts, preservatives, drug stabilizers, binders, excipients, disintegration agents, lubricants, sweeten
  • the carrier may be selected to minimize adverse side effects in the subject, and/or to minimize degradation of the active ingredient(s). 35 An adjuvant may also be included in any of these formulations.
  • excipient refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient.
  • Formulations for parenteral administration can, for example, contain excipients such as sterile water or saline, polyalkylene glycols such as polyethylene glycol, vegetable oils, or 40 hydrogenated napthalenes.
  • excipients include, but are not limited to, calcium bicarbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, PAT059646-WO-PCT 5 gelatin, ethylene-vinyl acetate co-polymer particles, and surfactants, including, for example, polysorbate 20.
  • pharmaceutically acceptable salt refers to a salt which does not abrogate the biological activity and properties of the compounds disclosed herein, and does not cause significant irritation to a subject to which it is administered.
  • salts 10 include, but are not limited to: (a) acid addition salts formed with inorganic acids, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid and the like; and salts formed with organic acids, for example, acetic acid, oxalic acid, tartaric acid, succinic acid, maleic acid, fumaric acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, tannic acid, palmitic acid, alginic acid, polyglutamic acid, naphthalenesulfonic acid, 15 methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfonic acid, polygalacturonic acid, and the like; and (b) salts formed from elemental anions such as chlorine, bromine, and iodine.
  • inorganic acids for example, hydrochloric acid, hydrobromic acid, sulfuric acid,
  • the compounds described herein can contain a monovalent anionic counterion M 1 -. Any suitable anionic counterion can be used. In certain embodiments, the monovalent anionic counterion is a pharmaceutically acceptable monovalent anionic counterion.
  • the monovalent anionic 25 counterion M 1 - can be selected from bromide, chloride, iodide, acetate, trifluoroacetate, benzoate, mesylate, tosylate, triflate, formate, or the like. In some embodiments, the monovalent anionic counterion M 1 - is trifluoroacetate or formate.
  • the term “therapeutically effective amount” or “therapeutically effective dose,” refers to an amount of a compound described herein, e.g., a compound or composition 30 described herein, to effect the desired therapeutic result (i.e., reduction or inhibition of an enzyme or a protein activity, amelioration of symptoms, alleviation of symptoms or conditions, delay of disease progression, a reduction in tumor size, inhibition of tumor growth, prevention of metastasis).
  • a therapeutically effective amount does not induce or cause undesirable side effects.
  • a therapeutically effective amount 35 induces or causes side effects but only those that are acceptable by a treating clinician in view of a patient’s condition.
  • a therapeutically effective amount is effective for detectable killing, reduction, and/or inhibition of the growth or spread of cancer cells, the size or number of tumors, and/or other measure of the level, stage, progression and/or severity of a cancer.
  • the term also applies to a dose that will induce a particular response in target cells, 40 e.g., a reduction, slowing, or inhibition of cell growth.
  • a therapeutically effective amount can be determined by first administering a low dose, and then incrementally increasing that dose until PAT059646-WO-PCT 5 the desired effect is achieved.
  • a therapeutically effective amount can also vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • the specific amount may vary depending on, for example, the particular pharmaceutical 10 composition, the subject and their age and existing health conditions or risk for health conditions, the dosing regimen to be followed, the severity of the disease, whether it is administered in combination with other agents, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • a therapeutically effective amount of a compound may reduce the number of cancer cells, reduce 15 tumor size, inhibit (e.g., slow or stop) tumor metastasis, inhibit (e.g., slow or stop) tumor growth, and/or relieve one or more symptoms.
  • the term “prophylactically effective amount” or “prophylactically effective dose,” refers to an amount of a compound disclosed herein, e.g., a panRAS inhibitor compound or composition described herein, that is effective, at dosages and for periods of time necessary, 20 to achieve the desired prophylactic result.
  • a prophylactically effective amount will be less than the therapeutically effective amount.
  • a prophylactically effective amount can prevent the onset of disease symptoms, including symptoms associated with a cancer. 25 [120]
  • alkyl refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation.
  • C 1 -C 6 alkyl refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to six carbon atoms, and which is attached to the rest of the molecule by a single bond.
  • Non-30 limiting examples of "C 1 -C 6 alkyl” groups include methyl (a C 1 alkyl), ethyl (a C 2 alkyl), 1- methylethyl (a C 3 alkyl), n-propyl (a C 3 alkyl), isopropyl (a C 3 alkyl), n-butyl (a C 4 alkyl), isobutyl (a C 4 alkyl), sec-butyl (a C 4 alkyl), tert-butyl (a C 4 alkyl), n-pentyl (a C 5 alkyl), isopentyl (a C 5 alkyl), neopentyl (a C5alkyl) and hexyl (a C6alkyl).
  • alkenyl refers to a straight or branched hydrocarbon chain 35 radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond.
  • C 2 -C 6 alkenyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, having from two to six carbon atoms, which is attached to the rest of the molecule by a single bond.
  • C 2 -C 6 alkenyl groups include ethenyl (a C 2 alkenyl), 40 prop-1-enyl (a C 3 alkenyl), but-1-enyl (a C 4 alkenyl), pent-1-enyl (a C 5 alkenyl), pent-4-enyl (a C 5 alkenyl), penta-1,4-dienyl (a C 5 alkenyl), hexa-1-enyl (a C 6 alkenyl), hexa-2-enyl (a C 6 alkenyl), PAT059646-WO-PCT 5 hexa-3-enyl (a C6alkenyl), hexa-1-,4-dienyl (a C6alkenyl), hexa-1-,5-dienyl (a C6alkenyl) and hexa-2-,4-dienyl (a C 6 alkenyl).
  • C 2 -C 3 alkenyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, having from two to three carbon atoms, which is attached to the rest of the molecule by a single bond.
  • Non-limiting examples of "C 2 -C 3 alkenyl” groups 10 include ethenyl (a C 2 alkenyl) and prop-1-enyl (a C 3 alkenyl).
  • alkylene refers to a bivalent straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms and containing no unsaturation.
  • C 1 -C 6 alkylene refers to a bivalent straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no 15 unsaturation, having from one to six carbon atoms.
  • C 1 -C 6 alkylene groups include methylene (a C 1 alkylene), ethylene (a C 2 alkylene), 1-methylethylene (a C 3 alkylene), n-propylene (a C 3 alkylene), isopropylene (a C 3 alkylene), n-butylene (a C 4 alkylene), isobutylene (a C 4 alkylene), sec-butylene (a C 4 alkylene), tert-butylene (a C 4 alkylene), n- pentylene (a C5alkylene), isopentylene (a C5alkylene), neopentylene (a C5alkylene), and 20 hexylene (a C 6 alkylene).
  • alkenylene refers to a bivalent straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms and containing at least one double bond.
  • C 2 -C 6 alkenylene refers to a bivalent straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, 25 containing at least one double bond, and having from two to six carbon atoms.
  • Non-limiting examples of "C 2 -C 6 alkenylene” groups include ethenylene (a C 2 alkenylene), prop-1-enylene (a C 3 alkenylene), but-1-enylene (a C 4 alkenylene), pent-1-enylene (a C 5 alkenylene), pent-4- enylene (a C 5 alkenylene), penta-1,4-dienylene (a C 5 alkenylene), hexa-1-enylene (a C 6 alkenylene), hexa-2-enylene (a C 6 alkenylene), hexa-3-enylene (a C 6 alkenylene), hexa-1-,4- 30 dienylene (a C 6 alkenylene), hexa-1-,5-dienylene (a C 6 alkenylene) and hexa-2-,4-dienylene (a C 6 alkenylene).
  • C 2 -C 6 alkenylene refers to a bivalent straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, and having from two to three carbon atoms.
  • Non-limiting examples of "C 2 -C 3 alkenylene” groups include ethenylene (a C 2 alkenylene) and prop-1-enylene 35 (a C 3 alkenylene).
  • cycloalkyl refers to a non-aromatic, monocyclic, fused bicyclic, fused tricyclic or bridged polycyclic ring system.
  • the cycloalkyl is a mono- or bi-cyclic saturated carbocyclic group containing from 3 to 10 ring members, which may include fused, bridged or spiro ring systems.
  • fused bicyclic or 40 bridged polycyclic ring systems include bicyclo[1.1.1]pentane, bicyclo[2.1.1]hexane, bicyclo[2.2.1]heptane, bicyclo[3.1.1]heptane, bicyclo[3.2.1]octane, bicyclo[2.2.2]octane and 1 PAT059646-WO-PCT 5 adamantanyl.
  • Non-limiting examples monocyclic C3-C8cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl groups.
  • heteroarylene, cycloalkylene, heterocycloalkylene mean a divalent heteroaryl, cycloalkyl and heterocycloalkyl.
  • haloalkyl refers to a linear or branched alkyl chain 10 substituted with one or more halogen groups in place of hydrogens along the hydrocarbon chain.
  • halogen groups suitable for substitution in the haloalkyl group include Fluorine, Bromine, Chlorine, and Iodine.
  • Haloalkyl groups may include substitution with multiple halogen groups in place of hydrogens in an alkyl chain, wherein said halogen groups can be attached to the same carbon or to another carbon in the alkyl chain.
  • the alkyl, alkenyl, alkynyl, alkoxy, amino, aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups may be optionally substituted by 1 to 4 groups selected from optionally substituted linear or branched (C 1 -C 6 )alkyl, optionally substituted linear or branched (C 2 - C 6 )alkenyl group, optionally substituted linear or branched (C 2 -C 6 )alkynyl group, optionally substituted linear or branched (C1-C6)alkoxy, optionally substituted (C1-C6)alkyl-S-, hydroxy, oxo 20 (or N-oxide where appropriate), nitro, cyano, -C(O)-OR 0 ’, -O-C(O)-R 0 ’, -C(O)-NR 0 ’R 0 ’’, -NR 0 ’R 0 0 0 0 0
  • Cis and trans geometric isomers of PAT059646-WO-PCT 5 the compounds of the present disclosure are described and may be isolated as a mixture of isomers or as separated isomeric forms.
  • one or more compounds depicted herein may exist in different tautomeric forms.
  • references to such compounds encompass all such tautomeric forms.
  • tautomeric forms 10 result from the swapping of a single bond with an adjacent double bond and the concomitant migration of a proton.
  • a tautomeric form may be a prototropic tautomer, which is an isomeric protonation states having the same empirical formula and total charge as a reference form.
  • moieties with prototropic tautomeric forms are ketone – enol pairs, amide – imidic acid pairs, lactam – lactim pairs, amide – imidic acid pairs, enamine – imine 15 pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, such as, 1H- and 3H-imidazole, 1H-, 2H- and 4H-1,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole.
  • tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
  • tautomeric forms result from acetal interconversion.
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • Exemplary isotopes that can be incorporated into compounds disclosed herein 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 Cl, 123 I and 125 I.
  • Isotopically- 25 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 18 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 for compounds described herein, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
  • many chemical entities can adopt a variety of different solid forms such as, for example, amorphous forms or crystalline forms (e.g., polymorphs, hydrates, 40 solvate).
  • compounds disclosed herein may be utilized in any such form, PAT059646-WO-PCT 5 including in any solid form.
  • compounds described or depicted herein may be provided or utilized in hydrate or solvate form.
  • substituents of compounds of the present disclosure are disclosed in groups or in ranges. It is specifically intended that the present disclosure includes each and every individual subcombination of the members of such groups 10 and ranges.
  • the term “C 1 -C 6 alkyl” is specifically intended to individually disclose methyl, ethyl, C 3 alkyl, C 4 alkyl, C 5 alkyl, and C 6 alkyl.
  • a compound includes a plurality of positions at which substituents are disclosed in groups or in ranges, unless otherwise indicated, the present disclosure is intended to cover individual compounds and groups of compounds (e.g., genera and subgenera) containing each and every individual 15 subcombination of members at each position.
  • optionally substituted X e.g., “optionally substituted alkyl”
  • alkyl wherein said alkyl is optionally substituted
  • certain compounds of interest may contain one or more “optionally 20 substituted” moieties.
  • substituted whether preceded by the term “optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent, e.g., any of the substituents or groups described herein.
  • an “optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may 25 be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position.
  • the alkyl portion, the heteroaryl portion, or both may be optionally substituted.
  • Combinations of substituents envisioned by the present disclosure are preferably those that result in the formation of stable or chemically feasible compounds.
  • stable refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
  • Suitable divalent substituents that are bound to vicinal substitutable carbons of an “optionally substituted” group include: -O(CR * 2 ) 2-3 O-, wherein each independent occurrence of R * is selected from hydrogen, C 1-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected 30 from nitrogen, oxygen, or sulfur.
  • Suitable substituents on the aliphatic group of R * include halogen, -R • , -(haloR • ), -OH, - OR • , -O(haloR • ), -CN, -C(O)OH, -C(O)OR • , -NH 2 , -NHR • , -NR • 2 , or -NO 2 , wherein each R • is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C 1-4 aliphatic, -CH 2 Ph, -O(CH 2 ) 0-1 Ph, or a 5-6-membered saturated, partially 35 unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on a substitutable nitrogen of an “optionally substituted” group include - R ⁇ , -NR ⁇ 2, -C(O)R ⁇ , -C(O)OR ⁇ , -C(O)C(O)R ⁇ , -C(O)CH2 C(NH)NR ⁇ 2 , or -N(R ⁇ )S(O) 2 R ⁇ ; wherein each R ⁇ is independently hydrogen, C -6 aliphatic which may 40 be substituted as defined below, unsubstituted -OPh, or an unsubstituted 3-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, PAT059646-WO-PCT 5 oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R ⁇ , taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturation
  • Suitable substituents on an aliphatic group of R ⁇ are independently halogen, -R • , -(haloR • ), - 10 OH, -OR • , -O(haloR • ), -CN, -C(O)OH, -C(O)OR • , -NH 2 , -NHR • , -NR • 2 , or -NO 2 , wherein each R • is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C 1-4 aliphatic, -CH 2 Ph, -O(CH 2 )0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • acetyl refers to the group -C(O)CH 3
  • alkoxy refers to a -O-C 1 -C 20 alkyl group, wherein the alkoxy group is attached to the remainder of the compound through an oxygen atom.
  • alkyl refers to a saturated, straight or branched monovalent hydrocarbon group containing from 1 to 20 (e.g., from 1 to 10 or from 1 to 6) carbons.
  • an alkyl group is unbranched (i.e., is linear); in some embodiments, an alkyl group is branched.
  • Alkyl groups are exemplified by, but not limited to, methyl, ethyl, n- and iso-propyl, n-, sec-, iso- and tert-butyl, and neopentyl.
  • heteroalkyl refers to an "alkyl” group, as defined herein, in which at least one carbon atom has been replaced with a heteroatom (e.g., an O, N, or S atom), 25 however the heteroalkyl group must contain at least one carbon atom after all replacements; for example, -NH-CH 2 -CH 3 , -CH 2 -NH-CH 3 , -CH 2 -N-(CH 3 ) 2 , and -CH 2 -CH 2 -NH 2 are heteroalkyls but - NH 2 is not.
  • a heteroatom e.g., an O, N, or S atom
  • an appropriate number of hydrogens are adjusted to fill out valences; for example, -CH 2 - can be replaced by groups such as -O, -S-, and -NH-, while -CH- can be replaced by groups such as -N-.
  • the heteroatom may appear in 30 any part of the radical, for example in the middle or at either end of the radical.
  • the number of carbons listed in the heteroalkyl refers to the number of carbons after all heteroatom replacements.
  • C 1-6 heteroalkyl means that there are between 1 and 6 carbon atoms in the heteroalkyl group, but there are also one or more heteroatoms; for example, -O- CH2-CH3 is a C2 heteroalkyl, not a C3 heteroalkyl.
  • Heteroalkyl is thus inclusive of amines, 35 alkoxys, and thioethers, in addition to other groups.
  • the heteroatoms in a heteroalkyl are selected from the group consisting of N, O and S.
  • alkylene represents a saturated divalent hydrocarbon group derived from a straight or branched chain saturated hydrocarbon by the removal of two hydrogen atoms, and is exemplified by methylene, ethylene, isopropylene, and the like.
  • Cx-Cy 40 alkylene represents alkylene groups having between x and y carbons. Exemplary values for x are 1, 2, 3, 4, 5, and 6, and exemplary values for y are 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or PAT059646-WO-PCT 5 20 (e.g., C1-C6, C1-C10, C2-C20, C2-C6, C2-C10, or C2-C20 alkylene).
  • alkylene can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein.
  • alkenyl represents monovalent straight or branched chain groups of, unless otherwise specified, from 2 to 20 carbons (e.g., from 2 to 6 or from 2 to 10 carbons) containing one or more carbon-carbon double bonds and is exemplified by ethenyl, 1- 10 propenyl, 2-propenyl, 2-methyl-1-propenyl, 1-butenyl, and 2-butenyl.
  • Alkenyls include both cis and trans isomers.
  • alkenylene represents a divalent straight or branched chain groups of, unless otherwise specified, from 2 to 20 carbons (e.g., from 2 to 6 or from 2 to 10 carbons) containing one or more carbon-carbon double bonds.
  • alkynyl represents monovalent straight or branched chain 15 groups from 2 to 20 carbon atoms (e.g., from 2 to 4, from 2 to 6, or from 2 to 10 carbons) containing a carbon-carbon triple bond and is exemplified by ethynyl, and 1-propynyl.
  • amino represents -N(R ⁇ ) 2 , e.g., -NH 2 and -N(CH 3 ) 2 .
  • aminoalkyl represents an alkyl moiety substituted on one or more carbon atoms with one or more amino moieties.
  • aryl represents a monovalent monocyclic, bicyclic, or multicyclic ring system formed by carbon atoms, wherein the ring attached to the pendant group is aromatic. Examples of aryl groups are phenyl, naphthyl, phenanthrenyl, and anthracenyl.
  • aryl ring can be attached to its pendant group at any heteroatom or carbon ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise 25 specified.
  • the aryl refers to a phenyl, nahthyl, biphenyl or indenyl group.
  • C 0 represents a bond.
  • part of the term - N(C(O)-(C 0 -C 5 alkylene-H)- includes -N(C(O)-(C 0 alkylene-H)-, which is also represented by - N(C(O)-H)-.
  • Carbocyclic and “carbocyclyl,” as used herein, refer to a monovalent, 30 optionally substituted C 3 -C 12 monocyclic, bicyclic, or tricyclic ring structure, which may be bridged, fused or spirocyclic, in which all the rings are formed by carbon atoms and at least one ring is non-aromatic.
  • Carbocyclic structures include cycloalkyl, cycloalkenyl, and cycloalkynyl groups.
  • carbocyclyl groups are cyclohexyl, cyclohexenyl, cyclooctynyl, 1,2-dihydronaphthyl, 35 1,2,3,4-tetrahydronaphthyl, fluorenyl, indenyl, indanyl, decalinyl, and the like.
  • a carbocyclic ring can be attached to its pendant group at any ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified.
  • enantiomer means each individual optically active form of a compound disclosed herein, having an optical purity or enantiomeric excess (as determined by 10 methods standard in the art) of at least 80% (i.e., at least 90% of one enantiomer and at most 10% of the other enantiomer), preferably at least 90% and more preferably at least 98%.
  • haloalkyl represents an alkyl moiety substituted on one or more carbon atoms with one or more of the same or different halogen moieties.
  • halogen represents a halogen selected from 15 bromine, chlorine, iodine, and fluorine.
  • heteroaryl represents a monovalent, monocyclic or polycyclic ring structure that contains at least one fully aromatic ring: i.e., they contain 4n+2 pi electrons within the monocyclic or polycyclic ring system and contains at least one ring heteroatom selected from N, O, or S in that aromatic ring.
  • exemplary unsubstituted heteroaryl 20 groups are of 1 to 12 (e.g., 1 to 11, 1 to 10, 1 to 9, 2 to 12, 2 to 11, 2 to 10, or 2 to 9) carbons.
  • heteroaryl incIudes bicyclic, tricyclic, and tetracyclic groups in which any of the above heteroaromatic rings is fused to one or more, aryl or carbocyclic rings, e.g., a phenyl ring, or a cyclohexane ring.
  • heteroaryl groups include, but are not limited to, pyridyl, pyrazolyl, benzooxazolyl, benzoimidazolyl, benzothiazolyl, imidazolyl, thiazolyl, quinolinyl, 25 tetrahydroquinolinyl, and 4-azaindolyl.
  • a heteroaryl ring can be attached to its pendant group at any ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified.
  • the heteroaryl is substituted with 1, 2, 3, or 4 substituents groups.
  • the heteroaryl any mono- or bi-cyclic group composed of from 5 to 10 ring members, having at least one aromatic moiety and 30 containing from 1 to 4 hetero atoms selected from oxygen, sulfur and nitrogen (including quaternary nitrogens).
  • heterocycloalkyl represents a monovalent monocyclic, bicyclic or polycyclic ring system, which may be bridged, fused or spirocyclic, wherein at least one ring is non-aromatic and wherein the non-aromatic ring contains one, two, three, or four 35 heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur.
  • the 5-membered ring has zero to two double bonds, and the 6- and 7-membered rings have zero to three double bonds.
  • Exemplary unsubstituted heterocycloalkyl groups are of 1 to 12 (e.g., 1 to 11, 1 to 10, 1 to 9, 2 to 12, 2 to 11, 2 to 10, or 2 to 9) carbons.
  • heterocycloalkyl also represents a heterocyclic compound having a bridged 40 multicyclic structure in which one or more carbons or heteroatoms bridges two non-adjacent members of a monocyclic ring, e.g., a quinuclidinyl group.
  • heterocycloalkyl includes PAT059646-WO-PCT 5 bicyclic, tricyclic, and tetracyclic groups in which any of the above heterocyclic rings is fused to one or more aromatic, carbocyclic, heteroaromatic, or heterocyclic rings, e.g., an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring, a pyridine ring, or a pyrrolidine ring.
  • heterocycloalkyl groups are pyrrolidinyl, piperidinyl, 1,2,3,4- 10 tetrahydroquinolinyl, decahydroquinolinyl, dihydropyrrolopyridine, and decahydronapthyridinyl.
  • a heterocycloalkyl ring can be attached to its pendant group at any ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified.
  • the term “hydroxy,” as used herein, represents a -OH group. 15
  • the term “hydroxyalkyl,” as used herein, represents an alkyl moiety substituted on one or more carbon atoms with one or more -OH moieties.
  • the term “isomer,” as used herein, means any tautomer, stereoisomer, atropiosmer, enantiomer, or diastereomer of any compound disclosed herein. It is recognized that the compounds disclosed herein can have one or more chiral centers or double bonds and, 20 therefore, exist as stereoisomers, such as double-bond isomers (i.e., geometric E/Z isomers) or diastereomers (e.g., enantiomers (i.e., (+) or (-)) or cis/trans isomers).
  • stereomerically pure form e.g., geometrically pure, enantiomerically pure, or diastereomerically pure
  • enantiomeric and stereoisomeric 25 mixtures e.g., racemates.
  • Enantiomeric and stereoisomeric mixtures of compounds disclosed herein can typically be resolved into their component enantiomers or stereoisomers by well- known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent.
  • Enantiomers and stereoisomers can also be obtained from 30 stereomerically or enantiomerically pure intermediates, reagents, and catalysts by well-known asymmetric synthetic methods.
  • stereoisomer refers to all possible different isomeric as well as conformational forms which a compound may possess (e.g., a compound of any formula described herein), in particular all possible stereochemically and conformationally isomeric 35 forms, all diastereomers, enantiomers or conformers of the basic molecular structure, including atropisomers.
  • Some compounds of the present disclosure may exist in different tautomeric forms, all of the latter being included within the scope of the present invention.
  • compositions described herein e.g., the disclosed panRAS inhibitor compounds and compositions, in treating a subject for a disorder, e.g., a cancer.
  • compositions e.g., panRAS inhibitors
  • Treatment efficacy may be evaluated for toxicity as well as indicators of efficacy and adjusted accordingly.
  • Efficacy measures include, but are not limited to, a cytostatic and/or cytotoxic effect observed in vitro or in vivo, reduced tumor volume, tumor 15 growth inhibition, and/or prolonged survival. [173] Methods of determining whether a panRAS inhibitor exerts a cytostatic and/or cytotoxic effect on a cell are known.
  • the cytotoxic or cytostatic activity of a panRAS inhibitor can be measured by, e.g., exposing mammalian cells to the panRAS inhibitor in a cell culture medium; culturing the cells for a period from about 6 hr to about 6 days; and measuring cell 20 viability (e.g., using a CellTiter-Glo® (CTG) or MTT cell viability assay).
  • CCG CellTiter-Glo®
  • MTT cell viability assay Cell-based in vitro assays may also be used to measure viability (proliferation), cytotoxicity, and induction of apoptosis (caspase activation) of the panRAS inhibitor.
  • Necrosis is typically accompanied by increased permeability of the plasma 25 membrane, swelling of the cell, and rupture of the plasma membrane.
  • Apoptosis can be quantitated, for example, by measuring DNA fragmentation.
  • Commercial photometric methods for the quantitative in vitro determination of DNA fragmentation are available. Examples of such assays, including TUNEL (which detects incorporation of labeled nucleotides in fragmented DNA) and ELISA-based assays, are described in Biochemica (1999) 2:34-7 (Roche Molecular 30 Biochemicals).
  • Apoptosis may also be determined by measuring morphological changes in a cell.
  • loss of plasma membrane integrity can be determined by measuring uptake of certain dyes (e.g., a fluorescent dye such as, for example, acridine orange or ethidium bromide).
  • a fluorescent dye such as, for example, acridine orange or ethidium bromide.
  • a method for measuring apoptotic cell number has been described by Duke and 35 Cohen, Current Protocols in Immunology (Coligan et al., eds. (1992) pp.3.17.1-3.17.16).
  • Cells also can be labeled with a DNA dye (e.g., acridine orange, ethidium bromide, or propidium iodide) and the cells observed for chromatin condensation and margination along the inner nuclear membrane.
  • a DNA dye e.g., acridine orange, ethidium bromide, or propidium iodide
  • Apoptosis may also be determined, in some embodiments, by screening for caspase activity.
  • a Caspase-Glo® Assay can be used to measure 40 activity of caspase-3 and caspase-7.
  • the assay provides a luminogenic caspase-3/7 substrate in a reagent optimized for caspase activity, luciferase activity, and cell PAT059646-WO-PCT 5 lysis.
  • adding Caspase-Glo® 3/7 Reagent in an “add-mix-measure” format may result in cell lysis, followed by caspase cleavage of the substrate and generation of a “glow-type” luminescent signal, produced by luciferase.
  • luminescence may be proportional to the amount of caspase activity present, and can serve as an indicator of apoptosis.
  • Other morphological changes that can be measured to determine apoptosis include, 10 e.g., cytoplasmic condensation, increased membrane blebbing, and cellular shrinkage. Determination of any of these effects on cancer cells indicates that a panRAS inhibitor is useful in the treatment of cancers.
  • Cell viability may be measured, e.g., by determining in a cell the uptake of a dye such as neutral red, trypan blue, Crystal Violet, or ALAMARTM blue (see, e.g., Page et al. (1993) Intl J 15 Oncology 3:473-6).
  • Cell viability may also be measured, e.g., by quantifying ATP, an indicator of metabolically active cells.
  • in vitro potency and/or cell viability of 20 prepared panRAS inhibitor compounds may be assessed using a CellTiter-Glo® (CTG) cell viability assay, as described in the examples provided herein.
  • CCG CellTiter-Glo®
  • the single reagent (CellTiter-Glo® Reagent) is added directly to cells cultured in serum-supplemented medium.
  • reagent results in cell lysis and generation of a luminescent signal proportional to the amount of ATP present.
  • the amount of ATP is directly 25 proportional to the number of cells present in culture.
  • Cell viability may also be measured, e.g., by measuring the reduction of tetrazolium salts.
  • in vitro potency and/or cell viability of prepared panRAS inhibitor compounds may be assessed using an MTT cell viability assay, as described in the examples provided herein.
  • the yellow tetrazolium MTT (3-(4, 5- 30 dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide) is reduced by metabolically active cells, in part by the action of dehydrogenase enzymes, to generate reducing equivalents such as NADH and NADPH.
  • the resulting intracellular purple formazan can then be solubilized and quantified by spectrophotometric means.
  • the present disclosure features a method of killing, inhibiting or 35 modulating the growth of a cancer cell or tissue by disrupting the expression and/or activity of panRAS (e.g., K-Ras (including splice variants KRAS4A and KRAS4B), H-Ras and N-Ras) and/or one or more upstream modulators or downstream targets thereof.
  • panRAS e.g., K-Ras (including splice variants KRAS4A and KRAS4B), H-Ras and N-Ras) expression and/or activity provides a therapeutic 40 benefit.
  • Subjects that may benefit from disrupting panRAS include, but are 1 PAT059646-WO-PCT 5 not limited to, those having or at risk of having a cancer such as a tumor or a hematological cancer.
  • the cancer is a breast cancer including ER positive breast cancer, multiple myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, bladder cancer, brain cancer, bone marrow cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal 10 cancer, hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant prostate cancer, bladder urothelial carcinoma, melanoma, myelogenous leukemia, myeloma, oral cancer, ovarian cancer, non-small cell lung cancer, prostate cancer, small cell lung cancer, spleen cancer, or head and neck cancer.
  • ER positive breast cancer multiple myeloma, plasma cell myeloma, leuk
  • the disclosed panRAS inhibitors may be administered in any cell or tissue that expresses EphA2, such as a EphA2-expressing cancer cell or tissue.
  • An exemplary embodiment includes a method of killing a EphA2-expressing cancer cell or tissue. The method may be used with any cell or tissue that expresses EphA2, such as a cancerous cell or a metastatic lesion.
  • EphA2-expressing cancers include breast 20 cancer, non-small cell lung cancer, pancreatic cancer, esophageal cancer, head and neck cancer, gastric or stomach cancer, bladder cancer, and colorectal cancer.
  • the disclosed panRAS inhibitors may be administered in any cell or tissue that expresses B7-H3 (CD276), such as a B7-H3 (CD276)-expressing cancer cell or tissue.
  • An exemplary embodiment includes a method of killing a B7-H3 (CD276)-expressing 25 cancer cell or tissue. The method may be used with any cell or tissue that expresses B7-H3 (CD276), such as a cancerous cell or a metastatic lesion.
  • Non-limiting examples of B7-H3 (CD276)-expressing cancers include colorectal cancer, pancreatic cancer, lymphoma, non-small cell lung cancer, small cell lung cancer, breast cancer including ER positive breast cancer, metastatic castration resistant prostate cancer, melanoma, bladder urothelial carcinoma, head 30 and neck cancer, and leukemia (e.g., acute myeloid leukemia).
  • Exemplary methods include the steps of contacting a cell with a panRAS inhibitor, as described herein, in an effective amount, i.e., an amount sufficient to kill the cell. The method can be used on cells in culture, e.g., in vitro, in vivo, ex vivo, or in situ.
  • cells that express EphA2 can be cultured in vitro in culture medium and the contacting step can be affected by adding the panRAS inhibitor to the culture medium.
  • the method will result in killing of cells expressing EphA2, including in particular cancer cells expressing EphA2.
  • the panRAS inhibitor can be administered to a subject by any suitable administration route (e.g., intravenous, subcutaneous, or direct contact with a tumor 40 tissue) to have an effect in vivo.
  • panRAS inhibitor therapeutic composition can be evaluated in a suitable animal model.
  • xenogeneic cancer models can be used, wherein cancer explants or passaged xenograft tissues are introduced into immune compromised animals, such as nude or SCID mice (Klein et al. (1997) Nature Med.3:402-8). Efficacy may be predicted using assays that measure inhibition of tumor formation, tumor 10 regression or metastasis, and the like.
  • In vivo assays that evaluate the promotion of tumor death by mechanisms such as apoptosis may also be used.
  • xenografts from tumor bearing mice treated with the therapeutic composition can be examined for the presence of apoptotic foci and compared to untreated control xenograft-bearing mice. The extent to which apoptotic foci are 15 found in the tumors of the treated mice provides an indication of the therapeutic efficacy of the composition.
  • a disorder e.g., a cancer.
  • the compositions described herein, e.g., the panRAS inhibitors disclosed herein can be administered to a non-human mammal or human subject for therapeutic purposes.
  • the 20 therapeutic methods include administering to a subject having or suspected of having a cancer a therapeutically effective amount of a composition comprising a panRAS inhibitor.
  • An exemplary embodiment is a method of treating a subject having or suspected of having a cancer, comprising administering to the subject a therapeutically effective amount of a composition disclosed herein, e.g., a panRAS inhibitor, composition, or pharmaceutical 25 composition (e.g., any of the exemplary panRAS inhibitors, compositions, or pharmaceutical compositions disclosed herein).
  • the cancer is a tumor or a hematological cancer.
  • the cancer is a breast cancer including ER positive breast cancer, multiple myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, bladder cancer, brain cancer, bone marrow 30 cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal cancer, hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant prostate cancer, bladder urothelial carcinoma, melanoma, myelogenous leukemia, myeloma, oral cancer, ovarian cancer, non-small cell lung cancer, 35 prostate cancer, small cell lung cancer, spleen cancer, or head and neck cancer.
  • ER positive breast cancer multiple myeloma, plasma cell myeloma, le
  • the present disclosure further provides methods of reducing or inhibiting growth of a tumor, comprising administering a therapeutically effective amount of a panRAS inhibitor or composition comprising a panRAS inhibitor.
  • the treatment is sufficient to reduce or inhibit the growth of the patient's tumor, reduce the number 40 or size of metastatic lesions, reduce tumor load, reduce primary tumor load, reduce invasiveness, prolong survival time, and/or maintain or improve the quality of life.
  • the tumor is resistant or refractory to treatment with the panRAS inhibitor when administered alone.
  • An exemplary embodiment is a method of reducing or inhibiting the growth of a tumor in a subject, comprising administering to the subject a therapeutically effective amount of an panRAS inhibitor, composition, or pharmaceutical composition (e.g., any of the exemplary 10 panRAS inhibitors, compositions, or pharmaceutical compositions disclosed herein).
  • an panRAS inhibitor, composition, or pharmaceutical composition e.g., any of the exemplary 10 panRAS inhibitors, compositions, or pharmaceutical compositions disclosed herein.
  • the tumor is a breast cancer including ER positive breast cancer, multiple myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, bladder cancer, brain cancer, bone marrow cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal cancer, 15 hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant prostate cancer, bladder urothelial carcinoma, melanoma, myelogenous leukemia, myeloma, oral cancer, ovarian cancer, non-small cell lung cancer, prostate cancer, small cell lung cancer, spleen cancer, or head and neck cancer.
  • breast cancer including ER positive breast cancer, multiple myeloma, plasma cell myelom
  • the tumor is a gastric cancer. 20
  • administration of the panRAS inhibitor, composition, or pharmaceutical composition reduces or inhibits the growth of the tumor by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, or at least about 99%, as compared to growth in the absence of treatment.
  • Another exemplary embodiment is a method of delaying or slowing the growth of a tumor in a subject, comprising administering to the subject a therapeutically effective amount of a panRAS inhibitor, composition, or pharmaceutical composition (e.g., any of the exemplary panRAS inhibitors, compositions, or pharmaceutical compositions disclosed herein).
  • a panRAS inhibitor, composition, or pharmaceutical composition e.g., any of the exemplary panRAS inhibitors, compositions, or pharmaceutical compositions disclosed herein.
  • the tumor is a breast cancer including ER positive breast cancer, multiple 30 myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, bladder cancer, brain cancer, bone marrow cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal cancer, hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant 35 prostate cancer, bladder urothelial carcinoma, melanoma, myelogenous leukemia, myeloma, oral cancer, ovarian cancer, non-small cell lung cancer, prostate cancer, small cell lung cancer, spleen cancer, or head and neck cancer.
  • ER positive breast cancer multiple 30 myeloma, plasma cell myeloma,
  • the tumor is a gastric cancer.
  • administration of the panRAS inhibitor, composition, or pharmaceutical composition delays or slows the growth of the tumor by at least about 10%, at least about 20%, 40 at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about PAT059646-WO-PCT 5 70%, at least about 80%, at least about 90%, at least about 95%, or at least about 99%, as compared to growth in the absence of treatment.
  • the present disclosure further provides methods of reducing or slowing the expansion of a cancer cell population, comprising administering a therapeutically effective amount of a panRAS inhibitor or composition comprising a panRAS inhibitor.
  • An exemplary embodiment is a method of reducing or slowing the expansion of a cancer cell population in a subject, comprising administering to the subject a therapeutically effective amount of a panRAS inhibitor, composition, or pharmaceutical composition (e.g., any of the exemplary panRAS inhibitors, compositions, or pharmaceutical compositions disclosed herein).
  • the cancer cell population is from a tumor or a hematological cancer.
  • the cancer cell population is a breast cancer including ER positive breast cancer, multiple myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, bladder cancer, brain cancer, bone marrow cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal cancer, hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, 20 follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant prostate cancer, bladder urothelial carcinoma, melanoma, myelogenous leukemia, myeloma, oral cancer, ovarian cancer, non-small cell lung cancer, prostate cancer, small cell lung cancer, spleen cancer, or head and neck cancer.
  • ER positive breast cancer multiple myeloma, plasma cell myeloma,
  • administration of the panRAS inhibitor, composition, or pharmaceutical composition reduces the cancer cell 25 population by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, or at least about 99%, as compared to the population in the absence of treatment.
  • administration of the panRAS inhibitor, composition, or pharmaceutical composition slows the expansion of the cancer cell population by at least about 30 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, or at least about 99%, as compared to expansion in the absence of treatment.
  • Also provided herein are methods of determining whether a subject having or suspected of having a cancer will be responsive to treatment with the disclosed panRAS inhibitors and 35 compositions.
  • An exemplary embodiment is a method of determining whether a subject having or suspected of having a cancer will be responsive to treatment with a panRAS inhibitor, composition, or pharmaceutical composition (e.g., any of the exemplary panRAS inhibitors, compositions, or pharmaceutical compositions disclosed herein) by providing a biological sample from the subject; contacting the sample with the panRAS inhibitor; and detecting binding 40 of the panRAS inhibitor to cancer cells in the sample.
  • the sample is a tissue biopsy sample, a blood sample, or a bone marrow sample.
  • the PAT059646-WO-PCT 5 method comprises providing a biological sample from the subject; contacting the sample with the panRAS inhibitor; and detecting one or more markers of cancer cell death in the sample (e.g., increased expression of one or more apoptotic markers, reduced expansion of a cancer cell population in culture, etc.).
  • one or more markers of cancer cell death in the sample e.g., increased expression of one or more apoptotic markers, reduced expansion of a cancer cell population in culture, etc.
  • An exemplary embodiment is a panRAS inhibitor, composition, or pharmaceutical composition (e.g., any of the exemplary panRAS inhibitors, compositions, or pharmaceutical compositions disclosed herein) for use in treating a subject having or suspected of having a cancer.
  • panRAS inhibitors of the present disclosure may be administered to a non- human mammal for veterinary purposes or as an animal model of human disease.
  • the therapeutic compositions used in the practice of the foregoing methods may be 25 formulated into pharmaceutical compositions comprising a pharmaceutically acceptable carrier suitable for the desired delivery method.
  • An exemplary embodiment is a pharmaceutical composition comprising an panRAS inhibitor of the present disclosure and a pharmaceutically acceptable carrier, e.g., one suitable for a chosen means of administration, e.g., intravenous administration.
  • the pharmaceutical composition may also comprise one or more additional 30 inactive and/or therapeutic agents that are suitable for treating or preventing, for example, a cancer (e.g., a standard-of-care agent, etc.).
  • the pharmaceutical composition may also comprise one or more carrier, excipient, and/or stabilizer components, and the like.
  • Methods of formulating such pharmaceutical compositions and suitable formulations are known in the art (see, e.g., “Remington’s Pharmaceutical Sciences,” Mack Publishing Co., Easton, PA).
  • Suitable carriers include any material that, when combined with the therapeutic composition, retains the anti-tumor function of the therapeutic composition and is generally non- reactive with the patient’s immune system.
  • Pharmaceutically acceptable carriers include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • Examples of 40 pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol, mesylate salt, and the like, as well as combinations thereof.
  • PAT059646-WO-PCT 5 isotonic agents are included, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition.
  • Pharmaceutically acceptable carriers may further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the panRAS inhibitor. 10 [197]
  • a pharmaceutical composition of the present disclosure can be administered by a variety of methods known in the art.
  • the route and/or mode of administration may vary depending upon the desired results.
  • the therapeutic formulation is solubilized and administered via any route capable of delivering the therapeutic composition to the cancer site.
  • Potentially effective routes of administration include, but are not limited to, parenteral (e.g., 15 intravenous, subcutaneous), intraperitoneal, intramuscular, intratumor, intradermal, intraorgan, orthotopic, and the like.
  • the administration is intravenous, subcutaneous, intraperitoneal, or intramuscular.
  • the pharmaceutically acceptable carrier should be suitable for the route of administration, e.g., intravenous or subcutaneous administration (e.g., by injection or infusion).
  • the 20 active compound(s), i.e., the panRAS inhibitor and/or any additional therapeutic agent, may be coated in a material to protect the compound(s) from the action of acids and other natural conditions that may inactivate the compound(s).
  • Administration can be either systemic or local.
  • the therapeutic compositions disclosed herein may be sterile and stable under the conditions of manufacture and storage, and may be in a variety of forms. These include, for 25 example, liquid, semi-solid, and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes, and suppositories. The form depends on the intended mode of administration and therapeutic application.
  • the disclosed panRAS inhibitors can be incorporated into a pharmaceutical composition suitable for parenteral administration.
  • the injectable solution may30 be composed of either a liquid or lyophilized dosage form in a flint or amber vial, ampule, or pre- filled syringe, or other known delivery or storage device.
  • one or more of the panRAS inhibitors or pharmaceutical compositions is supplied as a dry sterilized lyophilized powder or water free concentrate in a hermetically sealed container and can be reconstituted (e.g., with water or saline) to the appropriate concentration for administration to a subject.
  • a therapeutically effective amount or efficacious amount of a disclosed composition is employed in the pharmaceutical compositions of the present disclosure.
  • the composition e.g., one comprising an panRAS inhibitor, may be formulated into a pharmaceutically acceptable dosage form by conventional methods known in the art. Dosages and administration protocols for the treatment of cancers 40 using the foregoing methods will vary with the method and the target cancer, and will generally depend on a number of other factors appreciated in the art.
  • compositions disclosed herein may be adjusted to provide the optimum desired response (e.g., a therapeutic response).
  • a single bolus of one or both agents may be administered at one time, several divided doses may be administered over a predetermined period of time, or the dose of one or 10 both agents may be proportionally increased or decreased as indicated by the exigencies of the therapeutic situation.
  • treatment involves single bolus or repeated administration of the panRAS inhibitor preparation via an acceptable route of administration.
  • the panRAS inhibitor is administered to the patient daily, weekly, monthly, or any time period in between.
  • specific dosage regimens may be 15 adjusted over time according to the individual’s need, and the professional judgment of the treating clinician.
  • Parenteral compositions may be formulated in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect 20 in association with the required pharmaceutical carrier.
  • compositions comprising a panRAS inhibitor and/or any additional therapeutic agent(s) may be selected based on the unique characteristics of the active compound(s), and the particular therapeutic effect to be achieved.
  • a physician or veterinarian can start doses of the panRAS inhibitor employed in the pharmaceutical composition at levels 25 lower than that required to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • effective doses of the compositions of the present disclosure, for the treatment of a cancer may vary depending upon many different factors, including means of administration, target site, physiological state of the patient, whether the patient is human or an animal, other medications administered, and whether treatment is 30 prophylactic or therapeutic.
  • the selected dosage level may also depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present disclosure employed, or the ester, salt, or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular 35 compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors. Treatment dosages may be titrated to optimize safety and efficacy. [202] Toxicity and therapeutic efficacy of compounds provided herein can be determined by standard pharmaceutical procedures in cell culture or in animal models.
  • LD50, 40 ED50, EC50, and IC50 may be determined, and the dose ratio between toxic and therapeutic effects (LD50/ED50) may be calculated as the therapeutic index.
  • the data obtained from in PAT059646-WO-PCT 5 vitro and in vivo assays can be used in estimating or formulating a range of dosage for use in humans.
  • the compositions and methods disclosed herein may initially be evaluated in xenogeneic cancer models (e.g., an NCI-H929 multiple myeloma mouse model).
  • a panRAS inhibitor or composition comprising a panRAS inhibitor is administered on a single occasion.
  • a panRAS inhibitor or composition 10 comprising a panRAS inhibitor is administered on multiple occasions. Intervals between single dosages can be, e.g., daily, weekly, monthly, or yearly. Intervals can also be irregular, based on measuring blood levels of the administered agent (e.g., the panRAS inhibitor) in the patient in order to maintain a relatively consistent plasma concentration of the agent.
  • the dosage and frequency of administration of a panRAS inhibitor or composition comprising a panRAS inhibitor 15 may also vary depending on whether the treatment is prophylactic or therapeutic. In prophylactic applications, a relatively low dosage may be administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives.
  • panRAS inhibitors or compositions disclosed herein are co-formulated and/or co-administered 25 with one or more additional therapeutic agents, e.g., one or more chemotherapeutic agents, one or more standard-of-care agents for the particular condition being treated.
  • Kits for use in the therapeutic and/or diagnostic applications described herein are also provided.
  • Such kits may comprise a carrier, package, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) 30 comprising one of the separate elements to be used in a method disclosed herein.
  • a label may be present on or with the container(s) to indicate that a panRAS inhibitor or composition within the kit is used for a specific therapy or non-therapeutic application, such as a prognostic, prophylactic, diagnostic, or laboratory application.
  • a label may also indicate directions for either in vivo or in vitro use, such as those described herein.
  • Directions and or other information may 35 also be included on an insert(s) or label(s), which is included with or on the kit.
  • the label may be on or associated with the container.
  • a label may be on a container when letters, numbers, or other characters forming the label are molded or etched into the container itself.
  • a label may be associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert.
  • the label may indicate that a panRAS inhibitor or 40 composition within the kit is used for diagnosing or treating a condition, such as a cancer a described herein.
  • a kit comprises a panRAS inhibitor or composition comprising a panRAS inhibitor.
  • the kit further comprises one or more additional components, including but not limited to: instructions for use; other reagents, e.g., a therapeutic agent (e.g., a standard-of-care agent); devices, containers, or other materials for preparing the panRAS inhibitor for administration; pharmaceutically acceptable carriers; and devices, 10 containers, or other materials for administering the panRAS inhibitor to a subject.
  • Instructions for use can include guidance for therapeutic applications including suggested dosages and/or modes of administration, e.g., in a patient having or suspected of having a cancer.
  • the kit comprises a panRAS inhibitor and instructions for use of the panRAS inhibitor in treating, preventing, and/or diagnosing a cancer.
  • elevated panRAS e.g., K-Ras (including splice variants KRAS4A and KRAS4B), H-Ras and N-Ras
  • PanRAS inhibitors that may not be sufficiently effective as monotherapy to treat cancer can be administered in combination with other therapeutic agents (including non- targeted and targeted therapeutic agents) or radiation therapy (including radioligand therapy) to 20 provide therapeutic benefit.
  • panRAS inhibitors described herein sensitize tumor cells to the treatment with other therapeutic agents (including standard of care chemotherapeutic agents to which the tumor cells may have developed resistance) and/or radiation therapy.
  • panRAS inhibitors described herein are administered to a subject having cancer in an amount effective to 25 sensitize the tumor cells.
  • the term “sensitize” means that the treatment with panRAS inhibitor increases the potency or efficacy of the treatment with other therapeutic agents and/or radiation therapy against tumor cells.
  • the present disclosure provides methods of treatment wherein the panRAS inhibitors disclosed herein are administered in combination with one or more (e.g., 1 or 2) additional therapeutic agents.
  • additional therapeutic agents e.g. 1 or 2
  • Exemplary combination partners are disclosed herein.
  • a combination described herein comprises a PD-1 inhibitor.
  • the PD-1 inhibitor is chosen from PDR001 (Novartis), Nivolumab (Bristol- 35 Myers Squibb), Pembrolizumab (Merck & Co), Pidilizumab (CureTech), MEDI0680 (Medimmune), REGN2810 (Regeneron), TSR-042 (Tesaro), PF-06801591 (Pfizer), BGB-A317 (Beigene), BGB-108 (Beigene), INCSHR1210 (Incyte), or AMP-224 (Amplimmune).
  • the PD-1 inhibitor is PDR001.
  • PDR001 is also known as Spartalizumab.
  • a combination described herein comprises a LAG-3 inhibitor.
  • the LAG-3 inhibitor is chosen from LAG525 (Novartis), BMS-986016 (Bristol-Myers Squibb), or TSR-033 (Tesaro).
  • PAT059646-WO-PCT 5 [211] In certain embodiments, a combination described herein comprises a TIM-3 inhibitor.
  • the TIM-3 inhibitor is MBG453 (Novartis), TSR-022 (Tesaro), LY-3321367 (Eli Lily), Sym23 (Symphogen), BGB-A425 (Beigene), INCAGN-2390 (Agenus), BMS-986258 (BMS), RO-7121661 (Roche), or LY-3415244 (Eli Lilly).
  • a combination described herein comprises a PDL1 inhibitor.
  • the PDL1 inhibitor is chosen from FAZ053 (Novartis), atezolizumab (Genentech), durvalumab (Astra Zeneca), or avelumab (Pfizer).
  • a combination described herein comprises a GITR agonist.
  • the GITR agonist is chosen from GWN323 (NVS), BMS-986156, MK-4166 or MK-1248 (Merck), TRX518 (Leap Therapeutics), INCAGN1876 (Incyte/Agenus), AMG 228 15 (Amgen) or INBRX-110 (Inhibrx).
  • a combination described herein comprises an IAP inhibitor.
  • the IAP inhibitor comprises LCL161 or a compound disclosed in International Application Publication No. WO 2008/016893.
  • the combination comprises an mTOR inhibitor, e.g., RAD001 (also 20 known as everolimus).
  • the combination comprises a HDAC inhibitor, e.g., LBH589. LBH589 is also known as panobinostat.
  • the combination comprises an IL-17 inhibitor, e.g., CJM112.
  • a combination described herein comprises an estrogen receptor 25 (ER) antagonist.
  • the estrogen receptor antagonist is used in combination with a PD-1 inhibitor, a CDK4/6 inhibitor, or both.
  • the combination is used to treat an ER positive (ER+) cancer or a breast cancer (e.g., an ER+ breast cancer).
  • the estrogen receptor antagonist is a selective estrogen receptor 30 degrader (SERD).
  • SESDs are estrogen receptor antagonists which bind to the receptor and result in e.g., degradation or down-regulation of the receptor (Boer K. et al., (2017) Therapeutic Advances in Medical Oncology 9(7): 465-479).
  • ER is a hormone-activated transcription factor important for e.g., the growth, development and physiology of the human reproductive system. ER is activated by, e.g., the hormone estrogen (17beta estradiol).
  • ER expression and signaling 35 is implicated in cancers (e.g., breast cancer), e.g., ER positive (ER+) breast cancer.
  • the SERD is chosen from LSZ102, fulvestrant, brilanestrant, or elacestrant.
  • the SERD comprises a compound disclosed in International Application Publication No. WO 2014/130310, which is hereby incorporated by reference in its entirety. 40 [221] In some embodiments, the SERD comprises LSZ102.
  • LSZ102 has the chemical name: (E)-3-(4-((2-(2-(1,1-difluoroethyl)-4-fluorophenyl)-6-hydroxybenzo[b]thiophen-3- PAT059646-WO-PCT 5 yl)oxy)phenyl)acrylic acid.
  • the SERD comprises fulvestrant (CAS Registry Number: 129453-61-8), or a compound disclosed in International Application Publication No. WO 2001/051056, which is hereby incorporated by reference in its entirety.
  • the SERD comprises elacestrant (CAS Registry Number: 722533-56-4), or a compound disclosed in U.S.
  • Elacestrant is also known as RAD1901, ER-306323 or (6R)-6- ⁇ 2-[Ethyl( ⁇ 4-[2- (ethylamino)ethyl]phenyl ⁇ methyl)amino]-4-methoxyphenyl ⁇ -5,6,7,8-tetrahydronaphthalen-2-ol.
  • Elacestrant is an orally bioavailable, non-steroidal combined selective estrogens receptor modulator (SERM) and a SERD.
  • SERM selective estrogens receptor modulator
  • SERD selective estrogens receptor modulator
  • the SERD is brilanestrant (CAS Registry 15 Number: 1365888-06-7), or a compound disclosed in International Application Publication No. WO 2015/136017, which is incorporated by reference in its entirety.
  • the SERD is chosen from RU 58668, GW7604, AZD9496, apeledoxifene, pipendoxifene, arzoxifene, OP-1074, or acolbifene, e.g., as disclosed in McDonell et al. (2015) Journal of Medicinal Chemistry 58(12) 4883-4887.
  • a combination described herein comprises an inhibitor of Cyclin-Dependent Kinases 4 or 6 (CDK4/6).
  • CDK4/6 Cyclin-Dependent Kinases 4 or 6
  • the CDK4/6 inhibitor is 25 used in combination with a PD-1 inhibitor, an estrogen receptor (ER) antagonist, or both.
  • the combination is used to treat an ER positive (ER+) cancer or a breast cancer (e.g., an ER+ breast cancer).
  • the CDK4/6 inhibitor is chosen from ribociclib, abemaciclib (Eli Lilly), or palbociclib.
  • the CDK4/6 inhibitor comprises ribociclib (CAS Registry Number: 30 1211441-98-3), or a compound disclosed in U.S. Patent Nos.8,415,355 and 8,685,980, which are incorporated by reference in their entirety.
  • the CDK4/6 inhibitor comprises a compound disclosed in International Application Publication No. WO 2010/020675 and U.S. Patent Nos.8,415,355 and 8,685,980, which are incorporated by reference in their entirety.
  • the CDK4/6 inhibitor comprises ribociclib (CAS Registry Number: 1211441-98-3). Ribociclib is also known as LEE011, KISQALI®, or 7-cyclopentyl-N,N- dimethyl-2-((5-(piperazin-1-yl)pyridin-2-yl)amino)-7H-pyrrolo[2,3-d]pyrimidine-6-carboxamide. [228] In some embodiments, the CDK4/6 inhibitor comprises abemaciclib (CAS Registry Number: 1231929-97-7).
  • Abemaciclib is also known as LY835219 or N-[5-[(4-Ethyl-1-40 piperazinyl)methyl]-2-pyridinyl]-5-fluoro-4-[4-fluoro-2-methyl-1-(1-methylethyl)-1H-benzimidazol- PAT059646-WO-PCT 5 6-yl]-2-pyrimidinamine.
  • Abemaciclib is a CDK inhibitor selective for CDK4 and CDK6 and is disclosed, e.g., in Torres-Guzman R et al. (2017) Oncotarget 10.18632/oncotarget.17778.
  • the CDK4/6 inhibitor comprises palbociclib (CAS Registry Number: 571190-30-2).
  • Palbociclib is also known as PD-0332991, IBRANCE® or 6-Acetyl-8- cyclopentyl-5-methyl-2- ⁇ [5-(1-piperazinyl)-2-pyridinyl]amino ⁇ pyrido[2,3-d]pyrimidin-7(8H)-one.
  • 10 Palbociclib inhibits CDK4 with an IC50 of 11nM, and inhibits CDK6 with an IC50 of 16nM, and is disclosed, e.g., in Finn et al. (2009) Breast Cancer Research 11(5):R77.
  • a combination described herein comprises an inhibitor of chemokine (C-X-C motif) receptor 2 (CXCR2).
  • CXCR2 inhibitor is chosen from 6-chloro-3-((3,4-dioxo-2-(pentan-3-ylamino)cyclobut-1-en-1-yl)amino)-2-hydroxy-N- 15 methoxy-N-methylbenzenesulfonamide, danirixin, reparixin, or navarixin.
  • the CSF-1/1R binding agent is chosen from an inhibitor of macrophage colony-stimulating factor (M-CSF), e.g., a monoclonal antibody or Fab to M-CSF (e.g., MCS110), a CSF-1R tyrosine kinase inhibitor (e.g., 4-((2-(((1R,2R)-2- hydroxycyclohexyl)amino)benzo[d]thiazol-6-yl)oxy)-N-methylpicolinamide or BLZ945), a 20 receptor tyrosine kinase inhibitor (RTK) (e.g., pexidartinib), or an antibody targeting CSF-1R (e.g., emactuzumab or FPA008).
  • M-CSF macrophage colony-stimulating factor
  • MCS110 monoclonal antibody or Fab to M-CSF
  • CSF-1R tyrosine kinase inhibitor
  • the CSF-1/1R inhibitor is BLZ945.
  • the CSF-1/1R binding agent is MCS110.
  • the CSF- 1/1R binding agent is pexidartinib.
  • a combination described herein comprises a c-MET 25 inhibitor.
  • c-MET a receptor tyrosine kinase overexpressed or mutated in many tumor cell types, plays key roles in tumor cell proliferation, survival, invasion, metastasis, and tumor angiogenesis. Inhibition of c-MET may induce cell death in tumor cells overexpressing c-MET protein or expressing constitutively activated c-MET protein.
  • the c-MET inhibitor is chosen from capmatinib (INC280), JNJ-3887605, AMG 337, LY2801653, 30 MSC2156119J, crizotinib, tivantinib, or golvatinib.
  • a combination described herein comprises a transforming g rowth factor beta (also known as TGF- TGF , TGFb, or TGF-beta, used interchangeably herein) inhibitor.
  • the TGF- inhibitor is chosen from fresolimumab or XOMA 089.
  • a combination described herein comprises an adenosine A2a receptor (A2aR) antagonist (e.g., an inhibitor of A2aR pathway, e.g., an adenosine inhibitor, e.g., an inhibitor of A2aR or CD-73).
  • A2aR antagonist is used in combination with a PD-1 inhibitor, and one or more (e.g., two, three, four, five, or all) of a CXCR2 inhibitor, a CSF-1/1R binding agent, LAG-3 inhibitor, a GITR agonist, a c-MET inhibitor, 40 or an IDO inhibitor.
  • the combination is used to treat a pancreatic cancer, a colorectal cancer, a gastric cancer, or a melanoma (e.g., a refractory melanoma).
  • the A2aR antagonist is chosen from PBF509 (NIR178) (Palobiofarma/Novartis), CPI444/V81444 (Corvus/Genentech), AZD4635/HTL-1071 (AstraZeneca/Heptares), Vipadenant (Redox/Juno), GBV-2034 (Globavir), AB928 (Arcus Biosciences), Theophylline, Istradefylline (Kyowa Hakko Kogyo), Tozadenant/SYN-115 (Acorda), KW-6356 (Kyowa Hakko Kogyo), ST- 4206 (Leadiant Biosciences), or Preladen
  • a combination described herein comprises an inhibitor of indoleamine 2,3-dioxygenase (IDO) and/or tryptophan 2,3-dioxygenase (TDO).
  • IDO indoleamine 2,3-dioxygenase
  • TDO tryptophan 2,3-dioxygenase
  • the IDO inhibitor is used in combination with a PD-1 inhibitor, and one or more 15 (e.g., two, three, four, or all) of a TGF- inhibitor, an A2aR antagonist, a CSF-1/1R binding agent, a c-MET inhibitor, or a GITR agonist.
  • the combination is used to treat a pancreatic cancer, a colorectal cancer, a gastric cancer, or a melanoma (e.g., a refractory melanoma).
  • the IDO inhibitor is chosen from (4E)-4-[(3- chloro-4-fluoroanilino)-nitrosomethylidene]-1,2,5-oxadiazol-3-amine (also known as epacadostat20 or INCB24360), indoximod (NLG8189), (1-methyl-D-tryptophan), -cyclohexyl-5H-Imidazo[5,1- a]isoindole-5-ethanol (also known as NLG919), indoximod, BMS-986205 (formerly F001287).
  • a combination described herein comprises a Galectin, e.g., Galectin-1 or Galectin-3, inhibitor.
  • the combination comprises a Galectin-1 inhibitor and a Galectin-3 inhibitor.
  • the combination 25 comprises a bispecific inhibitor (e.g., a bispecific antibody molecule) targeting both Galectin-1 and Galectin-3.
  • the Galectin inhibitor is used in combination with one or more therapeutic agents described herein.
  • the Galectin inhibitor is chosen from an anti-Galectin antibody molecule, GR-MD-02 (Galectin Therapeutics), Galectin- 3C (Mandal Med), Anginex, or OTX-008 (OncoEthix, Merck).
  • a combination described herein comprises an inhibitor of the MAP kinase pathway including ERK inhibitors, MEK inhibitors and RAF inhibitors. [237] In some embodiments, a combination described herein comprises a MEK inhibitor.
  • the MEK inhibitor is chosen from Trametinib, selumetinib, AS703026, BIX 02189, BIX 02188, CI-1040, PD0325901, PD98059, U0126, XL-518, G-38963, or 35 G02443714. [238] In some embodiments, the MEK inhibitor is trametinib.
  • Trametinib is also known as JTP-74057, TMT212, N-(3- ⁇ 3-cyclopropyl-5-[(2-fluoro-4-iodophenyl)amino]-6,8-dimethyl-2,4,7- trioxo-3,4,6,7-tetrahydropyrido[4,3-d]pyrimidin-1(2H)-yl ⁇ phenyl)acetamide, or Mekinist (CAS Number 871700-17-3).
  • the MEK inhibitor comprises selumetinib which has the chemical name: (5-[(4-bromo-2-chlorophenyl)amino]-4-fluoro-N-(2-hydroxyethoxy)-1-methyl-1H- PAT059646-WO-PCT 5 benzimidazole-6-carboxamide.
  • Selumetinib is also known as AZD6244 or ARRY 142886, e.g., as described in PCT Publication No. WO2003077914.
  • the MEK inhibitor comprises AS703026, BIX 02189 or BIX 02188.
  • the MEK inhibitor comprises 2-[(2-Chloro-4-iodophenyl)amino]-N- 10 (cyclopropylmethoxy)-3,4-difluoro-benzamide (also known as CI-1040 or PD184352), e.g., as described in PCT Publication No. WO2000035436).
  • the MEK inhibitor comprises N-[(2R)-2,3-Dihydroxypropoxy]-3,4- difluoro-2-[(2-fluoro-4-iodophenyl)amino]- benzamide (also known as PD0325901), e.g., as described in PCT Publication No.
  • the MEK inhibitor comprises 2’-amino-3’-methoxyflavone (also known as PD98059) which is available from Biaffin GmbH & Co., KG, Germany.
  • the MEK inhibitor comprises 2,3-bis[amino[(2- aminophenyl)thio]methylene]-butanedinitrile (also known as U0126), e.g., as described in US Patent No.2,779,780).
  • the MEK inhibitor comprises XL-518 (also known as GDC-0973) which has a CAS No.1029872-29-4 and is available from ACC Corp.
  • the MEK inhibitor comprises G-38963.
  • the MEK inhibitor comprises G02443714 (also known as AS703206) 25 [248] Additional examples of MEK inhibitors are disclosed in WO 2013/019906, WO 03/077914, WO 2005/121142, WO 2007/04415, WO 2008/024725 and WO 2009/085983, the contents of which are incorporated herein by reference.
  • MEK inhibitors include, but are not limited to, 2,3-Bis[amino[(2-aminophenyl)thio]methylene]-butanedinitrile (also known as U0126 and described in US Patent No.2,779,780); (3S,4R,5Z,8S,9S,11E)-14-30 (Ethylamino)-8,9,16-trihydroxy-3,4-dimethyl-3,4,9, 19-tetrahydro-1H-2-benzoxacyclotetradecine- 1,7(8H)-dione] (also known as E6201, described in PCT Publication No.
  • WO2003076424 vemurafenib (PLX-4032, CAS 918504-65-1); (R)-3-(2,3-Dihydroxypropyl)-6-fluoro-5-(2-fluoro-4- iodophenylamino)-8-methylpyrido[2,3-d]pyrimidine-4,7(3H,8H)-dione (TAK-733, CAS 1035555- 63-5); pimasertib (AS-703026, CAS 1204531-26-9); 2-(2-Fluoro-4-iodophenylamino)-N-(2-35 hydroxyethoxy)-1,5-dimethyl-6-oxo-1,6-dihydropyridine-3-carboxamide (AZD 8330); and 3,4- Difluoro-2-[(2-fluoro-4-iodophenyl)amino]-N-(2-hydroxyethoxy)-5-[(3-oxo-[1,
  • a combination described herein comprises a RAF inhibitor.
  • RAF inhibitors include, but are not limited to, Vemurafenib (or Zelboraf®, PLX-4032, 40 CAS 918504-65-1), GDC-0879, PLX-4720 (available from Symansis), Dabrafenib (or PAT059646-WO-PCT 5 GSK2118436), LGX 818, CEP-32496, UI-152, RAF 265, Regorafenib (BAY 73-4506), CCT239065, or Sorafenib (or Sorafenib Tosylate, or Nexavar®).
  • the RAF inhibitor is Dabrafenib.
  • the RAF inhibitor is LXH254.
  • a combination described herein comprises an ERK inhibitor. 10
  • ERK inhibitors include, but are not limited to, LTT462, ulixertinib (BVD-523), LY3214996, GDC-0994, KO-947 and MK-8353. [255] In some embodiments, the ERK inhibitor is LTT462.
  • LTT462 is 4-(3-amino-6- ((1S,3S,4S)-3-fluoro-4-hydroxy ⁇ cyclohexyl)pyrazin-2-yl)-N-((S)-1-(3-bromo-5-fluorophenyl)-2- (methylamino) ⁇ ethyl)-2-fluorobenzamide and is the compound of the following structure: 15 [256]
  • the preparation of LTT462 is described in PCT patent application publication WO2015/066188.
  • LTT462 is an inhibitor of extracellular signal-regulated kinases 1 and 2 (ERK 1/2).
  • a combination described herein comprises a taxane, a vinca 20 alkaloid, a MEK inhibitor, an ERK inhibitor, or a RAF inhibitor.
  • a combination described herein comprises at least two inhibitors selected, independently, from a MEK inhibitor, an ERK inhibitor, and a RAF inhibitor.
  • a combination described herein comprises an anti-mitotic drug.
  • a combination described herein comprises a taxane. 25 [261] Taxanes include, but are not limited to, docetaxel, paclitaxel, or cabazitaxel. In some embodiments, the taxane is docetaxel.
  • a combination described herein comprises a vinca alkaloid.
  • Vinca alkaloids include, but are not limited to, vincristine, vinblastine, and leurosine.
  • a combination described herein comprises a topoisomerase 30 inhibitor.
  • Topoisomerase inhibitors include, but are not limited to, topotecan, irinotecan, camptothecin, diflomotecan, lamellarin D, ellipticines, etoposide (VP-16), teniposide, doxorubicin, daunorubicin, mitoxantrone, amsacrine, aurintricarboxylic acid, and HU-331.
  • PAT059646-WO-PCT 5 [266]
  • a combination described herein includes an interleukin-1 beta (IL- 1 ) inhibitor.
  • the IL-1 inhibitor is chosen from canakinumab, gevokizumab, Anakinra, or Rilonacept.
  • a combination described herein comprises an IL-15/IL-15Ra complex.
  • the IL-15/IL-15Ra complex is chosen from NIZ985 (Novartis), 10 ATL-803 (Altor) or CYP0150 (Cytune).
  • a combination described herein comprises a mouse double minute 2 homolog (MDM2) inhibitor.
  • the human homolog of MDM2 is also known as HDM2.
  • an MDM2 inhibitor described herein is also known as a HDM2 inhibitor.
  • the MDM2 inhibitor is chosen from HDM201 or CGM097. 15 [269] In an embodiment the MDM2 inhibitor comprises (S)-1-(4-chlorophenyl)-7-isopropoxy-6- methoxy-2-(4-(methyl(((1r,4S)-4-(4-methyl-3-oxopiperazin-1- yl)cyclohexyl)methyl)amino)phenyl)-1,2-dihydroisoquinolin-3(4H)-one (also known as CGM097) or a compound disclosed in PCT Publication No. WO 2011/076786 to treat a disorder, e.g., a disorder described herein).
  • a therapeutic agent disclosed herein is used in 20 combination with CGM097.
  • a combination described herein comprises a hypomethylating agent (HMA).
  • HMA hypomethylating agent
  • the HMA is chosen from decitabine or azacitidine.
  • a combination described herein comprises a glucocorticoid.
  • the glucocorticoid is dexamethasone.
  • a combination described herein comprises asparaginase.
  • a combination described herein comprises an inhibitor acting on any pro-survival proteins of the Bcl2 family.
  • a combination described herein comprises a Bcl-2 inhibitor.
  • the Bcl-2 inhibitor is venetoclax (also known 30 [274]
  • the Bcl-2 inhibitor is selected from the compounds described in WO 2013/110890 and WO 2015/011400.
  • the Bcl-2 inhibitor comprises navitoclax (ABT-263), ABT-737, BP1002, SPC2996, APG-1252, obatoclax mesylate (GX15- 070MS), PNT2258, Zn-d5, BGB-11417, or oblimersen (G3139).
  • the Bcl-35 2 inhibitor is N-(4-hydroxyphenyl)-3-[6-[(3S)-3-(morpholinomethyl)-3,4-dihydro-1H-isoquinoline- PAT059646-WO-PCT 5 2-carbonyl]-1,3-benzodioxol-5-yl]-N-phenyl-5,6,7,8-tetrahydroindolizine-1-carboxamide, compound A1: (compound A1).
  • the Bcl-2 inhibitor is (S)-5-(5-chloro-2-(3-(morpholinomethyl)-10 1,2,3,4-tetrahydroisoquinoline-2-carbonyl)phenyl)-N-(5-cyano-1,2-dimethyl-1H-pyrrol-3-yl)-N-(4- hydroxyphenyl)-1,2-dimethyl-1H-pyrrole-3-carboxamide), compound A2: (compound A2).
  • the panRAS inhibitors or combinations disclosed herein are suitable for the treatment of cancer in vivo.
  • the combination can be used to inhibit the 15 growth of cancerous tumors.
  • the combination can also be used in combination with one or more of: a standard of care treatment (e.g., for cancers or infectious disorders), a vaccine (e.g., a therapeutic cancer vaccine), a cell therapy, a hormone therapy (e.g., with anti-estrogens or anti-androgens), a radiation therapy, surgery, or any other therapeutic agent or modality, to treat a disorder herein.
  • a standard of care treatment e.g., for cancers or infectious disorders
  • a vaccine e.g., a therapeutic cancer vaccine
  • a cell therapy e.g., a hormone therapy (e.g., with anti-estrogens or anti-androgens)
  • a radiation therapy e.g., surgery, or any other therapeutic agent or modality
  • the 20 combination can be administered together with an antigen of interest.
  • a combination disclosed herein can be administered in either order or simultaneously.
  • panRAS Inhibitors and Precursors thereof
  • Exemplary panRAS inhibitors and intermediates were synthesized using exemplary methods described in this example.
  • Materials, Methods & General Procedures Compounds of the present disclosure may be prepared by methods known in the art of organic synthesis. In all of the methods it is understood that protecting groups for sensitive or reactive groups may be employed where necessary in accordance with general principles of chemistry. 20 Protecting groups are manipulated according to standard methods of organic synthesis (T.W.
  • Tetramethylsilane was used as an i nternal standard. Chemical shifts are reported in ppm relative to dimethyl sulfoxide ( 2.50), methanol ( 3.31), chloroform ( 7.26) or other solvent as indicated in NMR spectral data. A small amount of the dry sample (2 to 5 mg) is dissolved in an appropriate deuterated solvent (1 mL). The chemical names were generated using ChemDraw Professional v22 from PerkinElmer.
  • Step b To a stirred solution of 1,2-di-tert-butyl 3-methyl 3,6-dihydropyridazine-1,2,3-tricarboxylate (7.50 g) in THF (20 mL) under a nitrogen atmosphere was added lithium borohydride (2 M in THF, 32.9 mL) dropwise.
  • the resulting mixture was stirred at 0°C for 2 hr then warmed to RT and stirred for 1 hr.
  • the reaction mixture was quenched by addition of sat. NaHCO 3 solution (20 mL), then diluted with EtOAc.
  • the layers were separated, and the aq. layer was extracted with EtOAc (3 x 20 20 mL).
  • the combined organic layers were washed with brine (20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure.
  • Step c To a stirred solution of di-tert-butyl 3-(hydroxymethyl)-3,6-dihydropyridazine-1,2- 10 dicarboxylate (5.30 g), DMAP (206 mg), and triethylamine (1.88 g) in DCM (30 mL) was added tert-butylchlorodiphenylsilane (4.63 g).
  • Step d To a vigorously stirred solution of di-tert-butyl 3-(((tert-butyldiphenylsilyl)oxy)methyl)-3,6- dihydropyridazine-1,2-dicarboxylate (150 mg) and benzyltriethylammonium chloride (2.2 mg) in DCM (0.50 mL) under a nitrogen atmosphere was added potassium hydroxide (8.0 M solution in water, 850 ⁇ L). The resulting mixture was cooled to 0°C, and bromoform (stabilized with ethanol, 1.71 g) was added dropwise over 30 min. using a syringe pump.
  • reaction mixture was stirred 25 at 0°C for 2 hr, then allowed to warm to RT and stirred for 16 hr.
  • the reaction was quenched by addition of water (2 mL) and DCM (10 mL), then passed through a phase separator, washing with DCM.
  • Step e To a stirred solution of di-tert-butyl 7,7-dibromo-2-(((tert-butyldiphenylsilyl)oxy)methyl)- 3,4-diazabicyclo[4.1.0]heptane-3,4-dicarboxylate (2.80 g) in toluene (20 mL) was added tributyltin hydride (5.62 g) and AIBN (317 mg). The resulting solution was purged with nitrogen gas and then stirred at 80°C for 2 hr. The reaction mixture was cooled to 0°C and diluted with EtOAc (20 mL), then quenched with 10% aq.
  • Step f To a stirred solution of di-tert-butyl 2-(((tert-butyldiphenylsilyl)oxy)methyl)-3,4- 15 diazabicyclo[4.1.0]heptane-3,4-dicarboxylate (2.10 g) in THF (10 mL) at 0°C was added TBAF (1.0 M in THF, 5.6 mL) and the resulting solution was stirred at 0°C for 1 hr. The reaction solution was then allowed to warm to RT and stir for 5 hr.
  • Step g To a stirred solution of di-tert-butyl 2-(hydroxymethyl)-3,4-diazabicyclo[4.1.0]heptane-3,4- dicarboxylate (1.05 g) in acetonitrile (10 mL) at RT under a nitrogen atmosphere was added TEMPO (30.0 mg), sodium chlorite (578 mg), and pH 7 phosphate buffer (10 mL). Vigorous stirring produced a pale-yellow emulsion, to which was added sodium hypochlorite (0.74 M aq. solution, 260 L) dropwise. The resulting deep-red mixture was stirred at RT for 4 hr, then cooled 30 to 0°C, and quenched with sat.
  • Step h Racemic 3,4-di-tert-butyl 2-methyl 3,4-diazabicyclo[4.1.0]heptane-2,3,4-tricarboxylate (670 mg) was purified by chiral SFC (Prep_SFC1 method) to give 3,4-di-tert-butyl 2-methyl 10 (1S,2S,6R)-3,4-diazabicyclo[4.1.0]heptane-2,3,4-tricarboxylate (260 mg) as the first eluting isomer.
  • Intermediate 2 Note: axial chirality of compounds is shown in below scheme.
  • PAT059646-WO-PCT 5 Procedure Step a: To a mixture of 3-bromo-5-fluoropicolinonitrile (910 g) in toluene (9.1 L) was added MeMgBr (3 M in 2-MeTHF, 1.96 L) dropwise at 5°C. The mixture was stirred at 5°C for 1 hr. The 10 reaction mixture was poured into 10% aq. HCl solution (4.55 L) and stirred for 30 min. at 10°C. The reaction mixture was partitioned between water and toluene and the aq. phase was extracted with toluene (4.55 L).
  • Step b To a mixture of 1-(3-bromo-5-fluoropyridin-2-yl)ethan-1-one (855 g) in DMF (8.55 L) was added benzyl piperazine-1-carboxylate (867 g) and K 2 CO 3 (742 g). The reaction was stirred at 50°C for 16 hr. The reaction mixture was poured into ice/water (17 L) and stirred at RT for 3 hr. The mixture was filtered and rinsed with water.
  • Step c To a mixture of benzyl 4-(6-acetyl-5-bromopyridin-3-yl)piperazine-1-carboxylate (533 g) in DCM (5.0 L) was added DIEA (463 g) and formic acid (275 g). The headspace of the flask was 15 evacuated and backfilled with nitrogen and then RuCl (p-cymene) [(S,S)-Ts-DPEN] (37.7 g) was added. The reaction was stirred at 35°C for 16 hr. The reaction mixture was cooled down to 5°C and water (2.5 L) was added. The resulting mixture was stirred for 30 min. at 25°C.
  • reaction mixture was partitioned between water and DCM; the organic layer was washed with 5% NaCl (2.5 L) and concentrated under reduced pressure at 40°C.
  • the residue was purified by column 20 chromatography (SiO 2 , 33% EtOAc in heptane) to afford the desired product as a brown solid (484 g).
  • Step d To a mixture of benzyl (S)-4-(5-bromo-6-(1-hydroxyethyl)pyridin-3-yl)piperazine-1- carboxylate (400.0 g) in DMF (3.6 L) was added t-BuOLi (105.0 g) at 25°C. Next, MeI (248 g) was dropwise to the mixture. The resulting reaction mixture was stirred at 25°C for 16. To the reaction mixture was added water (1.8 L) and stirred 25°C for 2 hr, which was filtered and rinsed with water (360 mL). The filter cake was treated with MTBE (1.5 L) and heptane (735 mL) at 25°C for 16 hr.
  • Step e A mixture of benzyl (S)-4-(5-bromo-6-(1-methoxyethyl)pyridin-3-yl)piperazine-1- carboxylate (132 g), bis(pinacolato)diboron (108 g), Pd(dppf)Cl 2 (22.2 g), KOAc (74.5 g) in toluene (1.2 L) was degassed and purged with nitrogen (3x) and stirred at 100°C for 10 hr. The reaction mixture was concentrated then redissolved in EtOAc (600 mL). Sulfhydryl silica gel (150 g) was added, and the mixture was stirred at 25°C for 30 min.
  • Step f To a solution of 5-bromo-3-(3-((tert-butyldiphenylsilyl)oxy)-2,2-dimethylpropyl)-2-iodo-1H- indole (141 g)) and benzyl (S)-4-(6-(1-methoxyethyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- 10 yl)pyridin-3-yl)piperazine-1-carboxylate (157 g) in dioxane (600 mL), water (200 mL) and toluene (200 mL) was added K 3 PO 4 (115 g) and Pd(dppf)Cl 2 (15.9 g).
  • Step h A mixture of benzyl (S)-4-(5-(5-bromo-3-(3-((tert-butyldiphenylsilyl)oxy)-2,2- dimethylpropyl)-1-ethyl-1H-indol-2-yl)-6-(1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate 35 (83.9 g) and CsF (42.3 g) in DMF (800 mL) was evacuated and backfilled with nitrogen (3x). The reaction was stirred at 66°C for 24 hr.
  • Tween80:lecithin (8:2 w:w ratio, 11 mL) and isopropanol (5.5 mL) was added.
  • the resulting solution was degassed for 5 min. via sparging with nitrogen.1,1'-Bis (di-t-butylphosphino)ferrocene palladium dichloride (18.1 20 mg) and triethylamine (282 mg) were sequentially charged to the degassed mixture and the solution was stirred at 0°C for 1 hr.
  • the reaction vessel was 10 evacuated and backfilled with hydrogen. The reaction stirred at RT for 3 h. The reaction mixture was filtered under nitrogen by washing with MeOH, and then concentrated to afford methyl (S)-2- ((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1- methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)-5- ((triisopropylsilyl)oxy)phenyl)propanoate (475 mg) as an orange foam.
  • Step c To a stirred solution of methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3- hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)-1H-indol-5- yl)-5-((triisopropylsilyl)oxy)phenyl)propanoate (475 mg) in isopropanol (5.0 mL) under nitrogen 20 was charged aq.
  • Step d To a solution of methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)- 5-((triisopropylsilyl)oxy)phenyl)propanoate (393 mg) in DCE (10 mL) was added trimethyltin hydroxide (389 mg) under a nitrogen atmosphere. The resulting suspension was stirred at 70°C 40 for 4 hr.
  • Step b A mixture of benzyl (S)-4-(5-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-5-(4,4,5,5-15 tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indol-2-yl)-6-(1-methoxyethyl)pyridin-3-yl)piperazine-1- carboxylate (270 mg), methyl (S)-3-(4-bromothiazol-2-yl)-2-((tert- butoxycarbonyl)amino)propanoate (153 mg), PdCl 2 (dtbpf) (24.8 mg) and potassium carbonate (105 mg) in 1,4-dioxane (2.0 mL) and water (0.4 mL) was stirred at 85°C for 3 hr under nitrogen.
  • Step c A flask containing benzyl 4-(5-(5-(2-((S)-2-((tert-butoxycarbonyl)amino)-3-methoxy-3- oxopropyl)thiazol-4-yl)-1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-1H-indol-2-yl)-6-((S)-1- methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (250 mg) and Pd(OH) 2 on carbon (212 mg, 35 20% wt.) in MeOH (2.0 mL) under nitrogen atmosphere was evacuated and backfilled with hydrogen (2x).
  • reaction stirred at RT for 2 h. Additional formaldehyde solution (24 L) and sodium triacetoxyborohydride 10 (45 mg) were added and the reaction stirred at RT for 1 hr. The reaction was quenched directly with a volumetric equivalent of sat. NaHCO 3 Solution. and DMSO and stirred vigorously. The mixture was then partially dried under reduced pressure to remove MeOH.
  • Step a part 2: To a stirred solution of Intermediate 3 (trifluoroacetic acid salt; 25.0 mg) and methyl (1S,2S,6R)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylate bis-trifluoroacetate salt (14.2 mg) in DMF (0.50 mL) was added DIPEA (52 ⁇ L) and HATU (18.7 mg). The resulting solution was stirred at RT for 30 min.
  • Intermediate 3 trifluoroacetic acid salt; 25.0 mg
  • methyl (1S,2S,6R)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylate bis-trifluoroacetate salt 14.2 mg
  • DIPEA 52 ⁇ L
  • HATU 18.7 mg
  • Step a part 2: To a solution of Intermediate 4 (47.8 mg) in NMP (0.10 mL) was added methyl (1S,2S,6R)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylate bis-trifluoroacetate salt (27.5 mg), 15 followed by DIPEA (0.11 mL) and HATU (49.5 mg) under nitrogen. The solution was stirred at RT for 30 min. The reaction mixture was diluted with minimal DMSO and purified by column chromatography (C18, 10 to 40% acetonitrile in water + 0.1% TFA) to yield a yellow solid after lyophilization. This solid was reconstituted in EtOAc and neutralized via vigorous stirring in the presence of sat.
  • Step a To a solution of N-methyl-L-valine benzyl ester 4-toluenesulfonate (464 mg) in DCM (6.2 35 mL) at 0°C was added DIPEA (1.23 mL). The solution was stirred at 0°C for 10 min. Triphosgene (174.9 mg) was added and the reaction was stirred at 0°C for 10 min, then allowed to warm to RT and stirred for 30 min. Morpholine (103 mg) was added to the reaction at 0°C. The reaction mixture was warmed to RT and stirred for 1 hr. The reaction mixture was quenched with sat.
  • Step b A flask containing benzyl N-methyl-N-(morpholine-4-carbonyl)-L-valinate (231 mg) and Pd on carbon (732 mg, 10 wt%) in MeOH (8.1 mL) at RT was evacuated and backfilled with nitrogen. Triethylsilane (240 mg) was added over the course of 5 min. The reaction was stirred at RT for 30 min. The reaction mixture was filtered over a pad of Celite®, rinsed with methanol, and 15 then concentrated to yield N-methyl-N-(morpholine-4-carbonyl)-L-valine (171 mg) as a colorless oil that solidifies over time.
  • Step b To a solution of 3-bromo-5-iodo-2-isopropoxypyridine (1.04 g) and xantphos (176 mg) in toluene (6.1 mL) was added 1-methyl piperazine (320 mg), bis(dibenzylideneacetone)dipalladium (137 mg), and sodium tert-butoxide (2M in THF, 3.80 mL). The solution was sparged with nitrogen for 5 min. then stirred at 60°C for 20 min. The reaction was cooled to RT and poured into brine. 35 The aq.
  • Step c A vial containing 1-(5-bromo-6-isopropoxypyridin-3-yl)-4-methylpiperazine (209 mg), bis(pinacolato)diborane (338 mg), potassium acetate (196 mg), and PdCl 2 (dtbpf) (43.4 mg) was evacuated and backfilled with nitrogen.
  • reaction mixture was stirred at 85 C for 4 hr.
  • the resulting mixture was combined with other two batches (5.0 g and 9.3 g of 3-(5-bromo-1H-indol-3-yl)-2,2-dimethylpropyl acetate were used in the same manner) for the subsequent workup.
  • the reaction mixture was concentrated under reduced pressure to PAT059646-WO-PCT 5 remove dioxane and water.
  • Step b To a solution of methyl (S)-3-(3-(3-(3-acetoxy-2,2-dimethylpropyl)-1H-indol-5-yl)-5- ((triisopropylsilyl)oxy)phenyl)-2-((tert-butoxycarbonyl)amino)propanoate (65.0 g) and NaHCO 3 15 (9.3 g) in THF (650 mL) was added AgOTf (22.7 g) in THF (100 mL) and I 2 (16.8 g) in THF (100 mL) at 0 C. The reaction mixture was stirred at 0 C for 2 hr.
  • Step c To a solution of methyl (S)-3-(3-(3-(3-acetoxy-2,2-dimethylpropyl)-2-iodo-1H-indol-5-yl)-5- ((triisopropylsilyl)oxy)phenylycarbo)-2-((tert-butoxnyl)amino)propanoate (750 mg) in DCE (5.0 mL) was added trimethyltin hydroxide (496 mg). The reaction was stirred at 80°C for 24 hr. 35 Additional trimethyltin hydroxide (248 mg) was added and the reaction mixture stirred at 80°C for 24 hr.
  • the reaction was cooled to RT, 25 diluted with water, stirred vigorously for 5 min, diluted with DCM, passed through a phase separator, rinsed with DCM, and concentrated under reduced pressure.
  • the crude material was purified by column chromatography (C18, 20 to 100% acetonitrile in water + 0.1% NH 4 OH). Fractions containing the product were concentrated and sat. NaHCO 3 solution (15 mL) was added.
  • Step h To a solution of tert-butyl ((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl-1 2 -iodo-10,10-dimethyl-5,7-dioxo-2 5 - ((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)carbamate (328 mg), Intermediate 8 (crude, 198 mg), aq.
  • K 3 PO 4 30 solution 155 mg, 730 L, 1.0 molar
  • mesylate[(di(1-adamantyl)-n-butylphosphine)-2-(2 - amino-1,1 -biphenyl)]palladium(II) 26.6 mg
  • 1,4-dioxane 3.0 mL
  • the reaction mixture was poured into separatory funnel and diluted with sat. NaHCO3 solution.
  • the aq. layer was extracted with EtOAc (3x), and the combined organic layers were washed with brine and concentrated.
  • the crude 35 material was purified by column chromatography (C18, 50 to 100% acetonitrile in water + 0.1% TFA) to yield tert-butyl ((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl-1 2 -(2-isopropoxy-5-(4-methylpiperazin-1- yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 -diaza-1(5,3)- indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)carbamate (200 mg) as a brownish orange solid.
  • the cylinder was irradiated with total 801W high power LEDs (365 nm) for 24 hr, and then the reaction mixture was concentrated at 40°C to yield 68.2 g of a viscous pale-brown oil that was dissolved in 250 mL MTBE and washed twice with 100 mL water followed by 216 mL 1 N NaOH solution.
  • the basic aqueous solution was washed with 150 mL MTBE then acidified by slow addition of 12.8 g conc. H 2 SO 4 to a pH of ⁇ 3 and extracted with MTBE.
  • Step b To a solution of a crude cis/trans mixture of 3-phenyloxetane-2-carboxylic acid (25.9 g) in 150 mL absolute EtOH was added AmberChrom TM , 50WX4 (5.9 g) and the suspension heated at 60°C for 2 hr, then at 70°C for another 4.5 hr. The reaction mixture was cooled to RT, filtered, and concentrated to yield 28.0 g of orange oil.
  • Step c Ethyl cis-3-phenyloxetane-2-carboxylate (1.15 g) was dissolved in EtOH + 0.05 NH 3 (18 mL) and then purified by chiral SFC (Prep_SFC2 method) to give ethyl (2R,3S)-3-phenyloxetane- 2-carboxylate as the first eluting isomer as colorless oil (514 mg; 98% ee).
  • LC/MS Fluor_Analysis_2min method): m/z 207 [M+H] + .
  • Step b To a solution of (S)-4-(5-bromo-6-(1-methoxyethyl)pyridin-3-yl)morpholine (1.88 g) in15 DMSO (25 mL), 4,4,5,5-tetraethyl-2-(4,4,5,5,-tetraethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (5.71 g), Pd(dppf)Cl 2 -DCM adduct (127 mg) and potassium phenoxide (1.90 g) were added.
  • Step b To a solution of (S)-8-(5-bromo-6-((S)-1-methoxyethyl)pyridin-3-yl)octahydropyrazino[2,1- c][1,4]oxazine (281 mg) and bis(pinacolato)diboron (301 mg) in 2-MeTHF (7.7 mL) was added potassium 2-ethylhexanoate (316 mg) and Pd(dppf)Cl 2 (86.6 mg). The reaction was purged with nitrogen (3x) and heated at 80°C for 2 hr. The reaction was partitioned with 2% wt. aq. NaHCO 3 . The aq.
  • Step a To a suspension of racemic cis-methyl 4-phenylpyrrolidine-3-carboxylate hydrochloride 10 (2 g) and NaHCO 3 (2.09 g) in DCM (30 mL) and water (3 mL) was added methyl carbonochloridate (1.28 mL). The reaction was stirred at RT for 30 min. The reaction mixture was diluted with water, extracted with DCM (2x), passed through a phase separator, and concentrated.
  • Step b Racemic cis-dimethyl 4-phenylpyrrolidine-1,3-dicarboxylate (1.68 g) was purified by chiral SFC (Prep_SFC3 method) to yield dimethyl (3S,4S)-4-phenylpyrrolidine-1,3-dicarboxylate (0.766 g) as the first eluting isomer, a pale orange solid.
  • Step c A mixture of dimethyl (3S,4S)-4-phenylpyrrolidine-1,3-dicarboxylate (150 mg) and trimethyltin hydroxide (155 mg) in DCE (5.6 mL) was stirred at 85°C. After stirring for 24 hr, 25 additional trimethyltin hydroxide (52 mg) was added. After stirring for 6 hr, additional trimethyltin hydroxide (103 mg) was added. The reaction stirred for a total of 4 days. The reaction mixture was concentrated, suspended in EtOAc and 1N aq. HCl, and the phases were separated. The aq.
  • Step b Dimethyl (3S,4S)-4-(4-iodophenyl)pyrrolidine-1,3-dicarboxylate (105 mg), tert-butyl 10 piperazine-1-carboxylate (53 mg), Cs 2 CO 3 (219 mg), Pd 2 (dba) 3 (7.4 mg), and XPhos (7.7 mg) were dissolved in toluene (2.7 mL) and sparged with nitrogen for 5 min. The mixture was then stirred at 110°C for 20 h, after which time it was filtered and concentrated.
  • Step c To a solution of crude dimethyl (3S,4S)-4-(4-(4-(2-hydroxyacetyl)piperazin-1- yl)phenyl)pyrrolidine-1,3-dicarboxylate (37 mg) was added trimethyltin hydroxide (61 mg) in DCE (800 ⁇ L). The reaction was stirred at 85°C for 3 days, after which time the reaction mixture was concentrated and the residue treated with aq.1N HCl and EtOAc. The aq. layer was extracted PAT059646-WO-PCT 5 with EtOAc, and the combined organic extracts were washed with brine, passed through a phase separator, and concentrated.
  • Step b To a solution of ethyl (2R,3R)-3-phenyltetrahydrofuran-2-carboxylate (989 mg) and Ag(OTf) (2.31 g) in DCM (40 mL) and AcOH (135 mg, 129 L) at 0 °C was added I 2 (2.28 g). The reaction mixture was allowed to warm to RT and stir for 1.5 hr, after which time it was poured into PAT059646-WO-PCT 5 dilute aq. NH4OH and sat. aq. sodium thiosulfate. The resulting precipitate was filtered, and the filtrate was extracted with DCM (3x).
  • Step c A mixture of (2R,3R)-3-(4-iodophenyl)tetrahydrofuran-2-carboxylate (300 mg), tert-butyl 15 piperazine-1-carboxylate (254 mg), Cs 2 CO 3 (1.11 g), Pd 2 dba 3 (31.2 mg), and XPhos (32.5 mg) in toluene (12 mL) was sparged with nitrogen for 5 min. and heated at 110°C for 20 hr. The reaction mixture was filtered over a pad of Celite® and concentrated.
  • reaction solution was cooled to 0 °C and aq.1N NaOH (949 L) was added. Following this, Boc 2 O (99 mg) was added, and the reaction was allowed to warm to RT. Additional aq.1N NaOH (949 L) and Boc 2 O (99 mg) were added.
  • the reaction stirred for total of 29 hr, after which time the pH was adjusted to 10 with aq.1N NaOH.
  • the mixture was washed with Et 2 O and the 25 aq. layer was then acidified to pH 2 with aq.1N HCl and extracted with EtOAc (3x).
  • Step d To a solution of (tert-butoxycarbonyl)-L-alloisoleucine (70 mg) in THF (2.0 mL) at 0 °C was sequentially added MeI (344 mg, 151 L) and NaH (73 mg, 60% wt.). The reaction was allowed to warm to RT and stir for 28 hr. The reaction was then diluted with Et2O and washed with water (15 mL, 2x). Combined aq. extracts were acidified with citric acid (pH 3) and extracted 35 with EtOAc. Combined organic extracts were dried over Na 2 SO 4 , filtered, and concentrated.
  • Step a To a solution of methyl (tert-butoxycarbonyl)-L-threoninate (3 g) in acetonitrile (130 mL) 10 at 0 °C under nitrogen was added Ag 2 O (13.4 g) and MeI (14.1 g, 6.19 mL). The reaction allowed to warm to RT and stir for 4 days. After this time, the reaction was filtered, rinsed with acetonitrile, and concentrated.
  • Step b To a solution of N-(tert-butoxycarbonyl)-O-methyl-L-threoninate (2.39 g) in THF (16 mL) and MeOH (16 mL) at 0 °C was added aq.1.0N LiOH (810 mg, 34 mL). The reaction was allowed to warm to RT and stir for 2.5 hr. After this time, the mixture was cooled to 0 °C, 1N aq. HCl (21 20 mL) was added, and the mixture was concentrated. The remaining aq. mixture was extracted with EtOAc (3x), the organic layers were passed through a phase separator.
  • Step c To a solution of N-(tert-butoxycarbonyl)-O-methyl-L-threonine (500 mg) in THF (20 mL) at 0 °C under a nitrogen atmosphere was added NaH (176 mg, 60% wt.) in one portion. After 30 min., MeI (1.53 g, 670 L) and DMF (332 L) were added. The solution stirred at 0 °C for 2 hr then was allowed to warm to RT and stir for 20 hr. The reaction mixture was quenched with water, 30 diluted with aq. HCl and brine. The aq. layer was extracted with EtOAc (3x). The combined organic extracts were passed through a phase separator and concentrated.
  • the resulting 20 mixture was stirred at RT for 16 hr, upon which time additional LiBr (1 g) was added, and the PAT059646-WO-PCT 5 mixture was then heated to 50 °C for 4 hr. After this time, the reaction was cooled to RT and stirred for 48 hr, during which time a precipitate formed.
  • the first crop of solids was collected via vacuum filtration and rinsed with cold acetonitrile. The filtrate was concentrated to half of its volume and cooled to 5 °C to precipitate additional product.
  • This second crop was isolated via vacuum filtration and combined with the first crop to afford (2S)-3-(3-(2-(5-(4-10 ((benzyloxy)carbonyl)piperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-1-ethyl-3-(3-hydroxy- 2,2-dimethylpropyl)-1H-indol-5-yl)-5-((triisopropylsilyl)oxy)phenyl)-2-((tert- butoxycarbonyl)amino)propanoic acid (10.5 g) as a white solid after drying.
  • Step b part 1: To Intermediate 1 (220 mg) in DCM (35 mL) was added TFA (35 mL) and the solution was stirred at RT for 2 hr, after which time, the reaction mixture was concentrated and the residue was azeotropically dried with heptane and left to dry in vacuo for 16 hr to afford methyl (1S,2S,6R)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylate, trifluoroacetate salt (3.35 g) which was advanced directly to subsequent amide coupling.
  • Step b, part 2 Crude methyl (1S,2S,6R)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylate, trifluoroacetate salt was reconstituted in DMF (30 mL) and (2S)-3-(3-(2-(5-(4- ((benzyloxy)carbonyl)piperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-1-ethyl-3-(3-hydroxy- 2,2-dimethylpropyl)-1H-indol-5-yl)-5-((triisopropylsilyl)oxy)phenyl)-2-((tert- 25 butoxycarbonyl)amino)propanoic acid (10.5 g) was added.
  • the reaction apparatus was purged with nitrogen (3x) and the solution was cooled to 0 °C.
  • DIPEA (4.25 g, 5.73 mL) was added, and the mixture was stirred for 5 min., after which time HATU (3.93 g) was added, and the solution was allowed to stir for 5 min. at °C and then warm to RT. After stirring at RT for 1 hr, ice (75 mL) was added to triturate the product.
  • Step b To a solution of benzyl 4-(5-((6 1 R,6 5 S,6 6 S,4S)-4-amino-1 1 -ethyl-10,10-dimethyl-5,7- dioxo-2 5 -((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-20 2(1,3)-benzenacycloundecaphane-1 2 -yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1- carboxylate trifluoroacetate salt (370 mg) in DMF (2 mL) a 0 °C under nitrogen, was added Boc- N-Me-L-valine (101 mg) in DMF (2 mL) a 0 °C under nitrogen, was added Boc- N-Me-L-va
  • reaction mixture was allowed to warm to RT and stir for 15 min. After this time, the reaction mixture was diluted with water and extracted with EtOAc. The EtOAc extract was washed with sat. aq. NaHCO 3 , brine, 25 dried over Na 2 SO 4 , filtered, and concentrated.
  • Step c To a solution of benzyl 4-(5-((6 1 R,6 5 S,6 6 S,4S)-4-((S)-2-((tert- butoxycarbonyl)(methyl)amino)-3-methylbutanamido)-1 1 -ethyl-10,10-dimethyl-5,7-dioxo-2 5 - ((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-1 2 -yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate 15 (280 mg) in DCM (2 mL)
  • the reaction mixture was purged with H 2 and stirred under an H 2 atmosphere at RT for 30 min., after which time Celite® and water were added.
  • the mixture was filtered, and the filter cake was washed with methanol (10 mL).
  • the combined filtrate was concentrated, and the residue was purified by column chromatography (C18; 10 to 100% acetonitrile in water + 0.1% TFA).
  • the product fraction was concentrated, naturalized with sat. PAT059646-WO-PCT 5 aq. NaHCO3 and extracted with EtOAc.
  • Step b To a solution of methyl (S)-3-(5-(benzyloxy)-2-fluorophenyl)-2-((tert- butoxycarbonyl)amino)propanoate (1.23 g) in MeOH (15 mL) under nitrogen atmosphere was 20 charged Pd(OH) 2 on carbon (2.14 g, 10% wt.). The mixture was purged with H 2 and allowed to stir under a H 2 atmosphere for 2 hr, after which time the mixture was filtered and concentrated to afford crude methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(2-fluoro-5-hydroxyphenyl)propanoate (1.014 g) which was used without further purification.
  • Step c To a solution of crude methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(2-fluoro-5- hydroxyphenyl)propanoate (1.01 g) in 2-MeTHF (10 mL) under nitrogen atmosphere was sequentially charged imidazole (621 mg) and TIPS-Cl (880 mg, 976 ⁇ L). The reaction mixture was left to stir at RT for 16 hr.
  • Step c To a solution of methyl (2S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy- 2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)-2- fluoro-5-((triisopropylsilyl)oxy)phenyl)propanoate (80 mg) in MeOH (3 mL) was added aq.
  • reaction was cooled to RT and diluted with 10 volumetric equivalents of DCM.
  • the mixture was quenched with aq.0.05N sodium bisulfite (37.54 mL) with vigorous stirring for 5 min. The layers were separated. The aq.
  • the reaction was stirred for 1 hr and additional methyl carbonochloridate (11.0 mg, 9 L) and NEt 3 (67.5 mg, 93 L) were added.
  • the reaction stirred at RT for a total of 2 hr.
  • the reaction mixture was diluted with DCM and washed with brine.
  • the aq. layer was extracted with DCM (3x), and the combined organic extracts were passed through a phase separator and concentrated.
  • reaction was allowed to warm to RT and stirred at RT for 40 min.
  • the reaction mixture was diluted with EtOAc and washed with sat. aq. NaHCO 3 and brine.
  • the aq. layer was extracted with EtOAc (3x).
  • Step d A solution of 4-(5-((6 1 R,6 5 S,6 6 S,4S)-4-((2S,3R)-2-((tert-butoxycarbonyl)(methyl)amino)-3- methylpent-4-enamido)-1 1 -ethyl-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-1 1 H-8-oxa-40 6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-1 2 -yl)- 6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (95 mg) in DCM (650 L) was PAT059646-WO-PCT 5 charged TFA (160 L).
  • Step b To a solution of methyl 4-(5-((6 1 R,6 5 S,6 6 S,4S)-4-((2S,3R)-2-((3S,4S)-1-(tert- butoxycarbonyl)-N-methyl-4-phenylpyrrolidine-3-carboxamido)-3-methylpent-4-enamido)-1 1 -25 ethyl-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola- 6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-1 2 -yl)-6-((S)-1- methoxyethyl)pyridin-3-yl
  • Step b To a solution of (6 1 R,6 5 S,6 6 S,4S)-4-amino-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)-5-(4-20 methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-2 5 -((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 - diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-5,7-dione trifluoroacetate (1.45 g), N-(tert-butoxycarbonyl)-N-methyl-L-valine (515 mg), and DIPEA (1.00 mg, 1.16 mL) in NMP (10
  • Step c A solution of tert-butyl ((2S)-1-(((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)-5-15 (4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-1 1 H-8- oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)amino)-3-methyl-1-oxobutan-2-yl)(methyl)carbamate (657 mg) in DCM (4.8 mL) and TFA (1.2 mL) was stirred at RT for 1 hr.
  • Step b To a solution of tert-butyl 4-(4-((3S,4S)-4-(((2S,3R)-1-(((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl-1 2 -(5- (4-(methoxycarbonyl)piperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-2 5 -((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-25 2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopent-4
  • the reaction apparatus was evacuated and backfilled with hydrogen (3x) and the suspension was left stirring at RT under hydrogen 25 atmosphere for 2 hr. After this time, the apparatus was purged with nitrogen and the reaction mixture was filtered through a pad of Celite® topped with Na 2 SO 4 . The filter cake was thoroughly conditioned with MeOH.
  • Step b To a solution of tert-butyl ((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)-5-10 (piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 - diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)carbamate (706 mg) in 2-MeTHF (10 mL) under nitrogen atmosphere was charged Et 3 N (180 mg, 248 ⁇ L).
  • reaction mixture was PAT059646-WO-PCT 5 concentrated directly onto silica and purified by column chromatography (SiO2; 0 to 100 EtOAc/EtOH 3:1 in heptane) to afford (9H-fluoren-9-yl)methyl 4-(5-((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl- 10,10-dimethyl-4-((2S,3R)-3-methyl-2-((2R,3S)-N-methyl-3-phenyloxetane-2-carboxamido)pent- 4-enamido)-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-1 2 -yl)-6
  • Step b A mixture of benzyl ((1S)-1-cyclopentyl-2-(((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -15 ((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-2-oxoethyl)(methyl)carbamate (16 mg) and Pd on carbon (7.2 mg, 10%) in MeOH (
  • reaction mixture was allowed to stir at RT for 2 hr under nitrogen.
  • the reaction was diluted with EtOAc and water and stirred vigorously. The layers were separated.
  • the aq. layer 35 was extracted with EtOAc (3x), and the combined organic extracts were washed with brine, dried over Na 2 SO 4 , filtered and concentrated.
  • PAT059646-WO-PCT 5 Step a To a solution of Intermediate 5 (60 mg) in DCM (994 L) was added TFA (199 L). The reaction was stirred at RT for 40 min., after which time it was concentrated. The resulting yellow foam was dissolved in DCM and concentrated to dryness; this process was repeated (3x). To the 10 remaining residue was triturated with Et 2 O.
  • Step b To a vial charged with Intermediate 23 (15.6 mg) and HATU (24.4 mg) was charged DMF (641 L) and DIPEA (41.4 mg, 55.8 L) and the mixture was stirred at RT for 2 min.
  • Step b To a vial charged with Intermediate 24 (14 mg) and HATU (20 mg) was charged DMF (513 L) and DIPEA (27 mg, 35.8 L) and the mixture was stirred at RT for 2 min.
  • PAT059646-WO-PCT 5 Table 8: PanRAS inhibitor compounds PAT059646-WO-PCT 194 PAT059646-WO-PCT PAT059646-WO-PCT 196 PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT 5 Compound 123 Note: axial chirality of compounds is as shown in below scheme.
  • Step b To a solution of tert-butyl ((1 2 R,6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)-5-(piperazin- 1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro- 1 1 H-8-oxa-1(5,3)-indola-6(1,3)-pyridazina-2(1,3)-benzenacycloundecaphane-4-yl)carbamate (3.0 g) in MeOH (29 mL) was added formaldehyde (701 mg, 643 L, 37
  • the reaction was stirred for 10 min. and sodium triacetoxyborohydride (1.83 g) was added. The 20 reaction was stirred 30 min. Sat. aq. NaHCO3 solution was slowly added to the reaction. The aq. layer was extracted with DCM (2x).
  • Step c To a solution of tert-butyl ((1 2 R,6 3 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)- PAT059646-WO-PCT 5 6 1 ,6 2 ,6 3 ,6 4 ,6 5 ,6 6 -hexahydro-1 1 H-8-oxa-1(5,3)-indola-6(1,3)-pyridazina-2(1,3)- benzenacycloundecaphane-4-yl)carbamate (3.57 g) in DCM (36 mL) and triethylsilane (2
  • the reaction was stirred at RT for 2 hr. The reaction was concentrated, and the residue was dissolved in DCM. The DCM solution was washed with sat. aq. NaHCO 3 solution (0°C). The aq. layer was extracted with DCM (2x).
  • the reaction was warmed up to RT and stirred for 30 min.
  • the reaction was concentrated, dissolved in EtOAc, and washed with water, sat. aq. NaHCO 3 and brine.
  • the EtOAc layer was dried over Na 2 SO 4 and concentrated.
  • the 35 reaction was stirred at RT for 30 min. The reaction was concentrated, and the residue was dissolved in DCM. The DCM solution was washed with sat. aq. NaHCO 3 solution. The aq. layer was extracted with DCM (3x).
  • the reaction was warmed up to RT and stirred for 20 hr.
  • the reaction was concentrated, dissolved in EtOAc and filtered.
  • the filtrate 15 was washed with water, sat. aq. NaHCO 3 and brine.
  • the EtOAc layer was dried over Na 2 SO 4 and concentrated.
  • Step b To a solution of (2R,3S)-1-(cyanomethyl)-N-((2S)-1-(((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl-1 2 -(2- ((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 - ((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-35 benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3-phenyla
  • the reaction vessel was purged with hydrogen (3x). The mixture was stirred under hydrogen atmosphere at RT for 1.5 hr, after which time additional Pd(OH) 2 on carbon (15 mg, 10% wt.) was added and the reaction resumed for 50 min. The reaction mixture was filtered25 and washed with DCM.
  • Step b To the crude mixture of (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1- methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)- 11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-15 benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N
  • Step b To a mixture containing (2R,3S)-N-((2S)-1-(((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl-1 2 -(5-(4-(4- hydroxybutyl)piperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 - ((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N
  • Step b To a solution of tert-butyl ((6 1 R,6 5 S,6 6 S,4S)-1 2 -(5-(4-cyclopropylpiperazin-1-yl)-2-((S)-1- methoxyethyl)pyridin-3-yl)-1 1 -ethyl-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-1 1 H-8- PAT059646-WO-PCT 5 oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)carbamate (140 mg) in DCM (2 mL) was added TFA (1 mL).
  • Step c To a solution of (6 1 R,6 5 S,6 6 S,4S)-4-amino-1 2 -(5-(4-cyclopropylpiperazin-1-yl)-2-((S)-1-15 methoxyethyl)pyridin-3-yl)-1 1 -ethyl-10,10-dimethyl-2 5 -((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 - diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-5,7-dione trifluoroacetate salt in DMF (1 mL) was added N-(tert-butoxycarbonyl)-N-methyl-L-valine (60.3 mg), DIPEA (134.7 mg,
  • Step d To a solution of tert-butyl ((2S)-1-(((6 1 R,6 5 S,6 6 S,4S)-1 2 -(5-(4-cyclopropylpiperazin-1-yl)- 2-((S)-1-methoxyethyl)pyridin-3-yl)-1 1 -ethyl-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)- 1 1 H-8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- 30 benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)
  • Step b To a solution of crude methyl 4-(5-((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl-4-((2S,3R)-2-((3S,4S)-1- (methoxycarbonyl)-N-methyl-4-phenylpyrrolidine-3-carboxamido)-3-methylpent-4-enamido)-20 10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-1 2 -yl)-6-((S)-1-methoxyethyl)pyridin-3- yl)pipe
  • Step b To a solution of methyl 4-(5-((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl-4-((2S,3R)-2-((3S,4S)-1-20 (methoxycarbonyl)-N-methyl-4-(4-(4-methylpiperazin-1-yl)phenyl)pyrrolidine-3-carboxamido)-3- methylpent-4-enamido)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 - diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-1 2 -yl)-6-((S)- 1-methoxye
  • reaction mixture was sonicated then stirred at RT for 30 min.
  • the reaction mixture 25 was filtered, diluted with DMSO, and purified by preparatory HPLC (50 to 70% acetonitrile in water (+0.1% NH 4 OH)), and concentrated. The material was dissolved in DCM and washed with water.
  • PAT059646-WO-PCT 5 The aq.
  • reaction 35 was then stirred at 40°C for 24 hr, representing a total reaction time of 44 hr.
  • Step b To a solution of crude methyl 4-(5-((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl-4-((2S,3R)-2-((3S,4S)-4-(4- (4-(2-hydroxyacetyl)piperazin-1-yl)phenyl)-1-(methoxycarbonyl)-N-methylpyrrolidine-3- carboxamido)-3-methylpent-4-enamido)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-1 1 H- 8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane- 15 1 2 -yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperaz
  • Step c To a solution of benzyl (2R)-4-(5-((6 1 R,6 5 S,6 6 S,4S)-4-((tert-butoxycarbonyl)amino)-1 1 - ethyl-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola- 6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-1 2 -yl)-6-((S)-1- 20 methoxyethyl)pyridin-3-yl)-2-methylpiperazine-1-carboxylate (217 mg) in MeOH (3 mL) was added Pd(OH) 2 on
  • the reaction vessel was evacuated and backfilled with H 2 .
  • the reaction was stirred under an atmosphere of H 2 at RT for 16 hr.
  • the reaction mixture was filtered and concentrated to afford tert-butyl ((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1- methoxyethyl)-5-((R)-3-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -25 ((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)carbamate (182 mg).
  • Step d To a solution of tert-butyl ((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)-5-((R)-3-30 methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-1 1 H-8-oxa- 6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)carbamate (182 mg) in MeOH (2.7 mL) was added aq.
  • Step f To a solution of (6 1 R,6 5 S,6 6 S,4S)-4-amino-1 2 -(5-((R)-3,4-dimethylpiperazin-1-yl)-2-((S)-1- methoxyethyl)pyridin-3-yl)-1 1 -ethyl-10,10-dimethyl-2 5 -((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 - diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-5,7-dione trifluoroacetate salt in DMF (343 L) was added N-(tert-butoxycarbonyl)-N-methyl-L-
  • the reaction was stirred at 80°C under nitrogen for 2 hr. The reaction was then diluted with EtOAc 10 and water. The aq. layer was extracted with EtOAc (2x). The EtOAc extracts were washed with brine, dried with Na 2 SO 4 , filtered, and concentrated.
  • Step c To a solution of tert-butyl ((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl-1 2 -(5-((S)-hexahydropyrazino[2,1- c][1,4]oxazin-8(1H)-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 - ((triisopropylsilyl)oxy)-1 1 H-8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)carbamate (232 mg) in DCM (2 m
  • reaction 40 was purified by preparatory HPLC (C18; 25 to 50% acetonitrile in water + 0.1% NH 4 OH) to afford (2R,3S)-N-((2R,3R)-1-(((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl-1 2 -(5-((S)-hexahydropyrazino[2,1- PAT059646-WO-PCT 5 c][1,4]oxazin-8(1H)-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-2 5 -hydroxy-10,10-dimethyl-5,7-dioxo- 1 1 H-8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundeca
  • Step b To a solution of tert-butyl ((6 1 R,6 5 S,6 6 S,4S)-1 1 -ethyl-1 2 -(2-((S)-1-methoxyethyl)-5-(4- (oxetan-3-yl)piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-2 5 -((triisopropylsilyl)oxy)-1 1 H- 8-oxa-6 3 ,6 4 -diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane- 15 4-yl)carbamate (21.0 mg) in DCM (150 L) was added TFA (50 L).
  • Step f To the reaction mixture from Step e was added ammonium hydroxide (10.6 L, 30% wt.). 20 The reaction stirred for 15 min. then TMAF (6 mg) was added. The mixture was sonicated then stirred for 30 min. at RT. The reaction mixture was diluted with DMSO, filtered, and purified by HPLC (50-70% CH 3 CN in H 2 O + 12 mM NH 4 OH). The fractions containing the desired product were partially concentrated and the aqueous layer was extracted with DCM (3x). The combined organic layers were passed through a phase separator, concentrated, and lyophilized to afford 25 Compound 159 (9.2 mg) as an off-white solid.
  • NRAS_Q61R compound 101 and CYPA were mixed at 1.25mM : 1mM : 1mM molar ratio in a buffer of 20mM HEPES pH 7.5, 100mM NaCl.
  • the protein complex was crystallized under 0.1 M Bis-Tris propane/HCl, pH 7 and 2 M ammonium citrate dibasic using sitting drop vapor diffusion method. Single crystal was harvested and diffracted under synchrotron radiation. 10
  • the dataset was processed to 1.94 angstrom resolution. Structure was solved by molecular replacement and refined to Rwork/Rfree: 0.182/0.230.
  • NRAS_Q61R is a peptide with the following amino acid sequence: SEQ ID No.1: GPMTEYKLVVVGAGGVGKSALTIQLIQNHFVDEYDPTIEDSYRKQVVIDGETCLLDILDTAG 15 REEYSAMRDQYMRTGEGFLCVFAINNSKSFADINLYREQIKRVKDSDDVPMVLVGNKCDL PTRTVDTKQAHELAKSYGIPFIETSAKTRQGVEDAFYTLVREIRQYRMK.
  • CypA is a peptide with the following sequence: SEQ ID No.2: GMVNPTVFFDIAVDGEPLGRVSFELFADKVPKTAENFRALSTGEKGFGYKGSCFHRIIPGFMCQ 20 GGDFTRHNGTGGKSIYGEKFEDENFILKHTGPGILSMANAGPNTNGSQFFICTAKTEWLDGKH VVFGKVKEGMNIVEAMERFGSRNGKTSKKITIADCGQLE.
  • the structure including axial chirality was determined to be according to the below structure: 25 The axial chirality of 101 derives from the axial chirality of Intermediate 2.
  • cell viability and 20 cell density were determined using a cell counter (Vi-Cell XR Cell Viability Analyzer, Beckman Coulter). Cells with higher than 85% viability were seeded in white clear bottom 384-well TC treated plates (Corning cat. # 3765). Cells were seeded at a density of 1,000 cells per well in 45 L of standard growth media. Plates were incubated at 5% CO 2 , 37°C overnight in a tissue culture incubator. The next day, indicated compounds were prepared at 10X in standard growth media. 25 The prepared compounds were then added to the cells resulting in final concentrations of 0.005 – 100 nM and a final volume of 50 uL per well. Each compound concentration was tested in quadruplets.
  • IC50 35 and Amax values were extrapolated from the resultant curves.
  • concentrations of treatment required to inhibit 50% of cell growth or survival (GI50) were calculated with representative GI50 values of the cell lines tested summarized in Table 17 along with associated structures in Table 20.
  • 40 Cell activities PAT059646-WO-PCT 5
  • Table 17 Biological assay results for panRAS inhibitors
  • PAT059646-WO-PCT 5 Binary SPR assay Materials and instruments: 1. Avi-tagged Cyclophilin A (human) 10 2. Streptavidin(SA) sensor chip: Cytiva series S Cat# BR100531 3.
  • SPR assay buffer 20 mM HEPES pH 7.5, 150 mM NaCl, 1 mM TECP, 0.025% P20, 2% DMSO 4.8K Biacore: Cytiva 5. Biomek liquid handler: Beckman 15 6. ECHO 550: Labcyte 7. Dragonfly Discovery: SPT Labtech Method: The binary SPR assay was used to measure the direct binding between a Pan-RAS inhibitor 20 and Cyclophilin A (CypA). The assay was run using the multi-cycle kinetics binding assay format with 8K Biacore instrument employing 10-points and 1 to 3 serial dilution compound plate (prepared with ECHO instrument).0% DMSO assay buffer was added to the plate to DMSO final concentration at 2%.
  • Compound final concentration was from 10 uM to 0.51 nM and the last point was a blank control.
  • Avi-CypA was immobilized to SA-chip surface to 300 RU level. 25 The assay was run at 25°C . Flow rate was set to 50 ⁇ l/min, contact time 60 sec., dissociation time 600 sec. and 15 start-up cycles. DMSO solvent correction was run before and after the sample running. All analysis of data were performed using Cytiva’s Biacore Insight Evaluation Software. The predefined evaluation method “LMW multi-cycle kinetics” was selected for data analysis.1:1 binding kinetics fit model was applied, and steady state affinity fit mode was 30 applied to weak compounds which could’t be analyzed with kinetics fit mode.
  • the ternary SPR assay was used to measure the binding between KRAS G12C and (Pan-RAS 20 inhibitor + Cyclophilin A).
  • the assay was run using the multi-cycle kinetics binding assay format with 8K Biacore instrument employing 10-points and 1 to 3 serial dilution compound plate (prepared with ECHO instrument).2.5 uM Cyclophilin A solution, which was made with 0% DMSO assay buffer, was added to the plate to DMSO final concentration at 2%. Compound final concentration was from 10 uM to 0.51 nM and the last point was blank control.
  • Avi-KRAS G12C 25 GMPPNP was immobilized to SA-chip surface to 100 RU level.
  • the assay was run at 22°C .
  • the flow rate was set to 30 ⁇ l/min, contact time 60 sec., dissociation time 600 sec. and 15 start- up cycles.7-points DMSO solvent correction was run before and after the sample running. All analysis of data were performed using Cytiva’s Biacore Insight Evaluation Software.
  • the predefined evaluation method “LMW multi-cycle kinetics” was selected for data analysis.1:1 30 binding kinetics fit model was applied, and steady state affinity fit mode was applied to weak compounds which could’t be analyzed with kinetics fit mode.
  • mice SPR activities Table 18: Biological assay results for panRAS inhibitors PAT059646-WO-PCT 259 PAT059646-WO-PCT 5
  • Table 19 Pharmacokinetic data for select compounds 15
  • Table 20 Structures of panRAS inhibitors Note: axial chirality of compounds is as shown in the structures in Table 20. 260 PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT

Landscapes

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

Abstract

The present invention relates to compounds of the formulae (I) or (XX) that are panRAS inhibitors for use in a method of treating cancer.

Description

PAT059646-WO-PCT 5 PANRAS INHIBITORS AND METHODS OF USE THEREOF TECHNICAL FIELD [01] The present disclosure relates to panRAS inhibitors. The disclosure further relates to methods and compositions useful in the treatment and/or diagnosis of cancers that express 10 RAS and/or are amenable to treatment by modulating panRAS expression and/or activity, as well as methods of making those compositions. BACKGROUND [02] Ras proteins (K-Ras, H-Ras and N-Ras) play an essential role in various human cancers 15 and are therefore appropriate targets for anticancer therapy. Indeed, mutations in Ras proteins account for approximately 30% of all human cancers in the United States, many of which are fatal. Dysregulation of Ras proteins by activating mutations, overexpression or upstream activation is common in human tumors, and activating mutations in Ras are frequently found in human cancer. For example, activating mutations at codon 12 in Ras proteins function by 20 inhibiting both GTPase-activating protein (GAP)-dependent and intrinsic hydrolysis rates of GTP, significantly skewing the population of Ras mutant proteins to the “on” (GTP-bound) state (Ras(ON)), leading to oncogenic MAPK signaling. Notably, Ras exhibits a picomolar affinity for GTP, enabling Ras to be activated even in the presence of low concentrations of this nucleotide. Mutations at codons 13 (e.g., G13D) and 61 (e.g., Q61K) of Ras are also responsible for 25 oncogenic activity in some cancers. [03] Despite extensive drug discovery efforts against Ras during the last several decades, additional efforts are needed to uncover additional medicines for cancers driven by the various Ras mutations. SUMMARY 30 [04] In one aspect, the present disclosure provides, 1 PAT059646-WO-PCT 5 A compound of formula (I*) or formula (XX*), wherein 10 ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 15 N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; 20 each R1 is independently selected from the group consisting of H, Halo, OH, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, NRxCOR6, and COR6; R2 is optionally substituted 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; R3 is selected from the group consisting of H, Halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 25 haloalkyl, NH2, and COR6, or R3 is absent; PAT059646-WO-PCT 5 each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a 10 C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl, aryl, or 15 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; or R7 is C3-6 cycloalkyl; wherein R7 is optionally further substituted; R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; each Rx is independently selected from H, C1-6 alkyl, C3-6 cycloalkyl, and C1-6 haloalkyl; 20 W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, and R9 is optionally substituted; m is 0 or 1; and n is 0, 1, 2, or 3. 25 [05] In another aspect, the present disclosure provides,
PAT059646-WO-PCT 5 A compound of formula (I*) or formula (XX*), wherein 10 ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 15 N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; 20 each R1 is independently selected from the group consisting of H, Halo, OH, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, NRxCOR6, and COR6; R2 is optionally substituted 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; R3 is selected from the group consisting of H, Halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 25 haloalkyl, NH2, and COR6, or R3 is absent; PAT059646-WO-PCT 5 each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a 10 C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl, aryl, or 15 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; or R7 is C3-6 cycloalkyl; wherein R7 is optionally further substituted; R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; each Rx is independently selected from H, C1-6 alkyl, C3-6 cycloalkyl, and C1-6 haloalkyl; 20 W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, and R9 is optionally substituted; m is 0 or 1; and n is 0, 1, 2, or 3, 25 wherein at least one of the following is true: a) R9 is oxetane or pyrrolidine, each optionally substituted; or b) R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; wherein R7 is optionally further substituted. 30 [06] In another aspect, the present disclosure provides,
PAT059646-WO-PCT 5 A compound of formula (I*) or formula (XX*), wherein 10 ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 15 N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; 20 each R1 is independently selected from the group consisting of H, Halo, OH, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, NRxCOR6, and COR6; R2 is optionally substituted 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; R3 is selected from the group consisting of H, Halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 25 haloalkyl, NH2, and COR6, or R3 is absent; PAT059646-WO-PCT 5 each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a 10 C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl, aryl, or 15 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; or R7 is C3-6 cycloalkyl; wherein R7 is optionally further substituted; R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; each Rx is independently selected from H, C1-6 alkyl, C3-6 cycloalkyl, and C1-6 haloalkyl; 20 W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, and R9 is optionally substituted; m is 0 or 1; and n is 0, 1, 2, or 3, 25 wherein at least one of the following is true: a) R9 is 2-oxetanyl or 2-azetidinyl, each optionally substituted; or b) R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; wherein R7 is optionally further substituted. . 30 [07] In another aspect, the disclosure provides a pharmaceutical composition comprising a compound of the disclosure and a pharmaceutically acceptable carrier. In yet another aspect, the disclosure provides a method of treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective 35 amount of a compound of the disclosure, or a pharmaceutical composition comprising a compound of the disclosure. In yet another aspect, the disclosure provides methods of synthesis of a compound of the disclosure. [08] The disclosure provides, in part, novel compounds with biological activity against cancer 40 cells. The compounds may slow, inhibit, and/or reverse tumor growth in mammals, and/or may be useful for treating human cancer patients. The present disclosure more specifically relates, PAT059646-WO-PCT 5 in some embodiments, to panRAS inhibitor compounds that are capable of binding and killing cancer cells. BRIEF DESCRIPTION OF THE DRAWINGS [09] Fig.1 is a depiction of the 3-dimensional structure of Compound 101 derived from an X- 10 ray diffraction crystal study. [10] Fig.2 is a depiction of the PDB coordinates for Compound 101 derived from an X-ray diffraction crystal study. DETAILED DESCRIPTION 15 [11] The disclosed compositions and methods may be understood more readily by reference to the following detailed description. Provided herein are compounds (e.g., compounds of formula (I) or other formulas disclosed herein), and pharmaceutically acceptable salts thereof, which are panRAS inhibitors. [12] In one aspect, the present disclosure provides. 20 A compound of formula (I*) or formula (XX*), or in some embodiments a compound of formula (I) or formula (XX), PAT059646-WO-PCT 5 wherein 10 ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 15 N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; 20 each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, NRxCOR6, and COR6; R2 is optionally substituted 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 25 haloalkyl, NH2, and COR6, or R3 is absent; PAT059646-WO-PCT 5 each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a 10 C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl, aryl, or 15 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; or R7 is C3-6 cycloalkyl; wherein R7 is optionally further substituted; R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; each Rx is independently selected from H, C1-6 alkyl, C3-6 cycloalkyl, and C1-6 haloalkyl; 20 W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, and R9 is optionally substituted; m is 0 or 1; and n is 0, 1, 2, or 3. 25 [13] In one aspect, the present disclosure provides. A compound of formula (I*) or formula (XX*), or in some embodiments a compound of formula (I) or formula (XX), PAT059646-WO-PCT 5 10 wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- 15 membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; 1 PAT059646-WO-PCT 5 ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C2-6 10 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, NRxCOR6, and COR6; R2 is optionally substituted 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6, or R3 is absent; 15 each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a 20 C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl, aryl, or 25 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; or R7 is C3-6 cycloalkyl; wherein R7 is optionally further substituted; R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; each Rx is independently selected from H, C1-6 alkyl, C3-6 cycloalkyl, and C1-6 haloalkyl; 30 W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, and R9 is optionally substituted; m is 0 or 1; and n is 0, 1, 2, or 3, 35 wherein at least one of the following is true: a) R9 is oxetane or pyrrolidine, each optionally substituted; or b) R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; wherein R7 is optionally further substituted. PAT059646-WO-PCT 5 [14] In another aspect, the present disclosure provides. A compound of formula (I*) or formula (XX*), or in some embodiments a compound of formula (I) or formula (XX), 10 PAT059646-WO-PCT 5 wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 10 N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- 15 membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C 3-6 cycloalkyl, C 1-6 heteroalkyl, C 1-6 haloalkyl, NH 2 , NRxCOR6, and COR6; 20 R2 is optionally substituted 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6, or R3 is absent; each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 25 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a C3-6 cycloalkyl; 30 each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; PAT059646-WO-PCT 5 R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl, aryl, or 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; or R7 is C3-6 cycloalkyl; wherein R7 is optionally further substituted; R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; 10 each Rx is independently selected from H, C1-6 alkyl, C3-6 cycloalkyl, and C1-6 haloalkyl; W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, and R9 is optionally substituted; m is 0 or 1; and 15 n is 0, 1, 2, or 3, wherein at least one of the following is true: a) R9 is 2-oxetanyl or 2-azetidinyl, each optionally substituted; or b) R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; wherein R7 is optionally further substituted. 20 [15] In another aspect, the present disclosure provides,
PAT059646-WO-PCT 5 A compound of formula (I*) or formula (XX*), wherein 10 ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 15 N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; 20 each R1 is independently selected from the group consisting of H, Halo, OH, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, NRxCOR6, and COR6; R2 is optionally substituted 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; R3 is selected from the group consisting of H, Halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 25 haloalkyl, NH2, and COR6, or R3 is absent; PAT059646-WO-PCT 5 each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a 10 C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl, aryl, or 15 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; or R7 is C3-6 cycloalkyl; wherein R7 is optionally further substituted; R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; each Rx is independently selected from H, C1-6 alkyl, C3-6 cycloalkyl, and C1-6 haloalkyl; 20 W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, and R9 is optionally substituted; m is 0 or 1; and n is 0, 1, 2, or 3, 25 wherein at least one of the following is true: a) R9 is 2-oxetanyl or 2-azetidinyl, each optionally substituted; or b) R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; wherein R7 is optionally further substituted. 30 [16] In some embodiments, the present disclosure provides, A compound of formula (I) PAT059646-WO-PCT 5 ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 10 N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; 15 each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6; R2 is substituted pyridine or substituted phenyl; R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6, or R3 is absent; 20 each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a 25 C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl, aryl, or 30 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; or R7 is C3-6 cycloalkyl; wherein R7 is optionally further substituted; R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, and R9 is optionally substituted; 35 m is 0 or 1; and n is 0, 1, 2, or 3, wherein at least one of the following is true: a) R9 is oxetane or pyrrolidine, each optionally substituted; or b) R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; 40 wherein R7 is optionally further substituted. PAT059646-WO-PCT 5 [17] In some embodiments, the present disclosure provides, A compound of formula (I) wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- 10 membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; 15 ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 20 heteroalkyl, C1-6 haloalkyl, NH2, and COR6; R2 is substituted pyridine or substituted phenyl; R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6, or R3 is absent; each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 25 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a C3-6 cycloalkyl; 30 each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl, aryl, or 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; or R7 is C3-6 cycloalkyl; PAT059646-WO-PCT 5 wherein R7 is optionally further substituted; R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, and R9 is optionally substituted; m is 0 or 1; and 10 n is 0, 1, 2, or 3, wherein at least one of the following is true: a) R9 is 2-oxetanyl or 2-azetidinyl, each optionally substituted; or b) R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; wherein R7 is optionally further substituted. 15 [18] In some embodiments, the present disclosure provides a compound of formula (I) 20 wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- 25 membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and 30 ring B is edge-fused to ring C to form a BC ring system; each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6; R2 is substituted pyridine or substituted phenyl; PAT059646-WO-PCT 5 R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6, or R3 is absent; each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 10 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; 15 L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl; or R7 is C3-6 cycloalkyl; wherein R7 is optionally further substituted; R8 is H or C1-6 alkyl; 20 W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S; and R9 is optionally substituted; m is 0 or 1; and n is 0, 1, 2, or 3, wherein at least one of the following is true: 25 a) m is 0; or b) R9 is optionally substituted oxetane. [19] In some embodiments, the present disclosure provides a compound of formula (I) 30 PAT059646-WO-PCT 5 ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 10 N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; 15 each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6; R2 is substituted pyridine or substituted phenyl; R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6, or R3 is absent; 20 each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a 25 C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl; or R7 is 30 C3-6 cycloalkyl; wherein R7 is optionally further substituted; R8 is H or C1-6 alkyl; W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S; and R9 is optionally substituted; 35 m is 0 or 1; and n is 0, 1, 2, or 3, wherein at least one of the following is true: a) m is 0; or b) R9 is optionally substituted 2-oxetanyl. 40 PAT059646-WO-PCT 5 [20] In some embodiments, the compound is a compound of formula (II) wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 10 N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- 15 membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6; 20 R2 is substituted pyridine or substituted phenyl; R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6, or R3 is absent; each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; 25 each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, 30 C1-6 heteroalkyl, and NH2; L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; R7 is C1-6 alkyl, C2-6 alkenyl, or C1-6 alkynyl, each optionally substituted by C3-6 cycloalkyl; or R7 is C3-6 cycloalkyl; wherein R7 is optionally further substituted; PAT059646-WO-PCT 5 R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, and R9 is optionally substituted; n is 0, 1, 2, or 3; and wherein at least one of the following is true: 10 a) R9 is oxetane or pyrrolidine, each optionally substituted; or b) R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; wherein R7 is optionally further substituted. In some embodiments, the compound is a compound of compound of formula (I) 15 wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; 20 ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 25 N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6; R2 is substituted pyridine or substituted phenyl; 30 R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6, or R3 is absent; each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; PAT059646-WO-PCT 5 each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, 10 C1-6 heteroalkyl, and NH2; L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl; or R7 is C3-6 cycloalkyl; wherein R7 is optionally further substituted; R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; 15 W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S; and R9 is optionally substituted; m is 0 or 1; and n is 0, 1, 2, or 3; 20 wherein at least one of the following is true: a) R9 is 2-oxetanyl or 2-azetidinyl, each optionally substituted; or b) R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; wherein R7 is optionally further substituted. 25 [21] In some embodiments, the compound is a compound of formula (II) wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 30 N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; PAT059646-WO-PCT 5 ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 10 heteroalkyl, C1-6 haloalkyl, NH2, and COR6; R2 is substituted pyridine or substituted phenyl; R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6, or R3 is absent; each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 15 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a C3-6 cycloalkyl; 20 each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; R7 is C1-6 alkyl, C2-6 alkenyl, or C1-6 alkynyl, each optionally substituted by C3-6 cycloalkyl; or R7 is C3-6 cycloalkyl; 25 wherein R7 is optionally further substituted; R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, and R9 is optionally substituted; n is 0, 1, 2, or 3; and 30 wherein at least one of the following is true: a) R9 is 2-oxetanyl or 2-azetidinyl, each optionally substituted; or b) R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; wherein R7 is optionally further substituted. 35 [22] In some embodiments, the compound is a compound of formula (II) PAT059646-WO-PCT 5 wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; 10 ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 15 N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6; R2 is substituted pyridine or substituted phenyl; 20 R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6, or R3 is absent; each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 25 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; 30 L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; R7 is C1-6 alkyl, C1-6 alkenyl, or C1-6 alkynyl, each optionally substituted by C3-6 cycloalkyl; or R7 is C3-6 cycloalkyl; wherein R7 is optionally further substituted; R8 is H or C1-6 alkyl; PAT059646-WO-PCT 5 W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, and R9 is optionally substituted; and n is 0, 1, 2, or 3. 10 [23] In some embodiments, the compound is a compound of formula (Iw) or formula (XXw) wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 15 N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- 20 membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6; 25 R2 is substituted pyridine or substituted phenyl; PAT059646-WO-PCT 5 R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6, or R3 is absent; each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 10 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; 15 R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S; and R9 is optionally substituted; m is 0 or 1; and n is 0, 1, 2, or 3. [24] In some embodiments, the compound is a compound of formula (Iw) 20 wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; 25 ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 30 N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6; R2 is substituted pyridine or substituted phenyl; PAT059646-WO-PCT 5 R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6, or R3 is absent; each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 10 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; 15 R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S; and R9 is optionally substituted; m is 0 and n is 0; or m is 0 and n is 1. 20 [25] In some embodiments, ring A is phenyl, pyridine, thiazole, triazole, oxazole, or oxadiazole, and ring A is substituted with 0-3 R10; wherein R10 is selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6. [26] In some embodiments, ring A is 25 [27] In some embodiments, ring A is [28] In some embodiments, R10 is not H. [29] In some embodiments, ring B is optionally substituted pyrrole. 30 [30] In some embodiments, ring C is optionally substituted phenyl. PAT059646-WO-PCT 5 [31] In some embodiments, the BC ring system is indole, benzothiophene, benzoxazole, or indolizine, each of which optionally has 1-3 additional N ring atoms in its 6-membered ring, wherein each BC ring system is optionally substituted, for example wherein the BC ring system is optionally substituted with 1-3 R11, wherein each R11 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 10 heteroalkyl, C1-6 haloalkyl, NH2, and COR6. In some embodiments, each R11 is independently selected from the group consisting of H and halo. In some embodiments there are no R11 substituents. [32] In some embodiments, the BC ring system is , wherein 15 X1 is N, S, or O; X2 is N or CR11; for example wherein either 0 or 1 of X2 are N, and the remaining X2 are CR11, for example wherein the BC ring system is , 20 wherein each R11 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6. In some embodiments, each R11 is independently selected from the group consisting of H and halo. In some embodiments there are no R11 substituents. In some embodiments, R11 is not H. [33] In some embodiments, each R1 is independently selected from the group consisting of 25 H, halo, C1-6 alkyl, or C1-6 haloalkyl; for example wherein each R1 is H. [34] In some embodiments, R2 is substituted pyridine. In some embodiments, R2 is substituted 3-pyridyl. [35] In some embodiments, R2 is 1 PAT059646-WO-PCT 5 , wherein R2a is C1-10 alkyl, C3-6 cycloalkyl, or C1-10 heteroalkyl, and R2a is optionally substituted by 1-3 substituents independently selected from the group consisting of halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, -(CH2)0-2 phenyl, -(CH2)0-2-C3-6 cycloalkyl, C1-6 haloalkyl, NH2, and COR6; for example, wherein R2a is C1-10 optionally substituted heteroalkyl; 10 R2b is 5-6-membered heterocycloalkyl, with 1-3 ring atoms selected from the group consisting of N, O, P(O)xx, and S(O)xx, wherein xx is 0, 1, or 2, and R2b is optionally substituted, for example with 1-3 R2ba, wherein each R2ba is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1- 6 heteroalkyl, -(CH2)0-2 phenyl, -(CH2)0-2-C3-6 cycloalkyl, C1-6 haloalkyl, NH2, and COR6; and 15 wherein each phenyl and cycloalkyl is optionally substituted, for example with 1-3 substituents selected from the group consisting of Halo, OH, CN, C1-6 alkyl, C1-6 haloalkyl, and C1-6 heteroalkyl; and each R2c is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, and C1-6 heteroalkyl; for example, wherein each R2c is H. 20 [36] In some embodiments, R2 is , wherein R2a is C1-10 alkyl, C3-6 cycloalkyl, or C1-10 heteroalkyl, and R2a is optionally substituted by 1-3 substituents independently selected from the group consisting of halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, -(CH2)0-2 phenyl, -(CH2)0-2-C3-6 cycloalkyl, C1-6 haloalkyl, NH2, NRxCOR6, and COR6; 25 for example, wherein R2a is C1-10 optionally substituted heteroalkyl; R2b is 4-10-membered heterocycloalkyl, with 1-4 ring atoms selected from the group consisting of N, O, P(O)xx, and S(O)xx, wherein xx is 0, 1, or 2, and R2b is optionally substituted, for example with 1-3 R2ba, wherein each R2ba is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-30 6 heteroalkyl, -(CH2)0-2 phenyl, -(CH2)0-2-C3-6 cycloalkyl, C1-6 haloalkyl, NH2, COR6, and -(CH2)0-2- heterocycloalkyl wherein the heterocycloalkyl is 4-7 membered with 1-3 ring atoms selected from the group consisting of N, O, P(O)xx, and S(O)xx, wherein xx is 0, 1, or 2, and wherein each phenyl, heterocycloalkyl and cycloalkyl is optionally substituted, for example with 1-3 substituents selected from the group consisting of halo, OH, CN, C1-6 alkyl, C1-6 haloalkyl, and 35 C1-6 heteroalkyl; PAT059646-WO-PCT 5 each R2c is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, and C1-6 heteroalkyl; for example, wherein each R2c is H; and each Rx is independently selected from H, C1-6 alkyl, C3-6 cycloalkyl, and C1-6 haloalkyl. [37] In some embodiments, R2b is 10 wherein X3 is C, CRx, N, or P(O); is a single or double bond; and each Rx is independently selected from H, C1-6 alkyl, C3-6 cycloalkyl, and C1-6 haloalkyl; for example, wherein R2ba is H, C1-6 alkyl, -(CH2)0-2 phenyl, C1-6 heteroalkyl, C1-6 haloalkyl, C3-6 cycloalkyl, COR6,or heterocycloalkyl wherein the heterocycloalkyl is 4-7 membered with 1-3 ring 15 atoms selected from the group consisting of N, O, and S, and each phenyl, cycloalkyl, or heterocycloalkyl is optionally substituted. [38] In some embodiments, R2b is wherein X3 is CH, N, or P(O); and 20 is a single or double bond; for example, wherein R2ba is H, C1-6 alkyl, -(CH2)0-2 phenyl, C1-6 heteroalkyl, C1-6 haloalkyl, C3-6 cycloalkyl, COR6., or heterocycloalkyl wherein the heterocycloalkyl is 4-7 membered with 1-3 ring atoms selected from the group consisting of N, O, and S. 25 [39] In some embodiments, R2b is PAT059646-WO-PCT 5 for example, wherein R2ba is H, C1-6 alkyl, -(CH2)0-2 phenyl, C1-6 heteroalkyl, C1-6 haloalkyl, C3-6 cycloalkyl, COR6., or heterocycloalkyl wherein the heterocycloalkyl is 4-7 membered with 1-3 ring atoms selected from the group consisting of N, O, and S. [40] In some embodiments, R2a is 10 , wherein X4 is O or N, and X5 is CH; or X5 is O or N, and X4 is CH; and R2aa is C1-3 alkyl or C3-6 cycloalkyl, each optionally substituted with 1-3 halo, or R2aa is absent or H; 15 R2ab is C1-3 alkyl or C3-6 cycloalkyl, each optionally substituted with 1-3 halo, or R2ab is absent or H; and R2ac is C1-3 alkyl or C3-6 cycloalkyl, each optionally substituted with 1-3 halo or one OH, or R2ac is absent or H. [41] In some embodiments, R2a is 20 , wherein X4 is O or N, and X5 is CH; or X5 is O or N, and X4 is CH; and R2aa is C1-3 alkyl optionally substituted with 1-3 halo, or R2aa is absent or H; R2ab is C1-3 alkyl optionally substituted with 1-3 halo, or R2ab is absent or H; and 25 R2ac is C1-3 alkyl optionally substituted with 1-3 halo or one OH. [42] In some embodiments, R2 is PAT059646-WO-PCT 5 X3 is C, CRx, N, or P(O); X4 is O or N, and X5 is CH; or X5 is O or N, and X4 is CH; and is a single or double bond R2aa is C1-3 alkyl or C3-6 cycloalkyl, each optionally substituted with 1-3 halo, or R2aa is absent or 10 H; R2ab is C1-3 alkyl or C3-6 cycloalkyl, each optionally substituted with 1-3 halo, or R2ab is absent or H; R2ac is C1-3 alkyl or C3-6 cycloalkyl, each optionally substituted with 1-3 halo or one OH; and R2ba is H, C1-6 alkyl, -(CH2)0-2 phenyl, C1-6 heteroalkyl, C1-6 haloalkyl, C3-6 cycloalkyl, COR6., or 15 heterocycloalkyl wherein the heterocycloalkyl is 4-7 membered with 1-3 ring atoms selected from the group consisting of N, O, and S, and each phenyl, cycloalkyl, or heterocyloalkyl is optionally substituted. [43] In some embodiments, R2 is 20 , wherein X3 is CH, N, or P(O); X4 is O or N, and X5 is CH; or X5 is O or N, and X4 is CH; and is a single or double bond 25 R2aa is C1-3 alkyl optionally substituted with 1-3 halo, or R2aa is absent or H; R2ab is C1-3 alkyl optionally substituted with 1-3 halo, or R2ab is absent or H; R2ac is C1-3 alkyl optionally substituted with 1-3 halo or one OH; and R2ba is H, C1-6 alkyl, -(CH2)0-2 phenyl, C1-6 heteroalkyl, C1-6 haloalkyl, or COR6. 30 [44] In some embodiments, R2 is PAT059646-WO-PCT 5 R2aa is C1-3 alkyl optionally substituted with 1-3 halo, or R2aa is absent; 10 R2ab is C1-3 alkyl optionally substituted with 1-3 halo, or R2aa is absent; R2ac is C1-3 alkyl optionally substituted with 1-3 halo; and R2ba is H, C1-6 alkyl, -(CH2)0-2 phenyl, C1-6 heteroalkyl, C1-6 haloalkyl, C3-6 cycloalkyl, COR6., or heterocycloalkyl wherein the heterocycloalkyl is 4-7 membered with 1-3 ring atoms selected from the group consisting of N, O, and S, and each phenyl, cycloalkyl, or heterocycloalkyl is 15 optionally substituted. [45] In some embodiments, R2a is [46] In some embodiments, R2ba is C1-6 alkyl. 20 [47] In some embodiments, R3 is selected from the group consisting of H, C1-6 alkyl, C1-6 heteroalkyl, and C1-6 haloalkyl, or R3 is absent; for example, R3 is C1-6 alkyl. In some embodiments, R3 is methyl or ethyl. In some embodiments, R3 is ethyl. [48] In some embodiments, each R4 is independently selected from the group consisting of H, C1-6 alkyl, and C1-6 haloalkyl; for example, each R4 is H. 25 [49] In some embodiments, each R5 is independently selected from H and C1-6 alkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a C3-6 cycloalkyl. In some embodiments, each R5 is independently selected from H and C1-6 alkyl; for example both R5 are CH3. [50] In some embodiments, L is absent, or L is -COCHR7N(R8)-*, wherein * indicates the 30 point of attachment to W; R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl; or R7 is C3-6 cycloalkyl; PAT059646-WO-PCT 5 wherein R7 is optionally further substituted by 1-3 substituents selected from the group consisting of halo, OH, C1-3 haloalkyl, C1-3 heteroalkyl, and C1-3 alkyl; and R8 is H or C1-3 alkyl. [51] In some embodiments, R7 is C1-6 alkyl, C1-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, or C1-6 alkyl substituted by C3-6 cycloalkyl, wherein R7 is optionally further substituted by 1-3 10 substituents selected from the group consisting of halo and C1-3 alkyl. In some embodiments, R7 is C1-6 alkyl, C2-6 alkenyl, or C3-6 cycloalkyl, wherein R7 is optionally further substituted by 1-3 substituents selected from the group consisting of halo and C1-3 alkyl. [52] In some embodiments, R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; wherein R7 is optionally further substituted by 1-3 substituents 15 selected from the group consisting of halo and C1-3 alkyl. [53] In some embodiments, R7 is 20 [54] In some embodiments, R7 is 25 [55] In some embodiments, L is PAT059646-WO-PCT 5 r absent; for example, wherein R8 is CH3. In some embodiments, L is 10 In some embodiments, L is [57] In some embodiments, 15 W is -COR9; R9 is C3-7 cycloalkyl or 4-6 membered heterocycloalkyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, wherein each R9 is optionally substituted by 1-3 R9a, and optionally substituted with 1 R9b on a ring nitrogen atom, if present; each R9a is independently selected from the group consisting of halo, OH, CN, C1-6 alkyl, C1-6 20 heteroalkyl, -(CH2)0-2 phenyl, -(CH2)0-2-C3-6 cycloalkyl, C1-6 haloalkyl, alkenyl, C1-6 alkynyl, NH2, PAT059646-WO-PCT 5 COR6, NRxCOR6, and -(CH2)0-2 heterocyclyl of 4-7 ring atoms with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and wherein each phenyl, heterocyclyl and cycloalkyl is optionally substituted, for example with 1-3 substituents selected from the group consisting of halo, OH, CN, C1-6 alkyl, C1-6 haloalkyl, and C1-6 heteroalkyl. [58] In some embodiments, 10 W is -COR9; R9 is C3-7 cycloalkyl or 4-6 membered heterocycloalkyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, wherein each R9 is optionally substituted by 1-3 R9a; each R9a is independently selected from the group consisting of halo, OH, CN, C1-6 alkyl, C1-6 15 heteroalkyl, -(CH2)0-2 phenyl, -(CH2)0-2-C3-6 cycloalkyl, C1-6 haloalkyl, NH2, COR6, and -(CH2)0-2 heterocyclyl of 4-7 ring atoms with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and wherein each phenyl, heterocyclyl and cycloalkyl is optionally substituted, for example with 1-3 substituents selected from the group consisting of halo, OH, CN, C1-6 alkyl, C1-6 haloalkyl, and C1-6 heteroalkyl. 20 In some embodiments, R9 is oxetane or pyrrolidine, each optionally substituted. In some embodiments, R9 is optionally substituted oxetane. In some embodiments, R9 is oxetane optionally substituted with 1-3 R9a. In some embodiments, R9 is 2-oxetanyl optionally substituted with 1-3 R9a. In some embodiments R9a is optionally substituted phenyl. 25 [59] In some embodiments, W is PAT059646-WO-PCT 5 , wherein R9b is H, C1-6 alkyl, C1-6 heteroalkyl, -(CH2)0-2 phenyl, -(CH2)0-2-C3-6 cycloalkyl, C1-6 haloalkyl, NH2, COR6, NRxCOR6, and -(CH2)0-2 heterocyclyl of 4-7 ring atoms with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and wherein each 10 phenyl, heterocyclyl and cycloalkyl is optionally substituted, for example with 1-3 substituents selected from the group consisting of halo, OH, CN, C1-6 alkyl, C1-6 haloalkyl, and C1-6 heteroalkyl, for example wherein R9b is H, C1-3 alkyl, or NRxCO-C1-3 alkyl. [60] In some embodiments, W is 15 wherein R9b is H or C1-3 alkyl. [61] In some embodiments, W is PAT059646-WO-PCT 5 wherein R9b is H or C1-3 alkyl; for example wherein R9a is optionally substituted phenyl. In some embodiments, 10 each R9a’ is independently selected from the group consisting of halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, -(CH2)0-2 phenyl, -(CH2)0-2-C3-6 cycloalkyl, C1-6 haloalkyl, NH2, COR6, and -(CH2)0-2 heterocyclyl of 4-7 ring atoms with 1-3 ring atoms independently selected from the group 15 consisting of N, O, and S, and wherein each phenyl, heterocyclyl and cycloalkyl is optionally substituted, for example with 1-3 substituents selected from the group consisting of halo, OH, CN, C1-6 alkyl, C1-6 haloalkyl, and C1-6 heteroalkyl. In some embodiments R9a is optionally substituted phenyl. In some embodiments, W is 20 PAT059646-WO-PCT 5 In some embodiments, W is . 10 In some embodiments, W is , 15 In some embodiments, W is PAT059646-WO-PCT 5 [62] In some embodiments, the compound is a compound of formula (III), formula (XXIII), or formula (XXXIII) 10 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H, for example wherein n is 0 or 1; for example a compound of formula (IIIa) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H. [63] In some embodiments, the compound is a compound of formula (IV), formula (XXIV), or formula (XXXIV) 10 PAT059646-WO-PCT 5 wherein n is 0 or 1; wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H; for example, wherein there are no R11 substituents; 10 for example, wherein the compound is a compound of formula (IVa) wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H; for example, wherein there are no R11 substituents. 15 [64] In some embodiments, the compound is a compound of formula (V), formula (VI), formula (XXV), formula (XXVI), formula (XXXV), or formula (XXXVI)
PAT059646-WO-PCT 5 10 wherein n is 0 or 1; wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H; for example, wherein all R5 are CH3; for example, wherein there are no R11 substituents; 15 for example, a compound of formula (V), formula (XXV), or formula (XXXV) wherein there is one R10 selected from the group consisting of OH, NH2, and halo, and the remaining R10 are H; for example, a compound of formula (VI), formula (XXVI), or formula (XXXVI) wherein R10 is H. [65] In some embodiments, the compound is a compound of formula (Vw) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure, for example, wherein m is 0, and n is 0 or 1; for example, wherein all R1 are H; for example, wherein all R5 are CH3; for example, wherein there are no R11 substituents; 10 for example, a compound of formula (Vw) wherein there is one R10 selected from the group consisting of OH, NH2, and halo, and the remaining R10 are H. [66] In some embodiments, the compound is a compound of formula (Va) or formula (VIa) 15 are defined as in various embodiments of the disclosure; for example, wherein all R1 are H; for example, wherein all R5 are CH3; for example, wherein there are no R11 substituents; for example, a compound of formula (Va) wherein there is one R10 selected from the group consisting of OH, NH2, and halo, and the remaining R10 are H; 20 for example, a compound of formula (VIa) wherein R10 is H. [67] In some embodiments, the compound is a compound of formula (VII), formula (XXVII), or formula (XXXVII) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure; 10 for example, wherein all R1 are H. [68] In some embodiments, the compound is a compound of formula (Iy’) or formula (XXy’) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H, for example wherein m is 1 and n is 0 or 1. 10 [69] In some embodiments, the compound is a compound of formula (Iy) or formula (XXy) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H, for example wherein m is 1 and n is 0 or 1. [70] In some embodiments, the compound is a compound of formula (Iyz) 10 wherein the variables are defined as in various embodiments of the disclosure, and R9a is not H; for example, wherein all R1 are H, for example wherein m is 1 and n is 0 or 1. [71] In some embodiments, the compound is a compound of formula (IIIy’), formula (XXIIIy’), 15 or formula (XXXIIIy’) PAT059646-WO-PCT 5 10 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H, for example wherein n is 0 or 1; for example a compound of formula (IIIya’) 1 PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H. [72] In some embodiments, the compound is a compound of formula (IIIy’), formula (XXIIIy’), 10 or formula (XXXIIIy’) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H, for example wherein n is 0 or 1; 10 for example a compound of formula (IIIya’) wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H. 15 [73] In some embodiments, the compound is a compound of formula (IIIy), formula (XXIIIy), or formula (XXXIIIy) PAT059646-WO-PCT 5 ); 10 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H, for example wherein n is 0 or 1; for example a compound of formula (IIIya) 15 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H. [74] In some embodiments, the compound is a compound of formula (IIIyz), formula (XXIIIyz), or formula (XXXIIIyz) PAT059646-WO-PCT 5 10 wherein the variables are defined as in various embodiments of the disclosure, and R9a is not H; for example, wherein all R1 are H, for example wherein n is 0 or 1; for example a compound of formula (IIIyza) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure, and R9a is not H; for example, wherein all R1 are H. [75] In some embodiments, the compound is a compound of formula (IVy’), formula (XXIVy’), 10 or formula (XXXIVy’) 15 PAT059646-WO-PCT 5 wherein n is 0 or 1; wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H; for example, wherein there are no R11 substituents; for example, wherein the compound is a compound of formula (IVya’) 10 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H; for example, wherein there are no R11 substituents. [76] In some embodiments, the compound is a compound of formula (IVy), formula (XXIVy), or formula (XXXIVy) 15 PAT059646-WO-PCT 5 wherein n is 0 or 1; wherein the variables are defined as in various embodiments of the 10 disclosure; for example, wherein all R1 are H; for example, wherein there are no R11 substituents; for example, wherein the compound is a compound of formula (IVya) 15 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H; for example, wherein there are no R11 substituents. [77] In some embodiments, the compound is a compound of formula (IVyz), formula (XXIVyz), or formula (XXXIVyz) PAT059646-WO-PCT 5 10 wherein n is 0 or 1; wherein the variables are defined as in various embodiments of the disclosure, and R9a is not H; for example, wherein all R1 are H; for example, wherein there are no R11 substituents; for example, wherein the compound is a compound of formula (IVyza) PAT059646-WO-PCT 5 (IVyza); wherein the variables are defined as in various embodiments of the disclosure, and R9a is not H; for example, wherein all R1 are H; for example, wherein there are no R11 substituents. [78] In some embodiments, the compound is a compound of formula (Vy’) or formula (VIy’), formula (XXVy’), formula (XXVIy’), formula (XXXVIy’) or formula (XXXVIy’) 10
PAT059646-WO-PCT 5 10 wherein n is 0 or 1; wherein the variables are defined as in various embodiments of the disclosure; 1 PAT059646-WO-PCT 5 for example, wherein all R1 are H; for example, wherein all R5 are CH3; for example, wherein there are no R11 substituents; for example, a compound of formula (Vy’), formula (XXVy’), or formula (XXXVy’) wherein there is one R10 selected from the group consisting of OH, NH2, and halo, and the remaining R10 are H; 10 for example, a compound of formula (VIy’), formula (XXVIy’), or formula (XXXVIy’) wherein R10 is H. In some embodiments, the compound is a compound of formula (Vya’) or formula (Viya’) 15 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H; for example, wherein all R5 are CH3; for example, wherein 20 there are no R11 substituents; for example, a compound of formula (Vya’) wherein there is one R10 selected from the group consisting of OH, NH2, and halo, and the remaining R10 are H; for example, a compound of formula (Viya’) wherein R10 is H. [79] In some embodiments, the compound is a compound of formula (Vy) or formula (VIy), 25 formula (XXVy), formula (XXVIy), formula (XXXVIy) or formula (XXXVIy)
PAT059646-WO-PCT 5 wherein n is 0 or 1; wherein the variables are defined as in various embodiments of the 10 disclosure; for example, wherein all R1 are H; for example, wherein all R5 are CH3; for example, wherein there are no R11 substituents; for example, a compound of formula (Vy), formula (XXVy), or formula (XXXVy) wherein there is one R10 selected from the group consisting of OH, NH2, and halo, and the remaining R10 are H; 15 for example, a compound of formula (VIy), formula (XXVIy), or formula (XXXVIy) wherein R10 is H. In some embodiments, the compound is a compound of formula (Vya) or formula (VIya)
PAT059646-WO-PCT 5 (VIya); wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H; for example, wherein all R5 are CH3; for example, wherein 10 there are no R11 substituents; for example, a compound of formula (Vya) wherein there is one R10 selected from the group consisting of OH, NH2, and halo, and the remaining R10 are H; for example, a compound of formula (VIya) wherein R10 is H. [80] In some embodiments, the compound is a compound of formula (Vyz) or formula (VIyz), 15 formula (XXVyz), formula (XXVIyz), formula (XXXVyz), or formula (XXXVIyz) PAT059646-WO-PCT 5 10 wherein n is 0 or 1; wherein the variables are defined as in various embodiments of the disclosure, and R9a is not H; for example, wherein all R1 are H; for example, wherein all R5 are CH3; for example, wherein there are no R11 substituents; PAT059646-WO-PCT 5 for example, wherein R9a is optionally subsituted phenyl; for example, a compound of formula (Vyz), formula (XXVyz), or formula (XXXVyz) wherein there is one R10 selected from the group consisting of OH, NH2, and halo, and the remaining R10 are H; for example, a compound of formula (VIyz), formula (XXVIyz), or formula (XXXVIyz) wherein 10 R10 is H. [81] In some embodiments, the compound is a compound of formula (Vyz*) 15 for example, wherein m is 0 and n is 0 or 1; wherein the variables are defined as in various embodiments of the disclosure, and R9a is not H; for example, wherein all R1 are H; for example, wherein all R5 are CH3; for example, wherein there are no R11 substituents; for example, wherein R9a is optionally subsituted phenyl; 20 for example, wherein there is one R10 selected from the group consisting of OH, NH2, and halo, and the remaining R10 are H. [82] In some embodiments, the compound is a compound of formula (Vyza) or formula (VIyza) PAT059646-WO-PCT 5 embodiments of the disclosure, and R9a is not H; for example, wherein all R1 are H; for example, wherein all R5 are CH3; for example, wherein there are no R11 substituents; for example, wherein R9a is optionally subsituted phenyl; 10 for example, a compound of formula (Vyza) wherein there is one R10 selected from the group consisting of OH, NH2, and halo, and the remaining R10 are H; for example, a compound of formula (VIyza) wherein R10 is H. [83] In some embodiments, the compound is a compound of formula (VIIy), formula (VIIIy), 15 formula (XXVIIy), formula (XXVIIIy), formula (XXXVIIy), or formula (XXXVIIIy)
PAT059646-WO-PCT 5 10 PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H. 10 [84] In some embodiments, the compound is a compound of formula (VIIyz) (VIIyz); wherein the variables are defined as in various embodiments of the disclosure, and R9a is not H; for example, wherein R9a is optionally subsituted phenyl; for example, wherein all R1 are H. 15 [85] In some embodiments, the compound is a compound of formula (Iw) or formula (XXw) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure. [86] In some embodiments, the compound is a compound of formula (Iw*) or formula (XXw*) 10 1 PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure. [87] In some embodiments, L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W; 10 R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; wherein R7 is optionally further substituted; and R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; for example wherein R7 is 15 for example wherein R7 is for example wherein R7 is . [88] In some embodiments, the compound is selected from the compounds disclosed in the 20 specification. In some embodiments, the compound is selected from the compounds in Table 20. In some embodiments, the compound is selected from the group consisting of the following compound numbers: 103, 106, 107, 109, 112, 116, 117, 122, 123, 124, 125, 126, 130, 132, 133, 134, 135, 136, 137, 138, 139, 140, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 157, 158, 159, 160, 161, and 162. In some embodiments, the compound is selected from the group 25 consisting of the following compound numbers: 103, 106, 107, 109, 112, 116, 117, 122, 123, PAT059646-WO-PCT 5 130, 132, 133, 134, 135, 136, 137, 138, 139, 140, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 157, 158, 159, 160, 161, and 162. In some embodiments, the compound is selected from the group consisting of the following compound numbers: 112, 116, 122, 123, 132, 133, 134, 135, 136, 137, 138, 139, 140, 147, 148, 149, 150, 151, 152, 153, 154, 155, 157, 159, 160, 161, and 162. 10 [89] In some embodiments, any of the compounds disclosed herein may be in the form of a pharmaceutically acceptable salt. [90] In another aspect, disclosed is a pharmaceutical composition comprising a compound of the disclosure and a pharmaceutically acceptable carrier. [91] In another aspect, disclosed is a method of treating cancer in a subject in need thereof, 15 the method comprising administering to the subject a therapeutically effective amount of a compound of the disclosure, or a pharmaceutical composition of the disclosure. [92] In some embodiments, the cancer is a tumor or a hematological cancer, optionally, the cancer is a breast cancer including ER positive breast cancer, multiple myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, 20 bladder cancer, brain cancer, bone marrow cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal cancer, hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant prostate cancer, bladder urothelial carcinoma, melanoma, myelogenous leukemia, myeloma, oral cancer, ovarian cancer, 25 non-small cell lung cancer, prostate cancer, small cell lung cancer, spleen cancer, or head and neck cancer. [93] In some embodiments, the cancer is non-small cell lung cancer, pancreatic cancer, colorectal cancer, melanoma, head and neck cancer, acute myeloid leukemia, and bladder cancer. 30 [94] Throughout this text, the descriptions refer to compositions and methods of using the compositions. Where the disclosure describes or claims a feature or embodiment associated with a composition, such a feature or embodiment is equally applicable to the methods of using the composition. Likewise, where the disclosure describes or claims a feature or embodiment associated with a method of using a composition, such a feature or embodiment is equally 35 applicable to the composition. [95] When a range of values is expressed, it includes embodiments using any particular value within the range. Further, reference to values stated in ranges includes each and every value within that range. All ranges are inclusive of their endpoints and combinable. When values are expressed as approximations, by use of the antecedent “about,” it will be understood 40 that the particular value forms another embodiment. Reference to a particular numerical value includes at least that particular value, unless the context clearly dictates otherwise. The use of PAT059646-WO-PCT 5 “or” will mean “and/or” unless the specific context of its use dictates otherwise. All references cited herein are incorporated by reference for any purpose. Where a reference and the specification conflict, the specification will control. [96] It is to be appreciated that certain features of the disclosed compositions and methods, which are, for clarity, described herein in the context of separate embodiments, may also be 10 provided in combination in a single embodiment. Conversely, various features of the disclosed compositions and methods that are, for brevity, described in the context of a single embodiment, may also be provided separately or in any sub-combination. [97] Any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds. lsotopically labeled compounds have structures 15 depicted by the formulae given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Isotopes that can be incorporated into compounds disclosed herein include, for example, isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine, and chlorine, such as 3H, 11C, 13C, 14C, 15N, 18F, and 36Cl. Accordingly, it should be understood that the present disclosure includes compounds that incorporate one or more of 20 any of the aforementioned isotopes, including for example, radioactive isotopes, such as 3H and 14C, or those into which non-radioactive isotopes, such as 2H and 13C are present. Such isotopically labelled compounds are useful in metabolic studies (with 14C), reaction kinetic studies (with, for example 2H or 3H), detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or 25 substrate tissue distribution assays, or in radioactive treatment of patients. In particular, an 18F or labeled compound may be particularly desirable for PET or SPECT studies. Isotopically- labeled compounds can generally be prepared by conventional techniques known to those skilled in the art, e.g., using an appropriate isotopically-labeled reagents in place of the non- labeled reagent previously employed. 30 Definitions [98] Various terms relating to aspects of the description are used throughout the specification and claims. Such terms are to be given their ordinary meaning in the art unless otherwise indicated. Other specifically defined terms are to be construed in a manner consistent with the definitions provided herein. 35 [99] As used herein, the singular forms “a,” “an,” and “the” include plural forms unless the context clearly dictates otherwise. The terms “comprising”, “having”, “being of” as in “being of a chemical formula”, “including”, and “containing” are to be construed as open terms (i.e., meaning “including but not limited to”) unless otherwise noted. Additionally, whenever “comprising” or another open-ended term is used in an embodiment, it is to be understood that PAT059646-WO-PCT 5 the same embodiment can be more narrowly claimed using the intermediate term “consisting essentially of” or the closed term “consisting of”. [100] The term "about" or "approximately," when used in the context of numerical values and ranges, refers to values or ranges that approximate or are close to the recited values or ranges such that the embodiment may perform as intended, as is apparent to the skilled person from 10 the teachings contained herein. In some embodiments, about means plus or minus 20%, 15%, 10%, 5%, 1%, 0.5%, or 0.1% of a numerical amount. In one embodiment, the term “about” refers to a range of values which are 10% more or less than the specified value. In another embodiment, the term “about” refers to a range of values which are 5% more or less than the specified value. In another embodiment, the term “about” refers to a range of values which are 15 1% more or less than the specified value. [101] The term “agent” is used herein to refer to a chemical compound, a mixture of chemical compounds, a biological macromolecule, an extract made from biological materials, or a combination of two or more thereof. The term “therapeutic agent” or “drug” refers to an agent that is capable of modulating a biological process and/or has biological activity. The panRAS 20 inhibitors, as described herein, are exemplary therapeutic agents. [102] The term "chemotherapeutic agent" or “anti-cancer agent” is used herein to refer to all agents that are effective in treating cancer (regardless of mechanism of action). Inhibition of metastasis or angiogenesis is frequently a property of a chemotherapeutic agent. Chemotherapeutic agents include antibodies, biological molecules, and small molecules, and 25 encompass the panRAS inhibitors, as described herein. A chemotherapeutic agent may be a cytotoxic or cytostatic agent. The term “cytostatic agent” refers to an agent that inhibits or suppresses cell growth and/or multiplication of cells. The term "cytotoxic agent" refers to a substance that causes cell death primarily by interfering with a cell’s expression activity and/or functioning. 30 [103] The term “Rat Sarcoma Virus (Ras)” or “panRAS,” as used herein, refers to any native form of the human Ras protein family (e.g., K-Ras (including splice variants KRAS4A and KRAS4B), H-Ras and N-Ras). The term encompasses full-length human K-Ras (Kristen Rat Sarcoma Virus), N-Ras (Neuroblastoma Rat Sarcoma Virus) as well as any form of human Ras that may result from cellular processing. The term also encompasses functional variants or 35 fragments of human Ras proteins, including but not limited to splice variants, allelic variants, and isoforms that retain one or more biologic functions of human Ras proteins (i.e., variants and fragments are encompassed unless the context indicates that the term is used to refer to the wild-type protein only). Ras proteins can be isolated from human, or may be produced recombinantly or by synthetic methods. 40 [104] The term "inhibit" or "inhibition" or “inhibiting,” as used herein, means to reduce a biological activity or process by a measurable amount, and can include but does not require PAT059646-WO-PCT 5 complete prevention or inhibition. In some embodiments, “inhibition” means to reduce the expression and/or activity of panRAS and/or one or more upstream modulators or downstream targets thereof. [105] The term “panRAS inhibitor,” as used herein, refers to an agent capable of reducing the expression and/or activity of panRAS (e.g., K-Ras (including splice variants KRAS4A and 10 KRAS4B), H-Ras and N-Ras) and/or one or more upstream modulators or downstream targets thereof. Other exemplary panRAS modulators (including exemplary inhibitors of panRAS) are described in WO2021/091956 or WO2022/060836. [106] The term “cancer,” as used herein, refers to the presence of cells possessing characteristics typical of cancer-causing cells, such as uncontrolled proliferation, immortality, 15 metastatic potential, rapid growth and proliferation rate, and/or certain morphological features. Often, cancer cells can be in the form of a tumor or mass, but such cells may exist alone within a subject, or may circulate in the blood stream as independent cells, such as leukemic or lymphoma cells. The term "cancer" includes all types of cancers and cancer metastases, including hematological cancers, solid tumors, sarcomas, carcinomas and other solid and non- 20 solid tumor cancers. Hematological cancers may include B-cell malignancies, cancers of the blood (leukemias), cancers of plasma cells (myelomas, e.g., multiple myeloma), or cancers of the lymph nodes (lymphomas). Exemplary B-cell malignancies include chronic lymphocytic leukemia (CLL), follicular lymphoma, mantle cell lymphoma, and diffuse large B-cell lymphoma. Leukemias may include acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), 25 chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myelomonocytic leukemia (CMML), acute monocytic leukemia (AMoL), etc. The terms “acute lymphoblastic leukemia” and “acute lymphocytic leukemia” can be used interchangeably to describe ALL. Lymphomas may include Hodgkin's lymphoma, non-Hodgkin's lymphoma, etc. Other hematologic cancers may include myelodysplasia syndrome (MDS). Solid tumors may 30 include carcinomas such as adenocarcinoma, e.g., a breast cancer including ER positive breast cancer, multiple myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, bladder cancer, brain cancer, bone marrow cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal cancer, hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, 35 follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant prostate cancer, bladder urothelial carcinoma, melanoma, myelogenous leukemia, myeloma, oral cancer, ovarian cancer, non-small cell lung cancer, prostate cancer, small cell lung cancer, spleen cancer, or head and neck cancer. [107] As used herein, the term “tumor” refers to any mass of tissue that results from excessive 40 cell growth or proliferation, either benign or malignant, including precancerous lesions. In some embodiments, the tumor is a breast cancer including ER positive breast cancer, multiple PAT059646-WO-PCT 5 myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, bladder cancer, brain cancer, bone marrow cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal cancer, hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant 10 prostate cancer, bladder urothelial carcinoma, melanoma, myelogenous leukemia, myeloma, oral cancer, ovarian cancer, non-small cell lung cancer, prostate cancer, small cell lung cancer, spleen cancer, or head and neck cancer. [108] The terms “tumor cell” and “cancer cell” may be used interchangeably herein and refer to individual cells or the total population of cells derived from a tumor or cancer, including both 15 non-tumorigenic cells and cancer stem cells. The terms “tumor cell” and “cancer cell” will be modified by the term “non-tumorigenic” when referring solely to those cells lacking the capacity to renew and differentiate to distinguish those cells from cancer stem cells. [109] The terms “subject” and “patient” are used interchangeably herein to refer to any human or non-human animal in need of treatment. Non-human animals include all vertebrates (e.g., 20 mammals and non-mammals) such as any mammal. Non-limiting examples of mammals include humans, chimpanzees, apes, monkeys, cattle, horses, sheep, goats, swine, rabbits, dogs, cats, rats, mice, and guinea pigs. Non-limiting examples of non-mammals include birds and fish. In some embodiments, the subject is a human. [110] The term “a subject in need of treatment,” as used herein, refers to a subject that would 25 benefit biologically, medically, or in quality of life from a treatment (e.g., a treatment with any one or more of the exemplary compounds described herein). [111] As used herein, the term “treat,” “treating,” or “treatment” refers to any improvement of any consequence of disease, disorder, or condition, such as prolonged survival, less morbidity, and/or a lessening of side effects which result from an alternative therapeutic modality. In some 30 embodiments, treatment comprises delaying or ameliorating a disease, disorder, or condition (i.e., slowing or arresting or reducing the development of a disease or at least one of the clinical symptoms thereof). In some embodiments, treatment comprises delaying, alleviating, or ameliorating at least one physical parameter of a disease, disorder, or condition, including those which may not be discernible by the patient. In some embodiments, treatment comprises 35 modulating a disease, disorder, or condition, either physically (e.g., stabilization of a discernible symptom), physiologically (e.g., stabilization of a physical parameter), or both. In some embodiments, treatment comprises administration of a described compound or composition to a subject, e.g., a patient, to obtain a treatment benefit enumerated herein. The treatment can be to cure, heal, alleviate, delay, prevent, relieve, alter, remedy, ameliorate, palliate, improve, or 40 affect a disease, disorder, or condition (e.g., a cancer), the symptoms of a disease, disorder, or condition (e.g., a cancer), or a predisposition toward a disease, disorder, or condition (e.g., a PAT059646-WO-PCT 5 cancer). In some embodiments, in addition to treating a subject having a disease, disorder, or condition, a composition disclosed herein can also be provided prophylactically to prevent or reduce the likelihood of developing that disease, disorder, or condition. [112] As used herein, the term “prevent”, “preventing," or “prevention” of a disease, disorder, or condition refers to the prophylactic treatment of the disease, disorder, or condition; or 10 delaying the onset or progression of the disease, disorder, or condition. [113] As used herein, a "pharmaceutical composition" refers to a preparation of a composition, e.g., a panRAS inhibitor compound or composition, in addition to at least one other (and optionally more than one other) component suitable for administration to a subject, such as a pharmaceutically acceptable carrier, stabilizer, diluent, dispersing agent, suspending agent, 15 thickening agent, and/or excipient. The pharmaceutical compositions provided herein are in such form as to permit administration and subsequently provide the intended biological activity of the active ingredient(s) and/or to achieve a therapeutic effect. The pharmaceutical compositions provided herein preferably contain no additional components which are unacceptably toxic to a subject to which the formulation would be administered. 20 [114] As used herein, the terms "pharmaceutically acceptable carrier" and "physiologically acceptable carrier," which may be used interchangeably, refer to a carrier or a diluent that does not cause significant irritation to a subject and does not abrogate the biological activity and properties of the administered compound or composition and/or any additional therapeutic agent in the composition. Pharmaceutically acceptable carriers may enhance or stabilize the 25 composition or can be used to facilitate preparation of the composition. Pharmaceutically acceptable carriers can include solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drug stabilizers, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, and the like and combinations thereof, as 30 would be known to those skilled in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, pp.1289- 1329). Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the therapeutic or pharmaceutical compositions is contemplated. The carrier may be selected to minimize adverse side effects in the subject, and/or to minimize degradation of the active ingredient(s). 35 An adjuvant may also be included in any of these formulations. [115] As used herein, the term "excipient" refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient. Formulations for parenteral administration can, for example, contain excipients such as sterile water or saline, polyalkylene glycols such as polyethylene glycol, vegetable oils, or 40 hydrogenated napthalenes. Other exemplary excipients include, but are not limited to, calcium bicarbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, PAT059646-WO-PCT 5 gelatin, ethylene-vinyl acetate co-polymer particles, and surfactants, including, for example, polysorbate 20. [116] The term “pharmaceutically acceptable salt,” as used herein, refers to a salt which does not abrogate the biological activity and properties of the compounds disclosed herein, and does not cause significant irritation to a subject to which it is administered. Examples of such salts 10 include, but are not limited to: (a) acid addition salts formed with inorganic acids, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid and the like; and salts formed with organic acids, for example, acetic acid, oxalic acid, tartaric acid, succinic acid, maleic acid, fumaric acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, tannic acid, palmitic acid, alginic acid, polyglutamic acid, naphthalenesulfonic acid, 15 methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfonic acid, polygalacturonic acid, and the like; and (b) salts formed from elemental anions such as chlorine, bromine, and iodine. See, e.g., Haynes et al., “Commentary: Occurrence of Pharmaceutically Acceptable Anions and Cations in the Cambridge Structural Database,” J. Pharmaceutical Sciences, vol.94, no.10 (2005), and Berge et al., “Pharmaceutical Salts,” J. Pharmaceutical Sciences, vol.66, no.1 20 (1977), which are incorporated by reference herein. [117] In some embodiments, depending on their electronic charge, the compounds described herein can contain a monovalent anionic counterion M1-. Any suitable anionic counterion can be used. In certain embodiments, the monovalent anionic counterion is a pharmaceutically acceptable monovalent anionic counterion. In certain embodiments, the monovalent anionic 25 counterion M1- can be selected from bromide, chloride, iodide, acetate, trifluoroacetate, benzoate, mesylate, tosylate, triflate, formate, or the like. In some embodiments, the monovalent anionic counterion M1- is trifluoroacetate or formate. [118] As used herein, the term “therapeutically effective amount” or “therapeutically effective dose,” refers to an amount of a compound described herein, e.g., a compound or composition 30 described herein, to effect the desired therapeutic result (i.e., reduction or inhibition of an enzyme or a protein activity, amelioration of symptoms, alleviation of symptoms or conditions, delay of disease progression, a reduction in tumor size, inhibition of tumor growth, prevention of metastasis). In some embodiments, a therapeutically effective amount does not induce or cause undesirable side effects. In some embodiments, a therapeutically effective amount 35 induces or causes side effects but only those that are acceptable by a treating clinician in view of a patient’s condition. In some embodiments, a therapeutically effective amount is effective for detectable killing, reduction, and/or inhibition of the growth or spread of cancer cells, the size or number of tumors, and/or other measure of the level, stage, progression and/or severity of a cancer. The term also applies to a dose that will induce a particular response in target cells, 40 e.g., a reduction, slowing, or inhibition of cell growth. A therapeutically effective amount can be determined by first administering a low dose, and then incrementally increasing that dose until PAT059646-WO-PCT 5 the desired effect is achieved. A therapeutically effective amount can also vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art. The specific amount may vary depending on, for example, the particular pharmaceutical 10 composition, the subject and their age and existing health conditions or risk for health conditions, the dosing regimen to be followed, the severity of the disease, whether it is administered in combination with other agents, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried. In the case of cancer, a therapeutically effective amount of a compound may reduce the number of cancer cells, reduce 15 tumor size, inhibit (e.g., slow or stop) tumor metastasis, inhibit (e.g., slow or stop) tumor growth, and/or relieve one or more symptoms. [119] As used herein, the term “prophylactically effective amount” or “prophylactically effective dose,” refers to an amount of a compound disclosed herein, e.g., a panRAS inhibitor compound or composition described herein, that is effective, at dosages and for periods of time necessary, 20 to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount. In some embodiments, a prophylactically effective amount can prevent the onset of disease symptoms, including symptoms associated with a cancer. 25 [120] The term "alkyl", as used herein, refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation. The term "C1-C6alkyl", as used herein, refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to six carbon atoms, and which is attached to the rest of the molecule by a single bond. Non-30 limiting examples of "C1-C6alkyl" groups include methyl (a C1alkyl), ethyl (a C2alkyl), 1- methylethyl (a C3alkyl), n-propyl (a C3alkyl), isopropyl (a C3alkyl), n-butyl (a C4alkyl), isobutyl (a C4alkyl), sec-butyl (a C4alkyl), tert-butyl (a C4alkyl), n-pentyl (a C5alkyl), isopentyl (a C5alkyl), neopentyl (a C5alkyl) and hexyl (a C6alkyl). [121] The term “alkenyl”, as used herein, refers to a straight or branched hydrocarbon chain 35 radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond. The term “C2-C6alkenyl”, as used herein, refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, having from two to six carbon atoms, which is attached to the rest of the molecule by a single bond. Non-limiting examples of "C2-C6alkenyl" groups include ethenyl (a C2alkenyl), 40 prop-1-enyl (a C3alkenyl), but-1-enyl (a C4alkenyl), pent-1-enyl (a C5alkenyl), pent-4-enyl (a C5alkenyl), penta-1,4-dienyl (a C5alkenyl), hexa-1-enyl (a C6alkenyl), hexa-2-enyl (a C6alkenyl), PAT059646-WO-PCT 5 hexa-3-enyl (a C6alkenyl), hexa-1-,4-dienyl (a C6alkenyl), hexa-1-,5-dienyl (a C6alkenyl) and hexa-2-,4-dienyl (a C6alkenyl). The term “C2-C3alkenyl”, as used herein, refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, having from two to three carbon atoms, which is attached to the rest of the molecule by a single bond. Non-limiting examples of "C2-C3alkenyl" groups 10 include ethenyl (a C2alkenyl) and prop-1-enyl (a C3alkenyl). [122] The term "alkylene", as used herein, refers to a bivalent straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms and containing no unsaturation. The term "C1-C6alkylene", as used herein, refers to a bivalent straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no 15 unsaturation, having from one to six carbon atoms. Non-limiting examples of "C1-C6alkylene" groups include methylene (a C1alkylene), ethylene (a C2alkylene), 1-methylethylene (a C3alkylene), n-propylene (a C3alkylene), isopropylene (a C3alkylene), n-butylene (a C4alkylene), isobutylene (a C4alkylene), sec-butylene (a C4alkylene), tert-butylene (a C4alkylene), n- pentylene (a C5alkylene), isopentylene (a C5alkylene), neopentylene (a C5alkylene), and 20 hexylene (a C6alkylene). [123] The term “alkenylene”, as used herein, refers to a bivalent straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms and containing at least one double bond. The term “C2-C6alkenylene”, as used herein, refers to a bivalent straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, 25 containing at least one double bond, and having from two to six carbon atoms. Non-limiting examples of "C2-C6alkenylene" groups include ethenylene (a C2alkenylene), prop-1-enylene (a C3alkenylene), but-1-enylene (a C4alkenylene), pent-1-enylene (a C5alkenylene), pent-4- enylene (a C5alkenylene), penta-1,4-dienylene (a C5alkenylene), hexa-1-enylene (a C6alkenylene), hexa-2-enylene (a C6alkenylene), hexa-3-enylene (a C6alkenylene), hexa-1-,4- 30 dienylene (a C6alkenylene), hexa-1-,5-dienylene (a C6alkenylene) and hexa-2-,4-dienylene (a C6alkenylene). The term “C2-C6alkenylene”, as used herein, refers to a bivalent straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, and having from two to three carbon atoms. Non-limiting examples of "C2-C3alkenylene" groups include ethenylene (a C2alkenylene) and prop-1-enylene 35 (a C3alkenylene). [124] The term “cycloalkyl,” as used herein, refers to a non-aromatic, monocyclic, fused bicyclic, fused tricyclic or bridged polycyclic ring system. In some embodiments, the cycloalkyl is a mono- or bi-cyclic saturated carbocyclic group containing from 3 to 10 ring members, which may include fused, bridged or spiro ring systems. Non-limiting examples of fused bicyclic or 40 bridged polycyclic ring systems include bicyclo[1.1.1]pentane, bicyclo[2.1.1]hexane, bicyclo[2.2.1]heptane, bicyclo[3.1.1]heptane, bicyclo[3.2.1]octane, bicyclo[2.2.2]octane and 1 PAT059646-WO-PCT 5 adamantanyl. Non-limiting examples monocyclic C3-C8cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl groups. [125] The term heteroarylene, cycloalkylene, heterocycloalkylene mean a divalent heteroaryl, cycloalkyl and heterocycloalkyl. [126] The term “haloalkyl,” as used herein, refers to a linear or branched alkyl chain 10 substituted with one or more halogen groups in place of hydrogens along the hydrocarbon chain. Examples of halogen groups suitable for substitution in the haloalkyl group include Fluorine, Bromine, Chlorine, and Iodine. Haloalkyl groups may include substitution with multiple halogen groups in place of hydrogens in an alkyl chain, wherein said halogen groups can be attached to the same carbon or to another carbon in the alkyl chain. 15 [127] As used herein, the alkyl, alkenyl, alkynyl, alkoxy, amino, aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups may be optionally substituted by 1 to 4 groups selected from optionally substituted linear or branched (C1-C6)alkyl, optionally substituted linear or branched (C2- C6)alkenyl group, optionally substituted linear or branched (C2-C6)alkynyl group, optionally substituted linear or branched (C1-C6)alkoxy, optionally substituted (C1-C6)alkyl-S-, hydroxy, oxo 20 (or N-oxide where appropriate), nitro, cyano, -C(O)-OR0’, -O-C(O)-R0’, -C(O)-NR0’R0’’, -NR0’R0’’, -(C=NR0’)-OR0’’, linear or branched (C1-C6) haloalkyl, trifluoromethoxy, or halogen, wherein R0’ and R0’’ are each independently a hydrogen atom or an optionally substituted linear or branched (C1-C6)alkyl group, and wherein one or more of the carbon atoms of linear or branched (C1- C6)alkyl group is optionally deuterated. 25 Definitions for Terms in Drug Moieties [128] Those skilled in the art will appreciate that certain compounds described herein can exist in one or more different isomeric (e.g., stereoisomers, geometric isomers, atropisomers, tautomers) or isotopic (e.g., in which one or more atoms has been substituted with a different isotope of the atom, such as hydrogen substituted for deuterium) forms. Unless otherwise 30 indicated or clear from context, a depicted structure can be understood to represent any such isomeric or isotopic form, individually or in combination. [129] Compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. 35 [130] Compounds of the present disclosure that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C=N double bonds, and the like can also be present in the compounds described herein, and all such 40 stable isomers are contemplated in the present disclosure. Cis and trans geometric isomers of PAT059646-WO-PCT 5 the compounds of the present disclosure are described and may be isolated as a mixture of isomers or as separated isomeric forms. [131] In some embodiments, one or more compounds depicted herein may exist in different tautomeric forms. As will be clear from context, unless explicitly excluded, references to such compounds encompass all such tautomeric forms. In some embodiments, tautomeric forms 10 result from the swapping of a single bond with an adjacent double bond and the concomitant migration of a proton. In certain embodiments, a tautomeric form may be a prototropic tautomer, which is an isomeric protonation states having the same empirical formula and total charge as a reference form. Examples of moieties with prototropic tautomeric forms are ketone – enol pairs, amide – imidic acid pairs, lactam – lactim pairs, amide – imidic acid pairs, enamine – imine 15 pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, such as, 1H- and 3H-imidazole, 1H-, 2H- and 4H-1,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole. In some embodiments, tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution. In certain embodiments, tautomeric forms result from acetal interconversion. 20 [132] Unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. Exemplary isotopes that can be incorporated into compounds disclosed herein 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, 36Cl, 123I and 125I. Isotopically- 25 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 30 embodiments, 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 18F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy. [133] Preparations of isotopically labelled compounds are known to those of skill in the art. For 35 example, isotopically labeled compounds can generally be prepared by following procedures analogous to those disclosed for compounds described herein, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent. [134] As is known in the art, many chemical entities can adopt a variety of different solid forms such as, for example, amorphous forms or crystalline forms (e.g., polymorphs, hydrates, 40 solvate). In some embodiments, compounds disclosed herein may be utilized in any such form, PAT059646-WO-PCT 5 including in any solid form. In some embodiments, compounds described or depicted herein may be provided or utilized in hydrate or solvate form. [135] At various places in the present specification, substituents of compounds of the present disclosure are disclosed in groups or in ranges. It is specifically intended that the present disclosure includes each and every individual subcombination of the members of such groups 10 and ranges. For example, the term “C1-C6 alkyl” is specifically intended to individually disclose methyl, ethyl, C3 alkyl, C4 alkyl, C5 alkyl, and C6 alkyl. Furthermore, where a compound includes a plurality of positions at which substituents are disclosed in groups or in ranges, unless otherwise indicated, the present disclosure is intended to cover individual compounds and groups of compounds (e.g., genera and subgenera) containing each and every individual 15 subcombination of members at each position. [136] The term “optionally substituted X” (e.g., “optionally substituted alkyl”) is intended to be equivalent to “X, wherein X is optionally substituted” (e.g., “alkyl, wherein said alkyl is optionally substituted”). It is not intended to mean that the feature “X” (e.g., alkyl) per se is optional. As described herein, certain compounds of interest may contain one or more “optionally 20 substituted” moieties. In general, the term “substituted”, whether preceded by the term “optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent, e.g., any of the substituents or groups described herein. Unless otherwise indicated, an “optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may 25 be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. For example, in the term “optionally substituted C1-C6 alkyl-C2-C9 heteroaryl,” the alkyl portion, the heteroaryl portion, or both, may be optionally substituted. Combinations of substituents envisioned by the present disclosure are preferably those that result in the formation of stable or chemically feasible compounds. The 30 term “stable”, as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein. [137] Suitable monovalent substituents on a substitutable carbon atom of an “optionally substituted” group may be, independently, deuterium; halogen; -(CH2)0-4R°; -(CH2)0-4OR°; - 35 O(CH2)0-4Ro; -O-(CH2)0-4C(O)OR°; -(CH2)0-4CH(OR°)2; -(CH2)0-4SR°; -(CH2)0-4Ph, which may be substituted with R°; -(CH2)0-4O(CH2)0-1Ph which may be substituted with R°; -CH=CHPh, which may be substituted with R°; -(CH2)0-4O(CH2)0-1-pyridyl which may be substituted with R°; 4-8 membered saturated or unsaturated heterocycloalkyl (e.g., pyridyl); 3-8 membered saturated or unsaturated cycloalkyl (e.g., cyclopropyl, cyclobutyl, or cyclopentyl); -NO2; -CN; -N3; -(CH2)0-40 PAT059646-WO-PCT 5 (CH2)0-4C(O)R°; -C(S)R°; -(CH2)0-4C(O)OR°; -(CH2)0-4-C(O)-N(R°)2; -(CH2)0-4-C(O)-N(R°)-S(O)2- R°; -C(NCN)NR°2; -(CH2)0-4C(O)SR°; -(CH2)0-4C(O)OsiR°3; -(CH2)0-4OC(O)R°; -OC( O)(CH2)0- 4SR°; -SC(S)SR°; -(CH2)0-4SC(O)R°; -(CH2)0-4C(O)NR°2; -C(S)NR°2; -C(S)SR°; -(CH2)0- 4OC(O)NR°2; -C(O)N(OR°)R°; -C(O)C(O)R°; -C(O)CH2C(O)R°; -C(NOR°)R°; -(CH2)0-4SSR°; - (CH2)0-4S(O)2R°; -( CH2)0-4S(O)2OR°; -(CH2)0-4OS(O)2R°; -S(O)2NR°2; -(CH2)0-4S(O)R°; -10 N(R°)S(O)2NR°2; -N(R°)S(O)2R°; -N(OR°)R°; -C(NOR°)NR°2; -C(NH)NR°2; -P(O)2R°; -P(O)R°2; - P(O)(OR°)2; -OP(O)R°2; -OP(O)(OR°)2; -OP(O)(OR°)R°, -SiR°3; -(C1-4 straight or branched alkylene)O-N(R°)2; or –(C1-4 straight or branched alkylene)C(O)O-N(R°)2, wherein each R° may be substituted as defined below and is independently hydrogen, -C1-6 aliphatic, -CH2Ph, - O(CH2)0-1Ph, -CH2-(5-6 membered heteroaryl ring), or a 3-6-membered saturated, partially 15 unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which may be substituted as defined below. 20 [138] Suitable divalent substituents on a saturated carbon atom of an “optionally substituted” group include the following: =O, =S, =NNR* 2, =NNHC(O)R*, =NNHC(O)OR*, =NNHS(O)2R*, =NR*, =NOR*, -O(C(R*2))2-3O-, or -S(C(R* 2))2-3S-, wherein each independent occurrence of R* is selected from hydrogen, C1-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 25 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an “optionally substituted” group include: -O(CR* 2)2-3O-, wherein each independent occurrence of R* is selected from hydrogen, C1-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected 30 from nitrogen, oxygen, or sulfur. [139] Suitable substituents on the aliphatic group of R* include halogen, -R, -(haloR), -OH, - OR, -O(haloR), -CN, -C(O)OH, -C(O)OR, -NH2, -NHR, -NR 2, or -NO2, wherein each R is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C1-4 aliphatic, -CH2Ph, -O(CH2)0-1Ph, or a 5-6-membered saturated, partially 35 unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. [140] Suitable substituents on a substitutable nitrogen of an “optionally substituted” group include - R, -NR2, -C(O)R, -C(O)OR, -C(O)C(O)R, -C(O)CH2 C(NH)NR 2, or -N(R)S(O)2R; wherein each R is independently hydrogen, C -6 aliphatic which may 40 be substituted as defined below, unsubstituted -OPh, or an unsubstituted 3-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, PAT059646-WO-PCT 5 oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R, taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. [141] Suitable substituents on an aliphatic group of R are independently halogen, -R, -(haloR), - 10 OH, -OR, -O(haloR), -CN, -C(O)OH, -C(O)OR, -NH2, -NHR, -NR 2, or -NO2, wherein each R is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C1-4 aliphatic, -CH2Ph, -O(CH2)0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of R include =O and =S. 15 [142] The term “acetyl,” as used herein, refers to the group -C(O)CH3. [143] The term “alkoxy,” as used herein, refers to a -O-C1-C20 alkyl group, wherein the alkoxy group is attached to the remainder of the compound through an oxygen atom. [144] The term “alkyl,” as used herein, refers to a saturated, straight or branched monovalent hydrocarbon group containing from 1 to 20 (e.g., from 1 to 10 or from 1 to 6) carbons. In some 20 embodiments, an alkyl group is unbranched (i.e., is linear); in some embodiments, an alkyl group is branched. Alkyl groups are exemplified by, but not limited to, methyl, ethyl, n- and iso-propyl, n-, sec-, iso- and tert-butyl, and neopentyl. [145] The term "heteroalkyl,” as used herein, refers to an "alkyl" group, as defined herein, in which at least one carbon atom has been replaced with a heteroatom (e.g., an O, N, or S atom), 25 however the heteroalkyl group must contain at least one carbon atom after all replacements; for example, -NH-CH2-CH3, -CH2-NH-CH3, -CH2-N-(CH3)2, and -CH2-CH2-NH2 are heteroalkyls but - NH2 is not. When replacing a carbon with a heteroatom, an appropriate number of hydrogens are adjusted to fill out valences; for example, -CH2- can be replaced by groups such as -O, -S-, and -NH-, while -CH- can be replaced by groups such as -N-. The heteroatom may appear in 30 any part of the radical, for example in the middle or at either end of the radical. The number of carbons listed in the heteroalkyl refers to the number of carbons after all heteroatom replacements. For example, “C1-6 heteroalkyl” means that there are between 1 and 6 carbon atoms in the heteroalkyl group, but there are also one or more heteroatoms; for example, -O- CH2-CH3 is a C2 heteroalkyl, not a C3 heteroalkyl. Heteroalkyl is thus inclusive of amines, 35 alkoxys, and thioethers, in addition to other groups. In some embodiments the heteroatoms in a heteroalkyl are selected from the group consisting of N, O and S. [146] The term “alkylene,” as used herein, represents a saturated divalent hydrocarbon group derived from a straight or branched chain saturated hydrocarbon by the removal of two hydrogen atoms, and is exemplified by methylene, ethylene, isopropylene, and the like. The term “Cx-Cy 40 alkylene” represents alkylene groups having between x and y carbons. Exemplary values for x are 1, 2, 3, 4, 5, and 6, and exemplary values for y are 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or PAT059646-WO-PCT 5 20 (e.g., C1-C6, C1-C10, C2-C20, C2-C6, C2-C10, or C2-C20 alkylene). In some embodiments, the alkylene can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein. [147] The term “alkenyl,” as used herein, represents monovalent straight or branched chain groups of, unless otherwise specified, from 2 to 20 carbons (e.g., from 2 to 6 or from 2 to 10 carbons) containing one or more carbon-carbon double bonds and is exemplified by ethenyl, 1- 10 propenyl, 2-propenyl, 2-methyl-1-propenyl, 1-butenyl, and 2-butenyl. Alkenyls include both cis and trans isomers. The term “alkenylene,” as used herein, represents a divalent straight or branched chain groups of, unless otherwise specified, from 2 to 20 carbons (e.g., from 2 to 6 or from 2 to 10 carbons) containing one or more carbon-carbon double bonds. [148] The term “alkynyl,” as used herein, represents monovalent straight or branched chain 15 groups from 2 to 20 carbon atoms (e.g., from 2 to 4, from 2 to 6, or from 2 to 10 carbons) containing a carbon-carbon triple bond and is exemplified by ethynyl, and 1-propynyl. [149] The term “amino,” as used herein, represents -N(R)2, e.g., -NH2 and -N(CH3)2. [150] The term “aminoalkyl,” as used herein, represents an alkyl moiety substituted on one or more carbon atoms with one or more amino moieties. 20 [151] The term “aryl,” as used herein, represents a monovalent monocyclic, bicyclic, or multicyclic ring system formed by carbon atoms, wherein the ring attached to the pendant group is aromatic. Examples of aryl groups are phenyl, naphthyl, phenanthrenyl, and anthracenyl. An aryl ring can be attached to its pendant group at any heteroatom or carbon ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise 25 specified. In some embodiments, the aryl refers to a phenyl, nahthyl, biphenyl or indenyl group. [152] The term “C0,” as used herein, represents a bond. For example, part of the term - N(C(O)-(C0-C5 alkylene-H)- includes -N(C(O)-(C0 alkylene-H)-, which is also represented by - N(C(O)-H)-. [153] The terms “carbocyclic” and “carbocyclyl,” as used herein, refer to a monovalent, 30 optionally substituted C3-C12 monocyclic, bicyclic, or tricyclic ring structure, which may be bridged, fused or spirocyclic, in which all the rings are formed by carbon atoms and at least one ring is non-aromatic. [154] Carbocyclic structures include cycloalkyl, cycloalkenyl, and cycloalkynyl groups. Examples of carbocyclyl groups are cyclohexyl, cyclohexenyl, cyclooctynyl, 1,2-dihydronaphthyl, 35 1,2,3,4-tetrahydronaphthyl, fluorenyl, indenyl, indanyl, decalinyl, and the like. A carbocyclic ring can be attached to its pendant group at any ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified. [155] The term “carbonyl,” as used herein, represents a C(O) group, which can also be represented as C=O. 40 [156] The term “carboxyl,” as used herein, means -CO2H, (C=O)(OH), COOH, or C(O)OH or the unprotonated counterparts. PAT059646-WO-PCT 5 [157] The term “cyano,” as used herein, represents a -CN group. [158] The term “diastereomer,” as used herein, means stereoisomers that are not mirror images of one another and are non-superimposable on one another. [159] The term “enantiomer,” as used herein, means each individual optically active form of a compound disclosed herein, having an optical purity or enantiomeric excess (as determined by 10 methods standard in the art) of at least 80% (i.e., at least 90% of one enantiomer and at most 10% of the other enantiomer), preferably at least 90% and more preferably at least 98%. [160] The term “haloalkyl,” as used herein, represents an alkyl moiety substituted on one or more carbon atoms with one or more of the same or different halogen moieties. [161] The term “halogen,” or “halo” as used herein, represents a halogen selected from 15 bromine, chlorine, iodine, and fluorine. [162] The term “heteroaryl,” as used herein, represents a monovalent, monocyclic or polycyclic ring structure that contains at least one fully aromatic ring: i.e., they contain 4n+2 pi electrons within the monocyclic or polycyclic ring system and contains at least one ring heteroatom selected from N, O, or S in that aromatic ring. Exemplary unsubstituted heteroaryl 20 groups are of 1 to 12 (e.g., 1 to 11, 1 to 10, 1 to 9, 2 to 12, 2 to 11, 2 to 10, or 2 to 9) carbons. The term “heteroaryl” incIudes bicyclic, tricyclic, and tetracyclic groups in which any of the above heteroaromatic rings is fused to one or more, aryl or carbocyclic rings, e.g., a phenyl ring, or a cyclohexane ring. Examples of heteroaryl groups include, but are not limited to, pyridyl, pyrazolyl, benzooxazolyl, benzoimidazolyl, benzothiazolyl, imidazolyl, thiazolyl, quinolinyl, 25 tetrahydroquinolinyl, and 4-azaindolyl. A heteroaryl ring can be attached to its pendant group at any ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified. In some embodiments, the heteroaryl is substituted with 1, 2, 3, or 4 substituents groups. In some embodiments, the heteroaryl any mono- or bi-cyclic group composed of from 5 to 10 ring members, having at least one aromatic moiety and 30 containing from 1 to 4 hetero atoms selected from oxygen, sulfur and nitrogen (including quaternary nitrogens). [163] The term “heterocycloalkyl,” as used herein, represents a monovalent monocyclic, bicyclic or polycyclic ring system, which may be bridged, fused or spirocyclic, wherein at least one ring is non-aromatic and wherein the non-aromatic ring contains one, two, three, or four 35 heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. The 5-membered ring has zero to two double bonds, and the 6- and 7-membered rings have zero to three double bonds. Exemplary unsubstituted heterocycloalkyl groups are of 1 to 12 (e.g., 1 to 11, 1 to 10, 1 to 9, 2 to 12, 2 to 11, 2 to 10, or 2 to 9) carbons. [164] The term “heterocycloalkyl” also represents a heterocyclic compound having a bridged 40 multicyclic structure in which one or more carbons or heteroatoms bridges two non-adjacent members of a monocyclic ring, e.g., a quinuclidinyl group. The term “heterocycloalkyl” includes PAT059646-WO-PCT 5 bicyclic, tricyclic, and tetracyclic groups in which any of the above heterocyclic rings is fused to one or more aromatic, carbocyclic, heteroaromatic, or heterocyclic rings, e.g., an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring, a pyridine ring, or a pyrrolidine ring. [165] Examples of heterocycloalkyl groups are pyrrolidinyl, piperidinyl, 1,2,3,4- 10 tetrahydroquinolinyl, decahydroquinolinyl, dihydropyrrolopyridine, and decahydronapthyridinyl. A heterocycloalkyl ring can be attached to its pendant group at any ring atom that results in a stable structure and any of the ring atoms can be optionally substituted unless otherwise specified. [166] The term “hydroxy,” as used herein, represents a -OH group. 15 [167] The term “hydroxyalkyl,” as used herein, represents an alkyl moiety substituted on one or more carbon atoms with one or more -OH moieties. [168] The term “isomer,” as used herein, means any tautomer, stereoisomer, atropiosmer, enantiomer, or diastereomer of any compound disclosed herein. It is recognized that the compounds disclosed herein can have one or more chiral centers or double bonds and, 20 therefore, exist as stereoisomers, such as double-bond isomers (i.e., geometric E/Z isomers) or diastereomers (e.g., enantiomers (i.e., (+) or (-)) or cis/trans isomers). The chemical structures depicted herein, and therefore the compounds disclosed herein, encompass all the corresponding stereoisomers, that is, both the stereomerically pure form (e.g., geometrically pure, enantiomerically pure, or diastereomerically pure) and enantiomeric and stereoisomeric 25 mixtures, e.g., racemates. Enantiomeric and stereoisomeric mixtures of compounds disclosed herein can typically be resolved into their component enantiomers or stereoisomers by well- known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent. Enantiomers and stereoisomers can also be obtained from 30 stereomerically or enantiomerically pure intermediates, reagents, and catalysts by well-known asymmetric synthetic methods. [169] The term “stereoisomer,” as used herein, refers to all possible different isomeric as well as conformational forms which a compound may possess (e.g., a compound of any formula described herein), in particular all possible stereochemically and conformationally isomeric 35 forms, all diastereomers, enantiomers or conformers of the basic molecular structure, including atropisomers. Some compounds of the present disclosure may exist in different tautomeric forms, all of the latter being included within the scope of the present invention. [170] The term “sulfonyl” or "sulphonyl," as used herein, represents an -S(O)2- group. [171] The term “thiocarbonyl,” as used herein, refers to a -C(S)- group. 40 PAT059646-WO-PCT 5 Therapeutic Uses and Compositions [172] Disclosed herein are methods of using the compositions described herein, e.g., the disclosed panRAS inhibitor compounds and compositions, in treating a subject for a disorder, e.g., a cancer. Compositions, e.g., panRAS inhibitors, may be administered alone or in 10 combination with at least one additional inactive and/or active agent, e.g., at least one additional therapeutic agent, and may be administered in any pharmaceutically acceptable formulation, dosage, and dosing regimen. Treatment efficacy may be evaluated for toxicity as well as indicators of efficacy and adjusted accordingly. Efficacy measures include, but are not limited to, a cytostatic and/or cytotoxic effect observed in vitro or in vivo, reduced tumor volume, tumor 15 growth inhibition, and/or prolonged survival. [173] Methods of determining whether a panRAS inhibitor exerts a cytostatic and/or cytotoxic effect on a cell are known. For example, the cytotoxic or cytostatic activity of a panRAS inhibitor can be measured by, e.g., exposing mammalian cells to the panRAS inhibitor in a cell culture medium; culturing the cells for a period from about 6 hr to about 6 days; and measuring cell 20 viability (e.g., using a CellTiter-Glo® (CTG) or MTT cell viability assay). Cell-based in vitro assays may also be used to measure viability (proliferation), cytotoxicity, and induction of apoptosis (caspase activation) of the panRAS inhibitor. [174] For determining cytotoxicity, necrosis or apoptosis (programmed cell death) may be measured. Necrosis is typically accompanied by increased permeability of the plasma 25 membrane, swelling of the cell, and rupture of the plasma membrane. Apoptosis can be quantitated, for example, by measuring DNA fragmentation. Commercial photometric methods for the quantitative in vitro determination of DNA fragmentation are available. Examples of such assays, including TUNEL (which detects incorporation of labeled nucleotides in fragmented DNA) and ELISA-based assays, are described in Biochemica (1999) 2:34-7 (Roche Molecular 30 Biochemicals). [175] Apoptosis may also be determined by measuring morphological changes in a cell. For example, as with necrosis, loss of plasma membrane integrity can be determined by measuring uptake of certain dyes (e.g., a fluorescent dye such as, for example, acridine orange or ethidium bromide). A method for measuring apoptotic cell number has been described by Duke and 35 Cohen, Current Protocols in Immunology (Coligan et al., eds. (1992) pp.3.17.1-3.17.16). Cells also can be labeled with a DNA dye (e.g., acridine orange, ethidium bromide, or propidium iodide) and the cells observed for chromatin condensation and margination along the inner nuclear membrane. Apoptosis may also be determined, in some embodiments, by screening for caspase activity. In some embodiments, a Caspase-Glo® Assay can be used to measure 40 activity of caspase-3 and caspase-7. In some embodiments, the assay provides a luminogenic caspase-3/7 substrate in a reagent optimized for caspase activity, luciferase activity, and cell PAT059646-WO-PCT 5 lysis. In some embodiments, adding Caspase-Glo® 3/7 Reagent in an “add-mix-measure” format may result in cell lysis, followed by caspase cleavage of the substrate and generation of a “glow-type” luminescent signal, produced by luciferase. In some embodiments, luminescence may be proportional to the amount of caspase activity present, and can serve as an indicator of apoptosis. Other morphological changes that can be measured to determine apoptosis include, 10 e.g., cytoplasmic condensation, increased membrane blebbing, and cellular shrinkage. Determination of any of these effects on cancer cells indicates that a panRAS inhibitor is useful in the treatment of cancers. [176] Cell viability may be measured, e.g., by determining in a cell the uptake of a dye such as neutral red, trypan blue, Crystal Violet, or ALAMAR™ blue (see, e.g., Page et al. (1993) Intl J 15 Oncology 3:473-6). In such an assay, the cells are incubated in media containing the dye, the cells are washed, and the remaining dye, reflecting cellular uptake of the dye, is measured spectrophotometrically. [177] Cell viability may also be measured, e.g., by quantifying ATP, an indicator of metabolically active cells. In some embodiments, in vitro potency and/or cell viability of 20 prepared panRAS inhibitor compounds may be assessed using a CellTiter-Glo® (CTG) cell viability assay, as described in the examples provided herein. In this assay, in some embodiments, the single reagent (CellTiter-Glo® Reagent) is added directly to cells cultured in serum-supplemented medium. The addition of reagent results in cell lysis and generation of a luminescent signal proportional to the amount of ATP present. The amount of ATP is directly 25 proportional to the number of cells present in culture. [178] Cell viability may also be measured, e.g., by measuring the reduction of tetrazolium salts. In some embodiments, in vitro potency and/or cell viability of prepared panRAS inhibitor compounds may be assessed using an MTT cell viability assay, as described in the examples provided herein. In this assay, in some embodiments, the yellow tetrazolium MTT (3-(4, 5- 30 dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide) is reduced by metabolically active cells, in part by the action of dehydrogenase enzymes, to generate reducing equivalents such as NADH and NADPH. The resulting intracellular purple formazan can then be solubilized and quantified by spectrophotometric means. [179] In certain aspects, the present disclosure features a method of killing, inhibiting or 35 modulating the growth of a cancer cell or tissue by disrupting the expression and/or activity of panRAS (e.g., K-Ras (including splice variants KRAS4A and KRAS4B), H-Ras and N-Ras) and/or one or more upstream modulators or downstream targets thereof. The method may be used with any subject where disruption of panRAS (e.g., K-Ras (including splice variants KRAS4A and KRAS4B), H-Ras and N-Ras) expression and/or activity provides a therapeutic 40 benefit. Subjects that may benefit from disrupting panRAS (e.g., K-Ras (including splice variants KRAS4A and KRAS4B), H-Ras and N-Ras) expression and/or activity include, but are 1 PAT059646-WO-PCT 5 not limited to, those having or at risk of having a cancer such as a tumor or a hematological cancer. In some embodiments, the cancer is a breast cancer including ER positive breast cancer, multiple myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, bladder cancer, brain cancer, bone marrow cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal 10 cancer, hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant prostate cancer, bladder urothelial carcinoma, melanoma, myelogenous leukemia, myeloma, oral cancer, ovarian cancer, non-small cell lung cancer, prostate cancer, small cell lung cancer, spleen cancer, or head and neck cancer. 15 [180] In some embodiments, the disclosed panRAS inhibitors may be administered in any cell or tissue that expresses EphA2, such as a EphA2-expressing cancer cell or tissue. An exemplary embodiment includes a method of killing a EphA2-expressing cancer cell or tissue. The method may be used with any cell or tissue that expresses EphA2, such as a cancerous cell or a metastatic lesion. Non-limiting examples of EphA2-expressing cancers include breast 20 cancer, non-small cell lung cancer, pancreatic cancer, esophageal cancer, head and neck cancer, gastric or stomach cancer, bladder cancer, and colorectal cancer. [181] In some embodiments, the disclosed panRAS inhibitors may be administered in any cell or tissue that expresses B7-H3 (CD276), such as a B7-H3 (CD276)-expressing cancer cell or tissue. An exemplary embodiment includes a method of killing a B7-H3 (CD276)-expressing 25 cancer cell or tissue. The method may be used with any cell or tissue that expresses B7-H3 (CD276), such as a cancerous cell or a metastatic lesion. Non-limiting examples of B7-H3 (CD276)-expressing cancers include colorectal cancer, pancreatic cancer, lymphoma, non-small cell lung cancer, small cell lung cancer, breast cancer including ER positive breast cancer, metastatic castration resistant prostate cancer, melanoma, bladder urothelial carcinoma, head 30 and neck cancer, and leukemia (e.g., acute myeloid leukemia). [182] Exemplary methods include the steps of contacting a cell with a panRAS inhibitor, as described herein, in an effective amount, i.e., an amount sufficient to kill the cell. The method can be used on cells in culture, e.g., in vitro, in vivo, ex vivo, or in situ. For example, cells that express EphA2 (e.g., cells collected by biopsy of a tumor or metastatic lesion; cells from an 35 established cancer cell line; or recombinant cells), can be cultured in vitro in culture medium and the contacting step can be affected by adding the panRAS inhibitor to the culture medium. The method will result in killing of cells expressing EphA2, including in particular cancer cells expressing EphA2. Alternatively, the panRAS inhibitor can be administered to a subject by any suitable administration route (e.g., intravenous, subcutaneous, or direct contact with a tumor 40 tissue) to have an effect in vivo. PAT059646-WO-PCT 5 [183] The in vivo effect of a disclosed panRAS inhibitor therapeutic composition can be evaluated in a suitable animal model. For example, xenogeneic cancer models can be used, wherein cancer explants or passaged xenograft tissues are introduced into immune compromised animals, such as nude or SCID mice (Klein et al. (1997) Nature Med.3:402-8). Efficacy may be predicted using assays that measure inhibition of tumor formation, tumor 10 regression or metastasis, and the like. [184] In vivo assays that evaluate the promotion of tumor death by mechanisms such as apoptosis may also be used. In some embodiments, xenografts from tumor bearing mice treated with the therapeutic composition can be examined for the presence of apoptotic foci and compared to untreated control xenograft-bearing mice. The extent to which apoptotic foci are 15 found in the tumors of the treated mice provides an indication of the therapeutic efficacy of the composition. [185] Further provided herein are methods of treating a disorder, e.g., a cancer. The compositions described herein, e.g., the panRAS inhibitors disclosed herein, can be administered to a non-human mammal or human subject for therapeutic purposes. The 20 therapeutic methods include administering to a subject having or suspected of having a cancer a therapeutically effective amount of a composition comprising a panRAS inhibitor. [186] An exemplary embodiment is a method of treating a subject having or suspected of having a cancer, comprising administering to the subject a therapeutically effective amount of a composition disclosed herein, e.g., a panRAS inhibitor, composition, or pharmaceutical 25 composition (e.g., any of the exemplary panRAS inhibitors, compositions, or pharmaceutical compositions disclosed herein). In some embodiments, the cancer is a tumor or a hematological cancer. In some embodiments, the cancer is a breast cancer including ER positive breast cancer, multiple myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, bladder cancer, brain cancer, bone marrow 30 cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal cancer, hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant prostate cancer, bladder urothelial carcinoma, melanoma, myelogenous leukemia, myeloma, oral cancer, ovarian cancer, non-small cell lung cancer, 35 prostate cancer, small cell lung cancer, spleen cancer, or head and neck cancer. [187] In certain aspects, the present disclosure further provides methods of reducing or inhibiting growth of a tumor, comprising administering a therapeutically effective amount of a panRAS inhibitor or composition comprising a panRAS inhibitor. In some embodiments, the treatment is sufficient to reduce or inhibit the growth of the patient's tumor, reduce the number 40 or size of metastatic lesions, reduce tumor load, reduce primary tumor load, reduce invasiveness, prolong survival time, and/or maintain or improve the quality of life. In some PAT059646-WO-PCT 5 embodiments, the tumor is resistant or refractory to treatment with the panRAS inhibitor when administered alone. [188] An exemplary embodiment is a method of reducing or inhibiting the growth of a tumor in a subject, comprising administering to the subject a therapeutically effective amount of an panRAS inhibitor, composition, or pharmaceutical composition (e.g., any of the exemplary 10 panRAS inhibitors, compositions, or pharmaceutical compositions disclosed herein). In some embodiments, the tumor is a breast cancer including ER positive breast cancer, multiple myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, bladder cancer, brain cancer, bone marrow cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal cancer, 15 hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant prostate cancer, bladder urothelial carcinoma, melanoma, myelogenous leukemia, myeloma, oral cancer, ovarian cancer, non-small cell lung cancer, prostate cancer, small cell lung cancer, spleen cancer, or head and neck cancer. In some embodiments, the tumor is a gastric cancer. 20 In some embodiments, administration of the panRAS inhibitor, composition, or pharmaceutical composition reduces or inhibits the growth of the tumor by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, or at least about 99%, as compared to growth in the absence of treatment. 25 [189] Another exemplary embodiment is a method of delaying or slowing the growth of a tumor in a subject, comprising administering to the subject a therapeutically effective amount of a panRAS inhibitor, composition, or pharmaceutical composition (e.g., any of the exemplary panRAS inhibitors, compositions, or pharmaceutical compositions disclosed herein). In some embodiments, the tumor is a breast cancer including ER positive breast cancer, multiple 30 myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, bladder cancer, brain cancer, bone marrow cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal cancer, hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant 35 prostate cancer, bladder urothelial carcinoma, melanoma, myelogenous leukemia, myeloma, oral cancer, ovarian cancer, non-small cell lung cancer, prostate cancer, small cell lung cancer, spleen cancer, or head and neck cancer. In some embodiments, the tumor is a gastric cancer. In some embodiments, administration of the panRAS inhibitor, composition, or pharmaceutical composition delays or slows the growth of the tumor by at least about 10%, at least about 20%, 40 at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about PAT059646-WO-PCT 5 70%, at least about 80%, at least about 90%, at least about 95%, or at least about 99%, as compared to growth in the absence of treatment. [190] In certain aspects, the present disclosure further provides methods of reducing or slowing the expansion of a cancer cell population, comprising administering a therapeutically effective amount of a panRAS inhibitor or composition comprising a panRAS inhibitor. 10 [191] An exemplary embodiment is a method of reducing or slowing the expansion of a cancer cell population in a subject, comprising administering to the subject a therapeutically effective amount of a panRAS inhibitor, composition, or pharmaceutical composition (e.g., any of the exemplary panRAS inhibitors, compositions, or pharmaceutical compositions disclosed herein). In some embodiments, the cancer cell population is from a tumor or a hematological cancer. In 15 some embodiments, the cancer cell population is a breast cancer including ER positive breast cancer, multiple myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, bladder cancer, brain cancer, bone marrow cancer, cervical cancer, chronic lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal cancer, hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, 20 follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant prostate cancer, bladder urothelial carcinoma, melanoma, myelogenous leukemia, myeloma, oral cancer, ovarian cancer, non-small cell lung cancer, prostate cancer, small cell lung cancer, spleen cancer, or head and neck cancer. In some embodiments, administration of the panRAS inhibitor, composition, or pharmaceutical composition reduces the cancer cell 25 population by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, or at least about 99%, as compared to the population in the absence of treatment. In some embodiments, administration of the panRAS inhibitor, composition, or pharmaceutical composition slows the expansion of the cancer cell population by at least about 30 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, or at least about 99%, as compared to expansion in the absence of treatment. [192] Also provided herein are methods of determining whether a subject having or suspected of having a cancer will be responsive to treatment with the disclosed panRAS inhibitors and 35 compositions. An exemplary embodiment is a method of determining whether a subject having or suspected of having a cancer will be responsive to treatment with a panRAS inhibitor, composition, or pharmaceutical composition (e.g., any of the exemplary panRAS inhibitors, compositions, or pharmaceutical compositions disclosed herein) by providing a biological sample from the subject; contacting the sample with the panRAS inhibitor; and detecting binding 40 of the panRAS inhibitor to cancer cells in the sample. In some embodiments, the sample is a tissue biopsy sample, a blood sample, or a bone marrow sample. In some embodiments, the PAT059646-WO-PCT 5 method comprises providing a biological sample from the subject; contacting the sample with the panRAS inhibitor; and detecting one or more markers of cancer cell death in the sample (e.g., increased expression of one or more apoptotic markers, reduced expansion of a cancer cell population in culture, etc.). [193] Further provided herein are therapeutic uses of the disclosed panRAS inhibitors and 10 compositions. An exemplary embodiment is a panRAS inhibitor, composition, or pharmaceutical composition (e.g., any of the exemplary panRAS inhibitors, compositions, or pharmaceutical compositions disclosed herein) for use in treating a subject having or suspected of having a cancer. Another exemplary embodiment is a use of an panRAS inhibitor, composition, or pharmaceutical composition (e.g., any of the exemplary panRAS inhibitors, 15 compositions, or pharmaceutical compositions disclosed herein) in treating a subject having or suspected of having a cancer. Another exemplary embodiment is a use of an panRAS inhibitor, composition, or pharmaceutical composition (e.g., any of the exemplary panRAS inhibitors, compositions, or pharmaceutical compositions disclosed herein) in a method of manufacturing a medicament for treating a subject having or suspected of having a cancer. 20 [194] Moreover, panRAS inhibitors of the present disclosure may be administered to a non- human mammal for veterinary purposes or as an animal model of human disease. Regarding the latter, such animal models may be useful for evaluating the therapeutic efficacy of the disclosed panRAS inhibitors (e.g., testing of dosages and time courses of administration). [195] The therapeutic compositions used in the practice of the foregoing methods may be 25 formulated into pharmaceutical compositions comprising a pharmaceutically acceptable carrier suitable for the desired delivery method. An exemplary embodiment is a pharmaceutical composition comprising an panRAS inhibitor of the present disclosure and a pharmaceutically acceptable carrier, e.g., one suitable for a chosen means of administration, e.g., intravenous administration. The pharmaceutical composition may also comprise one or more additional 30 inactive and/or therapeutic agents that are suitable for treating or preventing, for example, a cancer (e.g., a standard-of-care agent, etc.). The pharmaceutical composition may also comprise one or more carrier, excipient, and/or stabilizer components, and the like. Methods of formulating such pharmaceutical compositions and suitable formulations are known in the art (see, e.g., “Remington’s Pharmaceutical Sciences,” Mack Publishing Co., Easton, PA). 35 [196] Suitable carriers include any material that, when combined with the therapeutic composition, retains the anti-tumor function of the therapeutic composition and is generally non- reactive with the patient’s immune system. Pharmaceutically acceptable carriers include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Examples of 40 pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol, mesylate salt, and the like, as well as combinations thereof. PAT059646-WO-PCT 5 In many cases, isotonic agents are included, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Pharmaceutically acceptable carriers may further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the panRAS inhibitor. 10 [197] A pharmaceutical composition of the present disclosure can be administered by a variety of methods known in the art. The route and/or mode of administration may vary depending upon the desired results. In some embodiments, the therapeutic formulation is solubilized and administered via any route capable of delivering the therapeutic composition to the cancer site. Potentially effective routes of administration include, but are not limited to, parenteral (e.g., 15 intravenous, subcutaneous), intraperitoneal, intramuscular, intratumor, intradermal, intraorgan, orthotopic, and the like. In some embodiments, the administration is intravenous, subcutaneous, intraperitoneal, or intramuscular. The pharmaceutically acceptable carrier should be suitable for the route of administration, e.g., intravenous or subcutaneous administration (e.g., by injection or infusion). Depending on the route of administration, the 20 active compound(s), i.e., the panRAS inhibitor and/or any additional therapeutic agent, may be coated in a material to protect the compound(s) from the action of acids and other natural conditions that may inactivate the compound(s). Administration can be either systemic or local. [198] The therapeutic compositions disclosed herein may be sterile and stable under the conditions of manufacture and storage, and may be in a variety of forms. These include, for 25 example, liquid, semi-solid, and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes, and suppositories. The form depends on the intended mode of administration and therapeutic application. In some embodiments, the disclosed panRAS inhibitors can be incorporated into a pharmaceutical composition suitable for parenteral administration. The injectable solution may30 be composed of either a liquid or lyophilized dosage form in a flint or amber vial, ampule, or pre- filled syringe, or other known delivery or storage device. In some embodiments, one or more of the panRAS inhibitors or pharmaceutical compositions is supplied as a dry sterilized lyophilized powder or water free concentrate in a hermetically sealed container and can be reconstituted (e.g., with water or saline) to the appropriate concentration for administration to a subject. 35 [199] Typically, a therapeutically effective amount or efficacious amount of a disclosed composition, e.g., a disclosed panRAS inhibitor, is employed in the pharmaceutical compositions of the present disclosure. The composition, e.g., one comprising an panRAS inhibitor, may be formulated into a pharmaceutically acceptable dosage form by conventional methods known in the art. Dosages and administration protocols for the treatment of cancers 40 using the foregoing methods will vary with the method and the target cancer, and will generally depend on a number of other factors appreciated in the art. PAT059646-WO-PCT 5 [200] Dosage regimens for compositions disclosed herein, e.g., those comprising panRAS inhibitors alone or in combination with at least one additional inactive and/or active therapeutic agent, may be adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single bolus of one or both agents may be administered at one time, several divided doses may be administered over a predetermined period of time, or the dose of one or 10 both agents may be proportionally increased or decreased as indicated by the exigencies of the therapeutic situation. In some embodiments, treatment involves single bolus or repeated administration of the panRAS inhibitor preparation via an acceptable route of administration. In some embodiments, the panRAS inhibitor is administered to the patient daily, weekly, monthly, or any time period in between. For any particular subject, specific dosage regimens may be 15 adjusted over time according to the individual’s need, and the professional judgment of the treating clinician. Parenteral compositions may be formulated in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect 20 in association with the required pharmaceutical carrier. [201] Dosage values for compositions comprising a panRAS inhibitor and/or any additional therapeutic agent(s), may be selected based on the unique characteristics of the active compound(s), and the particular therapeutic effect to be achieved. A physician or veterinarian can start doses of the panRAS inhibitor employed in the pharmaceutical composition at levels 25 lower than that required to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. In general, effective doses of the compositions of the present disclosure, for the treatment of a cancer may vary depending upon many different factors, including means of administration, target site, physiological state of the patient, whether the patient is human or an animal, other medications administered, and whether treatment is 30 prophylactic or therapeutic. The selected dosage level may also depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present disclosure employed, or the ester, salt, or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular 35 compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors. Treatment dosages may be titrated to optimize safety and efficacy. [202] Toxicity and therapeutic efficacy of compounds provided herein can be determined by standard pharmaceutical procedures in cell culture or in animal models. For example, LD50, 40 ED50, EC50, and IC50 may be determined, and the dose ratio between toxic and therapeutic effects (LD50/ED50) may be calculated as the therapeutic index. The data obtained from in PAT059646-WO-PCT 5 vitro and in vivo assays can be used in estimating or formulating a range of dosage for use in humans. For example, the compositions and methods disclosed herein may initially be evaluated in xenogeneic cancer models (e.g., an NCI-H929 multiple myeloma mouse model). [203] In some embodiments, a panRAS inhibitor or composition comprising a panRAS inhibitor is administered on a single occasion. In other embodiments, a panRAS inhibitor or composition 10 comprising a panRAS inhibitor is administered on multiple occasions. Intervals between single dosages can be, e.g., daily, weekly, monthly, or yearly. Intervals can also be irregular, based on measuring blood levels of the administered agent (e.g., the panRAS inhibitor) in the patient in order to maintain a relatively consistent plasma concentration of the agent. The dosage and frequency of administration of a panRAS inhibitor or composition comprising a panRAS inhibitor 15 may also vary depending on whether the treatment is prophylactic or therapeutic. In prophylactic applications, a relatively low dosage may be administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives. In therapeutic applications, a relatively higher dosage at relatively shorter intervals is sometimes required until progression of the disease is reduced or terminated, and preferably 20 until the patient shows partial or complete amelioration of one or more symptoms of disease. Thereafter, the patient may be administered a lower, e.g., prophylactic regime. [204] The above therapeutic approaches can be combined with any one of a wide variety of additional surgical, chemotherapy, or radiation therapy regimens. In some embodiments, the panRAS inhibitors or compositions disclosed herein are co-formulated and/or co-administered 25 with one or more additional therapeutic agents, e.g., one or more chemotherapeutic agents, one or more standard-of-care agents for the particular condition being treated. [205] Kits for use in the therapeutic and/or diagnostic applications described herein are also provided. Such kits may comprise a carrier, package, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) 30 comprising one of the separate elements to be used in a method disclosed herein. A label may be present on or with the container(s) to indicate that a panRAS inhibitor or composition within the kit is used for a specific therapy or non-therapeutic application, such as a prognostic, prophylactic, diagnostic, or laboratory application. A label may also indicate directions for either in vivo or in vitro use, such as those described herein. Directions and or other information may 35 also be included on an insert(s) or label(s), which is included with or on the kit. The label may be on or associated with the container. A label may be on a container when letters, numbers, or other characters forming the label are molded or etched into the container itself. A label may be associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert. The label may indicate that a panRAS inhibitor or 40 composition within the kit is used for diagnosing or treating a condition, such as a cancer a described herein. PAT059646-WO-PCT 5 [206] In some embodiments, a kit comprises a panRAS inhibitor or composition comprising a panRAS inhibitor. In some embodiments, the kit further comprises one or more additional components, including but not limited to: instructions for use; other reagents, e.g., a therapeutic agent (e.g., a standard-of-care agent); devices, containers, or other materials for preparing the panRAS inhibitor for administration; pharmaceutically acceptable carriers; and devices, 10 containers, or other materials for administering the panRAS inhibitor to a subject. Instructions for use can include guidance for therapeutic applications including suggested dosages and/or modes of administration, e.g., in a patient having or suspected of having a cancer. In some embodiments, the kit comprises a panRAS inhibitor and instructions for use of the panRAS inhibitor in treating, preventing, and/or diagnosing a cancer. 15 [207] It is known that elevated panRAS (e.g., K-Ras (including splice variants KRAS4A and KRAS4B), H-Ras and N-Ras) expression correlates with resistance to radiation therapy and chemotherapy. PanRAS inhibitors that may not be sufficiently effective as monotherapy to treat cancer can be administered in combination with other therapeutic agents (including non- targeted and targeted therapeutic agents) or radiation therapy (including radioligand therapy) to 20 provide therapeutic benefit. Without wishing to be bound by theory, it is believed that the panRAS inhibitor s described herein sensitize tumor cells to the treatment with other therapeutic agents (including standard of care chemotherapeutic agents to which the tumor cells may have developed resistance) and/or radiation therapy. In some embodiments, panRAS inhibitors described herein, are administered to a subject having cancer in an amount effective to 25 sensitize the tumor cells. As used herein, the term “sensitize” means that the treatment with panRAS inhibitor increases the potency or efficacy of the treatment with other therapeutic agents and/or radiation therapy against tumor cells. COMBINATION THERAPIES 30 [208] In some embodiments, the present disclosure provides methods of treatment wherein the panRAS inhibitors disclosed herein are administered in combination with one or more (e.g., 1 or 2) additional therapeutic agents. Exemplary combination partners are disclosed herein. [209] In certain embodiments, a combination described herein comprises a PD-1 inhibitor. In some embodiments, the PD-1 inhibitor is chosen from PDR001 (Novartis), Nivolumab (Bristol- 35 Myers Squibb), Pembrolizumab (Merck & Co), Pidilizumab (CureTech), MEDI0680 (Medimmune), REGN2810 (Regeneron), TSR-042 (Tesaro), PF-06801591 (Pfizer), BGB-A317 (Beigene), BGB-108 (Beigene), INCSHR1210 (Incyte), or AMP-224 (Amplimmune). In some embodiments, the PD-1 inhibitor is PDR001. PDR001 is also known as Spartalizumab. [210] In certain embodiments, a combination described herein comprises a LAG-3 inhibitor. In 40 some embodiments, the LAG-3 inhibitor is chosen from LAG525 (Novartis), BMS-986016 (Bristol-Myers Squibb), or TSR-033 (Tesaro). PAT059646-WO-PCT 5 [211] In certain embodiments, a combination described herein comprises a TIM-3 inhibitor. In some embodiments, the TIM-3 inhibitor is MBG453 (Novartis), TSR-022 (Tesaro), LY-3321367 (Eli Lily), Sym23 (Symphogen), BGB-A425 (Beigene), INCAGN-2390 (Agenus), BMS-986258 (BMS), RO-7121661 (Roche), or LY-3415244 (Eli Lilly). [212] In certain embodiments, a combination described herein comprises a PDL1 inhibitor. In 10 one embodiment, the PDL1 inhibitor is chosen from FAZ053 (Novartis), atezolizumab (Genentech), durvalumab (Astra Zeneca), or avelumab (Pfizer). [213] In certain embodiments, a combination described herein comprises a GITR agonist. In some embodiments, the GITR agonist is chosen from GWN323 (NVS), BMS-986156, MK-4166 or MK-1248 (Merck), TRX518 (Leap Therapeutics), INCAGN1876 (Incyte/Agenus), AMG 228 15 (Amgen) or INBRX-110 (Inhibrx). [214] In some embodiments, a combination described herein comprises an IAP inhibitor. In some embodiments, the IAP inhibitor comprises LCL161 or a compound disclosed in International Application Publication No. WO 2008/016893. [215] In an embodiment, the combination comprises an mTOR inhibitor, e.g., RAD001 (also 20 known as everolimus). [216] In an embodiment, the combination comprises a HDAC inhibitor, e.g., LBH589. LBH589 is also known as panobinostat. [217] In an embodiment, the combination comprises an IL-17 inhibitor, e.g., CJM112. [218] In certain embodiments, a combination described herein comprises an estrogen receptor 25 (ER) antagonist. In some embodiments, the estrogen receptor antagonist is used in combination with a PD-1 inhibitor, a CDK4/6 inhibitor, or both. In some embodiments, the combination is used to treat an ER positive (ER+) cancer or a breast cancer (e.g., an ER+ breast cancer). [219] In some embodiments, the estrogen receptor antagonist is a selective estrogen receptor 30 degrader (SERD). SERDs are estrogen receptor antagonists which bind to the receptor and result in e.g., degradation or down-regulation of the receptor (Boer K. et al., (2017) Therapeutic Advances in Medical Oncology 9(7): 465-479). ER is a hormone-activated transcription factor important for e.g., the growth, development and physiology of the human reproductive system. ER is activated by, e.g., the hormone estrogen (17beta estradiol). ER expression and signaling 35 is implicated in cancers (e.g., breast cancer), e.g., ER positive (ER+) breast cancer. In some embodiments, the SERD is chosen from LSZ102, fulvestrant, brilanestrant, or elacestrant. [220] In some embodiments, the SERD comprises a compound disclosed in International Application Publication No. WO 2014/130310, which is hereby incorporated by reference in its entirety. 40 [221] In some embodiments, the SERD comprises LSZ102. LSZ102 has the chemical name: (E)-3-(4-((2-(2-(1,1-difluoroethyl)-4-fluorophenyl)-6-hydroxybenzo[b]thiophen-3- PAT059646-WO-PCT 5 yl)oxy)phenyl)acrylic acid. In some embodiments, the SERD comprises fulvestrant (CAS Registry Number: 129453-61-8), or a compound disclosed in International Application Publication No. WO 2001/051056, which is hereby incorporated by reference in its entirety. In some embodiments, the SERD comprises elacestrant (CAS Registry Number: 722533-56-4), or a compound disclosed in U.S. Patent No.7,612,114, which is incorporated by reference in its10 entirety. Elacestrant is also known as RAD1901, ER-306323 or (6R)-6-{2-[Ethyl({4-[2- (ethylamino)ethyl]phenyl}methyl)amino]-4-methoxyphenyl}-5,6,7,8-tetrahydronaphthalen-2-ol. Elacestrant is an orally bioavailable, non-steroidal combined selective estrogens receptor modulator (SERM) and a SERD. Elacestrant is also disclosed, e.g., in Garner F et al., (2015) Anticancer Drugs 26(9):948-56. In some embodiments, the SERD is brilanestrant (CAS Registry 15 Number: 1365888-06-7), or a compound disclosed in International Application Publication No. WO 2015/136017, which is incorporated by reference in its entirety. [222] In some embodiments, the SERD is chosen from RU 58668, GW7604, AZD9496, bazedoxifene, pipendoxifene, arzoxifene, OP-1074, or acolbifene, e.g., as disclosed in McDonell et al. (2015) Journal of Medicinal Chemistry 58(12) 4883-4887. 20 [223] Other exemplary estrogen receptor antagonists are disclosed, e.g., in WO 2011/156518, WO 2011/159769, WO 2012/037410, WO 2012/037411, and US 2012/0071535, all of which are hereby incorporated by reference in their entirety. [224] In certain embodiments, a combination described herein comprises an inhibitor of Cyclin-Dependent Kinases 4 or 6 (CDK4/6). In some embodiments, the CDK4/6 inhibitor is 25 used in combination with a PD-1 inhibitor, an estrogen receptor (ER) antagonist, or both. In some embodiments, the combination is used to treat an ER positive (ER+) cancer or a breast cancer (e.g., an ER+ breast cancer). In some embodiments, the CDK4/6 inhibitor is chosen from ribociclib, abemaciclib (Eli Lilly), or palbociclib. [225] In some embodiments, the CDK4/6 inhibitor comprises ribociclib (CAS Registry Number: 30 1211441-98-3), or a compound disclosed in U.S. Patent Nos.8,415,355 and 8,685,980, which are incorporated by reference in their entirety. [226] In some embodiments, the CDK4/6 inhibitor comprises a compound disclosed in International Application Publication No. WO 2010/020675 and U.S. Patent Nos.8,415,355 and 8,685,980, which are incorporated by reference in their entirety. 35 [227] In some embodiments, the CDK4/6 inhibitor comprises ribociclib (CAS Registry Number: 1211441-98-3). Ribociclib is also known as LEE011, KISQALI®, or 7-cyclopentyl-N,N- dimethyl-2-((5-(piperazin-1-yl)pyridin-2-yl)amino)-7H-pyrrolo[2,3-d]pyrimidine-6-carboxamide. [228] In some embodiments, the CDK4/6 inhibitor comprises abemaciclib (CAS Registry Number: 1231929-97-7). Abemaciclib is also known as LY835219 or N-[5-[(4-Ethyl-1-40 piperazinyl)methyl]-2-pyridinyl]-5-fluoro-4-[4-fluoro-2-methyl-1-(1-methylethyl)-1H-benzimidazol- PAT059646-WO-PCT 5 6-yl]-2-pyrimidinamine. Abemaciclib is a CDK inhibitor selective for CDK4 and CDK6 and is disclosed, e.g., in Torres-Guzman R et al. (2017) Oncotarget 10.18632/oncotarget.17778. [229] In some embodiments, the CDK4/6 inhibitor comprises palbociclib (CAS Registry Number: 571190-30-2). Palbociclib is also known as PD-0332991, IBRANCE® or 6-Acetyl-8- cyclopentyl-5-methyl-2-{[5-(1-piperazinyl)-2-pyridinyl]amino}pyrido[2,3-d]pyrimidin-7(8H)-one. 10 Palbociclib inhibits CDK4 with an IC50 of 11nM, and inhibits CDK6 with an IC50 of 16nM, and is disclosed, e.g., in Finn et al. (2009) Breast Cancer Research 11(5):R77. [230] In certain embodiments, a combination described herein comprises an inhibitor of chemokine (C-X-C motif) receptor 2 (CXCR2). In some embodiments, the CXCR2 inhibitor is chosen from 6-chloro-3-((3,4-dioxo-2-(pentan-3-ylamino)cyclobut-1-en-1-yl)amino)-2-hydroxy-N- 15 methoxy-N-methylbenzenesulfonamide, danirixin, reparixin, or navarixin. [231] In some embodiments, the CSF-1/1R binding agent is chosen from an inhibitor of macrophage colony-stimulating factor (M-CSF), e.g., a monoclonal antibody or Fab to M-CSF (e.g., MCS110), a CSF-1R tyrosine kinase inhibitor (e.g., 4-((2-(((1R,2R)-2- hydroxycyclohexyl)amino)benzo[d]thiazol-6-yl)oxy)-N-methylpicolinamide or BLZ945), a 20 receptor tyrosine kinase inhibitor (RTK) (e.g., pexidartinib), or an antibody targeting CSF-1R (e.g., emactuzumab or FPA008). In some embodiments, the CSF-1/1R inhibitor is BLZ945. In some embodiments, the CSF-1/1R binding agent is MCS110. In other embodiments, the CSF- 1/1R binding agent is pexidartinib. [232] In certain embodiments, a combination described herein comprises a c-MET 25 inhibitor. c-MET, a receptor tyrosine kinase overexpressed or mutated in many tumor cell types, plays key roles in tumor cell proliferation, survival, invasion, metastasis, and tumor angiogenesis. Inhibition of c-MET may induce cell death in tumor cells overexpressing c-MET protein or expressing constitutively activated c-MET protein. In some embodiments, the c-MET inhibitor is chosen from capmatinib (INC280), JNJ-3887605, AMG 337, LY2801653, 30 MSC2156119J, crizotinib, tivantinib, or golvatinib. [233] In certain embodiments, a combination described herein comprises a transforming growth factor beta (also known as TGF- TGF , TGFb, or TGF-beta, used interchangeably herein) inhibitor. In some embodiments, the TGF- inhibitor is chosen from fresolimumab or XOMA 089. 35 [234] In certain embodiments, a combination described herein comprises an adenosine A2a receptor (A2aR) antagonist (e.g., an inhibitor of A2aR pathway, e.g., an adenosine inhibitor, e.g., an inhibitor of A2aR or CD-73). In some embodiments, the A2aR antagonist is used in combination with a PD-1 inhibitor, and one or more (e.g., two, three, four, five, or all) of a CXCR2 inhibitor, a CSF-1/1R binding agent, LAG-3 inhibitor, a GITR agonist, a c-MET inhibitor, 40 or an IDO inhibitor. In some embodiments, the combination is used to treat a pancreatic cancer, a colorectal cancer, a gastric cancer, or a melanoma (e.g., a refractory melanoma). In some PAT059646-WO-PCT 5 embodiments, the A2aR antagonist is chosen from PBF509 (NIR178) (Palobiofarma/Novartis), CPI444/V81444 (Corvus/Genentech), AZD4635/HTL-1071 (AstraZeneca/Heptares), Vipadenant (Redox/Juno), GBV-2034 (Globavir), AB928 (Arcus Biosciences), Theophylline, Istradefylline (Kyowa Hakko Kogyo), Tozadenant/SYN-115 (Acorda), KW-6356 (Kyowa Hakko Kogyo), ST- 4206 (Leadiant Biosciences), or Preladenant/SCH 420814 (Merck/Schering). Without wishing to 10 be bound by theory, it is believed that in some embodiments, inhibition of A2aR leads to upregulation of IL-1b. [235] In certain embodiments, a combination described herein comprises an inhibitor of indoleamine 2,3-dioxygenase (IDO) and/or tryptophan 2,3-dioxygenase (TDO). In some embodiments, the IDO inhibitor is used in combination with a PD-1 inhibitor, and one or more 15 (e.g., two, three, four, or all) of a TGF- inhibitor, an A2aR antagonist, a CSF-1/1R binding agent, a c-MET inhibitor, or a GITR agonist. In some embodiments, the combination is used to treat a pancreatic cancer, a colorectal cancer, a gastric cancer, or a melanoma (e.g., a refractory melanoma). In some embodiments, the IDO inhibitor is chosen from (4E)-4-[(3- chloro-4-fluoroanilino)-nitrosomethylidene]-1,2,5-oxadiazol-3-amine (also known as epacadostat20 or INCB24360), indoximod (NLG8189), (1-methyl-D-tryptophan), -cyclohexyl-5H-Imidazo[5,1- a]isoindole-5-ethanol (also known as NLG919), indoximod, BMS-986205 (formerly F001287). [236] In certain embodiments, a combination described herein comprises a Galectin, e.g., Galectin-1 or Galectin-3, inhibitor. In some embodiments, the combination comprises a Galectin-1 inhibitor and a Galectin-3 inhibitor. In some embodiments, the combination 25 comprises a bispecific inhibitor (e.g., a bispecific antibody molecule) targeting both Galectin-1 and Galectin-3. In some embodiments, the Galectin inhibitor is used in combination with one or more therapeutic agents described herein. In some embodiments, the Galectin inhibitor is chosen from an anti-Galectin antibody molecule, GR-MD-02 (Galectin Therapeutics), Galectin- 3C (Mandal Med), Anginex, or OTX-008 (OncoEthix, Merck). 30 In some embodiments, a combination described herein comprises an inhibitor of the MAP kinase pathway including ERK inhibitors, MEK inhibitors and RAF inhibitors. [237] In some embodiments, a combination described herein comprises a MEK inhibitor. In some embodiments, the MEK inhibitor is chosen from Trametinib, selumetinib, AS703026, BIX 02189, BIX 02188, CI-1040, PD0325901, PD98059, U0126, XL-518, G-38963, or 35 G02443714. [238] In some embodiments, the MEK inhibitor is trametinib. Trametinib is also known as JTP-74057, TMT212, N-(3-{3-cyclopropyl-5-[(2-fluoro-4-iodophenyl)amino]-6,8-dimethyl-2,4,7- trioxo-3,4,6,7-tetrahydropyrido[4,3-d]pyrimidin-1(2H)-yl}phenyl)acetamide, or Mekinist (CAS Number 871700-17-3). 40 [239] In some embodiments, the MEK inhibitor comprises selumetinib which has the chemical name: (5-[(4-bromo-2-chlorophenyl)amino]-4-fluoro-N-(2-hydroxyethoxy)-1-methyl-1H- PAT059646-WO-PCT 5 benzimidazole-6-carboxamide. Selumetinib is also known as AZD6244 or ARRY 142886, e.g., as described in PCT Publication No. WO2003077914. [240] In some embodiments, the MEK inhibitor comprises AS703026, BIX 02189 or BIX 02188. [241] In some embodiments, the MEK inhibitor comprises 2-[(2-Chloro-4-iodophenyl)amino]-N- 10 (cyclopropylmethoxy)-3,4-difluoro-benzamide (also known as CI-1040 or PD184352), e.g., as described in PCT Publication No. WO2000035436). [242] In some embodiments, the MEK inhibitor comprises N-[(2R)-2,3-Dihydroxypropoxy]-3,4- difluoro-2-[(2-fluoro-4-iodophenyl)amino]- benzamide (also known as PD0325901), e.g., as described in PCT Publication No. WO2002006213). 15 [243] In some embodiments, the MEK inhibitor comprises 2’-amino-3’-methoxyflavone (also known as PD98059) which is available from Biaffin GmbH & Co., KG, Germany. [244] In some embodiments, the MEK inhibitor comprises 2,3-bis[amino[(2- aminophenyl)thio]methylene]-butanedinitrile (also known as U0126), e.g., as described in US Patent No.2,779,780). 20 [245] In some embodiments, the MEK inhibitor comprises XL-518 (also known as GDC-0973) which has a CAS No.1029872-29-4 and is available from ACC Corp. [246] In some embodiments, the MEK inhibitor comprises G-38963. [247] In some embodiments, the MEK inhibitor comprises G02443714 (also known as AS703206) 25 [248] Additional examples of MEK inhibitors are disclosed in WO 2013/019906, WO 03/077914, WO 2005/121142, WO 2007/04415, WO 2008/024725 and WO 2009/085983, the contents of which are incorporated herein by reference. Further examples of MEK inhibitors include, but are not limited to, 2,3-Bis[amino[(2-aminophenyl)thio]methylene]-butanedinitrile (also known as U0126 and described in US Patent No.2,779,780); (3S,4R,5Z,8S,9S,11E)-14-30 (Ethylamino)-8,9,16-trihydroxy-3,4-dimethyl-3,4,9, 19-tetrahydro-1H-2-benzoxacyclotetradecine- 1,7(8H)-dione] (also known as E6201, described in PCT Publication No. WO2003076424); vemurafenib (PLX-4032, CAS 918504-65-1); (R)-3-(2,3-Dihydroxypropyl)-6-fluoro-5-(2-fluoro-4- iodophenylamino)-8-methylpyrido[2,3-d]pyrimidine-4,7(3H,8H)-dione (TAK-733, CAS 1035555- 63-5); pimasertib (AS-703026, CAS 1204531-26-9); 2-(2-Fluoro-4-iodophenylamino)-N-(2-35 hydroxyethoxy)-1,5-dimethyl-6-oxo-1,6-dihydropyridine-3-carboxamide (AZD 8330); and 3,4- Difluoro-2-[(2-fluoro-4-iodophenyl)amino]-N-(2-hydroxyethoxy)-5-[(3-oxo-[1,2]oxazinan-2- yl)methyl]benzamide (CH 4987655 or Ro 4987655). [249] In some embodiments, a combination described herein comprises a RAF inhibitor. [250] RAF inhibitors include, but are not limited to, Vemurafenib (or Zelboraf®, PLX-4032, 40 CAS 918504-65-1), GDC-0879, PLX-4720 (available from Symansis), Dabrafenib (or PAT059646-WO-PCT 5 GSK2118436), LGX 818, CEP-32496, UI-152, RAF 265, Regorafenib (BAY 73-4506), CCT239065, or Sorafenib (or Sorafenib Tosylate, or Nexavar®). [251] In some embodiments, the RAF inhibitor is Dabrafenib. [252] In some embodiments, the RAF inhibitor is LXH254. [253] In some embodiments, a combination described herein comprises an ERK inhibitor. 10 [254] ERK inhibitors include, but are not limited to, LTT462, ulixertinib (BVD-523), LY3214996, GDC-0994, KO-947 and MK-8353. [255] In some embodiments, the ERK inhibitor is LTT462. LTT462 is 4-(3-amino-6- ((1S,3S,4S)-3-fluoro-4-hydroxy¬cyclohexyl)pyrazin-2-yl)-N-((S)-1-(3-bromo-5-fluorophenyl)-2- (methylamino)¬ethyl)-2-fluorobenzamide and is the compound of the following structure: 15 [256] The preparation of LTT462 is described in PCT patent application publication WO2015/066188. LTT462 is an inhibitor of extracellular signal-regulated kinases 1 and 2 (ERK 1/2). [257] In some embodiments, a combination described herein comprises a taxane, a vinca 20 alkaloid, a MEK inhibitor, an ERK inhibitor, or a RAF inhibitor. [258] In some embodiments, a combination described herein comprises at least two inhibitors selected, independently, from a MEK inhibitor, an ERK inhibitor, and a RAF inhibitor. [259] In some embodiments, a combination described herein comprises an anti-mitotic drug. [260] In some embodiments, a combination described herein comprises a taxane. 25 [261] Taxanes include, but are not limited to, docetaxel, paclitaxel, or cabazitaxel. In some embodiments, the taxane is docetaxel. [262] In some embodiments, a combination described herein comprises a vinca alkaloid. [263] Vinca alkaloids include, but are not limited to, vincristine, vinblastine, and leurosine. [264] In some embodiments, a combination described herein comprises a topoisomerase 30 inhibitor. [265] Topoisomerase inhibitors include, but are not limited to, topotecan, irinotecan, camptothecin, diflomotecan, lamellarin D, ellipticines, etoposide (VP-16), teniposide, doxorubicin, daunorubicin, mitoxantrone, amsacrine, aurintricarboxylic acid, and HU-331. PAT059646-WO-PCT 5 [266] In one embodiment, a combination described herein includes an interleukin-1 beta (IL- 1 ) inhibitor. In some embodiments, the IL-1 inhibitor is chosen from canakinumab, gevokizumab, Anakinra, or Rilonacept. [267] In certain embodiments, a combination described herein comprises an IL-15/IL-15Ra complex. In some embodiments, the IL-15/IL-15Ra complex is chosen from NIZ985 (Novartis), 10 ATL-803 (Altor) or CYP0150 (Cytune). [268] In certain embodiments, a combination described herein comprises a mouse double minute 2 homolog (MDM2) inhibitor. The human homolog of MDM2 is also known as HDM2. In some embodiments, an MDM2 inhibitor described herein is also known as a HDM2 inhibitor. In some embodiments, the MDM2 inhibitor is chosen from HDM201 or CGM097. 15 [269] In an embodiment the MDM2 inhibitor comprises (S)-1-(4-chlorophenyl)-7-isopropoxy-6- methoxy-2-(4-(methyl(((1r,4S)-4-(4-methyl-3-oxopiperazin-1- yl)cyclohexyl)methyl)amino)phenyl)-1,2-dihydroisoquinolin-3(4H)-one (also known as CGM097) or a compound disclosed in PCT Publication No. WO 2011/076786 to treat a disorder, e.g., a disorder described herein). In one embodiment, a therapeutic agent disclosed herein is used in 20 combination with CGM097. [270] In some embodiments, a combination described herein comprises a hypomethylating agent (HMA). In some embodiments, the HMA is chosen from decitabine or azacitidine. [271] In some embodiments, a combination described herein comprises a glucocorticoid. In some embodiments, the glucocorticoid is dexamethasone. 25 [272] In some embodiments, a combination described herein comprises asparaginase. [273] In certain embodiments, a combination described herein comprises an inhibitor acting on any pro-survival proteins of the Bcl2 family. In certain embodiments, a combination described herein comprises a Bcl-2 inhibitor. In some embodiments, the Bcl-2 inhibitor is venetoclax (also known 30 [274] In one embodiment, the Bcl-2 inhibitor is selected from the compounds described in WO 2013/110890 and WO 2015/011400. In some embodiments, the Bcl-2 inhibitor comprises navitoclax (ABT-263), ABT-737, BP1002, SPC2996, APG-1252, obatoclax mesylate (GX15- 070MS), PNT2258, Zn-d5, BGB-11417, or oblimersen (G3139). In some embodiments, the Bcl-35 2 inhibitor is N-(4-hydroxyphenyl)-3-[6-[(3S)-3-(morpholinomethyl)-3,4-dihydro-1H-isoquinoline- PAT059646-WO-PCT 5 2-carbonyl]-1,3-benzodioxol-5-yl]-N-phenyl-5,6,7,8-tetrahydroindolizine-1-carboxamide, compound A1: (compound A1). [275] In some embodiments, the Bcl-2 inhibitor is (S)-5-(5-chloro-2-(3-(morpholinomethyl)-10 1,2,3,4-tetrahydroisoquinoline-2-carbonyl)phenyl)-N-(5-cyano-1,2-dimethyl-1H-pyrrol-3-yl)-N-(4- hydroxyphenyl)-1,2-dimethyl-1H-pyrrole-3-carboxamide), compound A2: (compound A2). [276] In one embodiment, the panRAS inhibitors or combinations disclosed herein are suitable for the treatment of cancer in vivo. For example, the combination can be used to inhibit the 15 growth of cancerous tumors. The combination can also be used in combination with one or more of: a standard of care treatment (e.g., for cancers or infectious disorders), a vaccine (e.g., a therapeutic cancer vaccine), a cell therapy, a hormone therapy (e.g., with anti-estrogens or anti-androgens), a radiation therapy, surgery, or any other therapeutic agent or modality, to treat a disorder herein. For example, to achieve antigen-specific enhancement of immunity, the 20 combination can be administered together with an antigen of interest. A combination disclosed herein can be administered in either order or simultaneously. EXAMPLES 25 PAT059646-WO-PCT 5 [277] The following examples provide illustrative embodiments of the disclosure. One of ordinary skill in the art will recognize the numerous modifications and variations that may be performed without altering the spirit or scope of the disclosure. Such modifications and variations are encompassed within the scope of the disclosure. The examples provided do not in any way limit the disclosure. 10 Example 1. Synthesis and Characterization of panRAS Inhibitors, and Precursors thereof [278] Exemplary panRAS inhibitors and intermediates were synthesized using exemplary methods described in this example. 15 Materials, Methods & General Procedures: Compounds of the present disclosure may be prepared by methods known in the art of organic synthesis. In all of the methods it is understood that protecting groups for sensitive or reactive groups may be employed where necessary in accordance with general principles of chemistry. 20 Protecting groups are manipulated according to standard methods of organic synthesis (T.W. Green and P.G.M. Wuts (1999) Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons). These groups are removed at a convenient stage of the compound synthesis using methods that are readily apparent to those skilled in the art. Analytical Methods, Materials, and Instrumentation 25 Unless otherwise noted, reagents and solvents were used as received from commercial suppliers. Proton nuclear magnetic resonance (1H NMR) spectra were acquired on Bruker AVANCE 400 MHz, 500 MHz or 600 MHz NMR spectrometers using ICON-NMR, under TopSpin program control unless otherwise noted. Spectra were measured at 298 K, unless indicated otherwise, and 30 were referenced relative to the solvent resonance. Tetramethylsilane (TMS) was used as an internal standard. Chemical shifts are reported in ppm relative to dimethyl sulfoxide ( 2.50), methanol ( 3.31), chloroform ( 7.26) or other solvent as indicated in NMR spectral data. A small amount of the dry sample (2 to 5 mg) is dissolved in an appropriate deuterated solvent (1 mL). The chemical names were generated using ChemDraw Professional v22 from PerkinElmer. 35 Mass spectra were acquired on LC-MS, SFC-MS, or GC-MS systems using electrospray, chemical and electron impact ionization methods from a range of instruments of the following configurations: Waters Acquity UPLC/SQD system, using a photodiode array detector and a single quadrupole mass detector; Agilent 1200 systems with G 6110 series mass detector; Agilent 40 1290 Infinity II with DAD (photodiode array detector) and single quadrupole mass detector with ESI and APCI ionization (multi-mode);Waters AcQuity UPLC with PDA (photodiode array PAT059646-WO-PCT 5 detector), ELSD and single quadrupole mass detector with ESI ionization; Waters AutoPurification System with PDA (photodiode array detector) and single quadrupole mass detector with ESI ionization; [M+H]+ refers to protonated molecular ion of the chemical species; [M-H]- refers to molecular ion of the chemical species with loss of one proton; [M+Na]+ refers to molecular ion of the chemical species with addition of one sodium ion; [M-Boc+H]+ refers to protonated molecular 10 ion of the chemical species without a Boc protecting group; [M-tBu+2H]+ refers to protonated molecular ion of the chemical species without a tert-butyl group. Abbreviations Some abbreviations used in the examples are as follows: 1,1-bis(di-tert-butylphosphino)-15 ferrocenedichloropalladium (II) (PdCl2(dtpf) or Pd(dtpf)Cl2); 1,1-bis(diphenylphosphino)- ferrocenedichloropalladium (II) (PdCl2(dppf) or Pd(dppf)Cl2); 1,1-carbonyldiimidazole (CDI);(1- cyano-2-ethoxy-2-oxoethylidenaminooxy)dimethylamino-morpholino-carbenium hexafluorophosphate (COMU); 1-hydroxy-7-azabenzotriazole (HOAt); 2-(1H-7-azabenzotriazol- 1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU); 2,2'-bis-diphenylphospphanyl- 20 [1,1']binaphthalenyl (BINAP); 2-(2,5-dioxopyrrolidin-1-yl)-1,1,3,3-tetramethylisouronium tetrafluoroborate (TSTU); 4-dimethylaminopyridine (DMAP); 3-morpholinopropane-1-sulfonic acid (MOPS); (9,9-dimethyl-9H-xanthene-4,5-diyl)bis(diphenylphosphane) (xantphos); acetic acid (AcOH); acetic anhydride (Ac2O); acetonitrile (CH3CN); ammonium hydroxide (NH4OH); aqueous (aq.); atmosphere (atm.); back pressure regulator (BPR); broad (br); bromo-tris-25 pyrrolidino-phosphonium hexafluorophosphate (PyBroP); benzotriazole-1-yl-oxy-tris- (dimethylamino)-phosphonium hexafluorophosphate (BOP); cesium carbonate (Cs2CO3); doublet (d); 1,2-dichloroethane (DCE); dichloromethane (DCM); dicyclohexyl(2’,6’-dimethoxy[1,1’- biphenyl]-2-yl)phosphane (SPhos); dicyclohexyl[2 ,4 ,6 -tris(propan-2-yl)[1,1 -biphenyl]-2- yl]phosphane (XPhos); diethyl ether (Et2O); diisopropyl azodicarboxylate (DIAD); dimethyl 30 sulfoxide (DMSO); diphenylphosphoryl azide (DPPA); di-tert-butyl dicarbonate (Boc2O); equivalent(s) (equiv.); ethanol (EtOH); ethyl acetate (EtOAc); ethyl iodide (EtI); ethyl (E)-2-(((((9H- fluoren-9-yl)methoxy)carbonyl)oxy)imino)-2-cyanoacetate (Fmoc-Oxyma); fetal bovine serum (FBS); Förster resonance energy transfer (FRET); gram(s) (g); high performance liquid chromatography (HPLC); high-resolution mass spectrum (HRMS); homogeneous time-resolved 35 FRET (HTRF); hour(s) (hr); hydrochloric acid (HCl); inner diameter (I.D.); isopropanol (iPrOH); isopropylamine (iPr2NH); liquid chromatography coupled with mass spectrometry (LCMS); liter(s) (L); lithium aluminium hydride (LAH); lithium bis(trimethylsilyl)amide (LHMDS); lithium diisopropylamide (LDA); lithium bromide (LiBr); lithium hydroxide (LiOH); luminescence (LUM); magnesium sulfate (MgSO4); mass spectrum (MS); mass-to-charge ratio (m/z or M/Z); meta- 40 chloroperoxybenzoic acid (mCPBA); metabolism (MT); methanol (MeOH); methyl iodide (MeI); methyl magnesium bromide (MeMgBr); methyl tert-butyl ether (MTBE); 2-methyl tetrahydrofuran PAT059646-WO-PCT 5 (2-MeTHF); microwave (MW); microliter(s) (μL); micrometer(s) (μm); micromole(s) (μmol); milliliter(s) (mL); millimeter(s) (mm); millimole(s) (mmol); minute(s) (min); mole(s) (mol); multiplet (m); N-(3-dimethylaminopropyl)-N -ethylcarbodiimide hydrochloride (EDC); n-butyllithium (n- BuLi); N-chlorosuccinimide (NCS); N-hydroxy succinimide (NHS); N,N-diisopropylethylamine (DIPEA); N,N-dimethylformamide (DMF); N-methylpyrrolidone (NMP); isopropanol (iPrOH); 10 pentet (p); potassium hydroxide (KOH); potassium tert-butoxide (KOtBu); palladium(II) acetate (Pd(OAc)2); palladium on carbon (Pd/C); palladium hydroxide (Pd(OH)2); para-toluene sulfonic acid (PTSA); para-toluenesulfonyl chloride (TsCl); phosphate buffered saline (PBS); quartet (q); retention time (Rt); Roswell Park Memorial Institute medium (RPMI); room temperature (RT); saturated (sat.); singlet (s); second (sec.); sodium bicarbonate (NaHCO3); sodium borohydride 15 (NaBH4); sodium carbonate (Na2CO3); sodium hydride (NaH); sodium hydroxide (NaOH); sodium potassium-2,3-dihydroxybutane-1,4-dioate (Rochelle’s salt); sodium sulfate (Na2SO4); sodium thiosulfate (Na2S2O3); supercritical fluid chromatography (SFC); tert-butoxycarbonyl (Boc); tert- butyldimethylsilyl chloride (TBSCl); tetrabutylammonium fluoride (TBAF); tetrahydrofuran (THF); tetramethylammonium fluoride (TMAF); toluenesulfonylmethyl isocyanide (TosMIC); 20 triethylamine (NEt3); triethylsilane (Et3SiH); trifluoroacetic acid (TFA); triisopropylsilyl chloride (TIPS-Cl); trimethylaluminum (AlMe3); trimethylsilyl trifluoromethanesulfonate (TMSOTf); triplet (t); tris(2-carboxyethyl)phosphine (TCEP); tris(dibenzylidene)dipalladium(0) (Pd2dba3); tri-tert- butylphosphonium tetrafluoroborate (TTBP-HBF4); thionyl chloride (SOCl2); trimethylsilyl chloride (TMSCl); weight (wt.); silica gel, silica (SiO2), silver(I) oxide (Ag2O); silver(I)25 trifluoromethanesulfonate (AgOTf); para-toluenesulfonyl 1,2-diphenyl-1,2-ethylenediamine (Ts- DPEN). LCMS CONDITIONS RNXMON-basic (Acquity BEH C18; particle size: 1.7 μm; column size: 2.1 x 30mm; eluent A: H2O 30 + 5 mM ammonium hydroxide; eluent B: CH3CN + 5 mM ammonium hydroxide; eluent: 2 to 98% B in 2.00 min; flow rate: 1.0 mL/min; column temperature: 50°C.) RXNMON-Acidic (Acquity BEH C18; particle size: 1.7 μm; column size: 2.1 x 50mm; eluent A: H2O + 0.1% formic acid; eluent B: CH3CN + 0.1% formic acid; eluent: 2 to 98% B in 2.00 min; flow rate: 1.0 mL/min; column temperature: 50°C.) 35 RXNMON-TFA (Acquity BEH C18; particle size: 1.7 μm; column size: 2.1 x 50mm; eluent A: H2O + 0.05% trifluoroacetic acid; eluent B: CH3CN + 0.05% trifluoroacetic acid; eluent: 2 to 98% B in 2.00 min; flow rate: 1.0 mL/min; column temperature: 50°C.) 1 PAT059646-WO-PCT 5 FinalAnalysis-Basic (Acquity BEH C18; particle size: 1.7 μm; column size: 2.1 x 30mm; eluent A: H2O + 5 mM ammonium hydroxide; eluent B: CH3CN + 5 mM ammonium hydroxide; eluent: 2 to 98% B in 5.00 min; flow rate: 1.0 mL/min; column temperature: 50°C.) FinalAnalysis-Acidic (Acquity BEH C18; particle size: 1.7 μm; column size: 2.1 x 50mm; eluent A: H2O + 0.1% formic acid; eluent B: CH3CN + 0.1% formic acid; eluent: 2 to 98% B in 5.00 min; flow 10 rate: 1.0 mL/min; column temperature: 50°C.) FinalAnalysis-TFA (Acquity BEH C18; particle size: 1.7 μm; column size: 2.1 x 50mm; eluent A: H2O + 0.05% trifluoroacetic acid; eluent B: CH3CN + 0.05% trifluoroacetic acid; eluent: 2 to 98% B in 5.00 min; flow rate: 1.0 mL/min; column temperature: 50°C.) FinalAnalysis-Acidic-NonPolar (Acquity BEH C18; particle size: 1.7 μm; column size: 2.1 x 50mm; 15 eluent A: H2O + 0.1% formic acid; eluent B: CH3CN + 0.1% formic acid; eluent: 40 to 98% B in 2.00 min; flow rate: 1.0 mL/min; column temperature: 50°C.) FinalAnalysis-Basic-NonPolar (Acquity BEH C18; particle size: 1.7 μm; column size: 2.1 x 30mm; eluent A: H2O + 5 mM ammonium hydroxide; eluent B: CH3CN + 5 mM ammonium hydroxide; eluent: 40 to 98% B in 2.00 min; flow rate: 1.0 mL/min; column temperature: 50°C.) 20 Peptide Method (Acquity CSH C18; particle size: 1.7 μm; column size: 2.1 x 50mm; eluent A: H2O + 0.05% trifluoroacetic acid; eluent B: CH3CN + 0.05% trifluoroacetic acid; eluent: 2 to 98% B in 5.00 min; flow rate: 1.0 mL/min; column temperature: 50°C.); Unless otherwise noted, HRMS was measured by Peptide Method with a MS detector. HPLC-quant.: (Zorbax Eclipse XDB-C18; particle size: 1.8 m; column size: 4.6 mm x 50 mm; 25 eluent A: H2O + 0.05% trifluoroacetic acid; eluent B: CH3CN + 0.05% trifluoroacetic acid; eluent: 5 to 100% B in 6.00 min; flow rate: 1.0 mL/min; column temperature: 35°C; detection at 215 nm) Final_Analysis_2min (Aquity CORTECS C18; particle size: 2.7 μm; column size: 2.1 x 50mm; eluent A: H2O + 0.05% formic acid + 3.75 mM ammonium acetate; eluent B: 2-PrOH + 0.05% formic acid; eluent: 5 to 50% B in 1.40 min, 50 to 98% B in 0.3 min; flow rate: 1.0 mL/min; column 30 temperature: 80°C) HRMS_2min (Acquity CSH C18; particle size: 1.7 μm; column size: 2.1 x 50mm; eluent A: H2O + 0.05% trifluoroacetic acid; eluent B: CH3CN + 0.05% trifluoroacetic acid; eluent: 2 to 98% B in 2.00 min; flow rate: 1.0 mL/min; column temperature: 50°C) Prep_SFC1 (Column: Chiralcel OX-H 21x250mm 5um, Flow Rate: 100 g per minute, Cosolvent: 35 15% 4:1 heptane:isopropanol in CO2, detection: 211nm, back pressure regulator: 125 bar, Injection Size: 24.0mg (30.0 mg/mL in 4:1 Hp/IPA)) PAT059646-WO-PCT 5 Prep_SFC2 (2 x CHIRALPAK IG; particle size: 5 μm; column size: 30 x 250mm; modifier: 14% EtOH + 0.05% NH3; flow rate: 95.0 mL/min; duration: 2.2 min; preheater temperature: 40°C; back pressure regulator: 100 bar; detection wavelength: 214 nm) Prep_SFC3 (ChiralPak IG; particle size: 5 μm; flow rate: 80 mL/min; column size: 21 x 250 mm; modifier: 20% MeOH in CO2; detection wavelength: 215 nm; back pressure regulator: 125 bar) 10 Prep_SFC4 (Chiralcel OJ-H; particle size: 5 μm; flow rate: 85 mL/min; column size: 30 x 150 mm; modifier: 15% CH3OH in CO2; pre-heater temperature: 35 °C; detection wavelength: 220 nm; back pressure regulator: 100 bar) Prep_SFC5 (ChiralPak AD-H; particle size: 5 μm; flow rate: 45 mL/min; column size: 20 x 250 mm; modifier: 15% CH3OH in CO2; pre-heater temperature: 35 °C; detection wavelength: 220 nm; 15 back pressure regulator: 100 bar) Prep_SFC6 (ChiralPak IC; particle size: 5 μm; flow rate: 80 mL/min; column size: 21 x 250 mm; modifier: 15% MeOH in CO2; detection wavelength: 220 nm; back pressure regulator: 125 bar) Prep_SFC7 (ChiralPak IG; particle size: 4 μm; flow rate: 150 g/min; column size: 30 x 250 mm; modifier: 12% MeOH in CO2; detection wavelength: 215 nm; back pressure regulator: 125 bar) 20 Prep_SFC8 (Waters diol; particle size: 5 μm; flow rate: 120 g/min; column size: 30 x 250 mm; modifier: 30% MeOH containing 10 mM NH3 in CO2; detection wavelength: 261 nm; back pressure regulator: 125 bar) Chiral_SFC1 (CHIRALPAK IG; particle size: 5 μm; column size: 4.6 x 100mm; modifier: 8% EtOH + NH3; flow rate: 3.0 mL/min; column temperature: 40°C; backpressure: 1800 psi) 25 Chiral_UPLC1 (OJ-RH; particle size: 5 μm; column size: 4.6 x 150mm; eluent 60% MeOH + 0.05% FA; flow rate: 0.5 mL/min; detection wavelength: 210 nm) Intermediate 1 3,4-di-tert-butyl 2-methyl (1S,2S,6R)-3,4-diazabicyclo[4.1.0]heptane-2,3,4-tricarboxylate 30 PAT059646-WO-PCT 5 Procedure Step a: To a stirred solution of di-tert-butyl (E)-diazene-1,2-dicarboxylate (8.00 g) in toluene (10 mL) was added methyl (E)-penta-2,4-dienoate (4.68 g). The resulting mixture was stirred at 80°C for 36 hr. The reaction mixture was purified directly by column chromatography (SiO2, 0 to 100% 10 EtOAc in heptane) to afford 1,2-di-tert-butyl 3-methyl 3,6-dihydropyridazine-1,2,3-tricarboxylate (7.70 g) as a white solid. LC/MS (RXNMON-Acidic method): M/Z = 343 [M+H]+.1H NMR (400 MHz, CDCl3) 5.93 (d, J = 2.9 Hz, 2H), 5.32 (s, 1H), 4.49 - 4.27 (m, 1H), 3.74 (s, 3H), 3.72 - 3.56 (m, 1H), 1.48 (s, 18H). 15 Step b: To a stirred solution of 1,2-di-tert-butyl 3-methyl 3,6-dihydropyridazine-1,2,3-tricarboxylate (7.50 g) in THF (20 mL) under a nitrogen atmosphere was added lithium borohydride (2 M in THF, 32.9 mL) dropwise. The resulting mixture was stirred at 0°C for 2 hr then warmed to RT and stirred for 1 hr. The reaction mixture was quenched by addition of sat. NaHCO3 solution (20 mL), then diluted with EtOAc. The layers were separated, and the aq. layer was extracted with EtOAc (3 x 20 20 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure. The crude material was purified by column chromatography (SiO2, 0 to 100% EtOAc in heptane) to afford di-tert-butyl 3-(hydroxymethyl)-3,6- 114 PAT059646-WO-PCT 5 dihydropyridazine-1,2-dicarboxylate (5.30 g) as a colorless oil. LC/MS (RXNMON-Acidic method): M/Z = 315 [M+H]+.1H NMR (400 MHz, CDCl3) 5.87 – 5.67 (m, 2H), 4.69 (s, 1H), 4.40 – 4.18 (m, 1H), 3.97 – 3.26 (m, 3H), 2.68 (s, 1H), 1.53 – 1.45 (m, 18H). Step c: To a stirred solution of di-tert-butyl 3-(hydroxymethyl)-3,6-dihydropyridazine-1,2- 10 dicarboxylate (5.30 g), DMAP (206 mg), and triethylamine (1.88 g) in DCM (30 mL) was added tert-butylchlorodiphenylsilane (4.63 g). The resulting mixture was stirred at 0°C for 4 hr. The reaction mixture was concentrated, and the resulting residue was taken up in minimal acetonitrile. The crude mixture was purified by column chromatography (C18, 40 to 100% acetonitrile in water + 0.1% formic acid), followed by lyophilization, to afford di-tert-butyl 3-(((tert- 15 butyldiphenylsilyl)oxy)methyl)-3,6-dihydropyridazine-1,2-dicarboxylate (7.10 g) as a colorless oil. LC/MS (RXNMON-Acidic-NonPolar method): M/Z = 575 [M+Na]+.1H NMR (400 MHz, CDCl3) 7.69 - 7.62 (m, 4H), 7.45 - 7.32 (m, 6H), 6.08 - 5.72 (m, 2H), 4.67 (d, J = 84.8 Hz, 1H), 4.42 - 4.14 (m, 1H), 4.03 - 3.50 (m, 3H), 1.45 (s, 9H), 1.27 (d, J = 5.5 Hz, 9H), 1.06 (s, 9H). 20 Step d: To a vigorously stirred solution of di-tert-butyl 3-(((tert-butyldiphenylsilyl)oxy)methyl)-3,6- dihydropyridazine-1,2-dicarboxylate (150 mg) and benzyltriethylammonium chloride (2.2 mg) in DCM (0.50 mL) under a nitrogen atmosphere was added potassium hydroxide (8.0 M solution in water, 850 μL). The resulting mixture was cooled to 0°C, and bromoform (stabilized with ethanol, 1.71 g) was added dropwise over 30 min. using a syringe pump. The reaction mixture was stirred 25 at 0°C for 2 hr, then allowed to warm to RT and stirred for 16 hr. The reaction was quenched by addition of water (2 mL) and DCM (10 mL), then passed through a phase separator, washing with DCM. The resulting organic layer was concentrated under reduced pressure and the crude mixture was purified by column chromatography (SiO2, 0 to 50% EtOAc in heptane) to afford di- tert-butyl 7,7-dibromo-2-(((tert-butyldiphenylsilyl)oxy)methyl)-3,4-diazabicyclo[4.1.0]heptane-3,4- 30 dicarboxylate (130mg) as a colorless oil. LC/MS (RXNMON-Acidic-NonPolar method): M/Z = 747 [M+Na]+.1H NMR (400 MHz, CDCl3) 7.69 - 7.62 (m, 4H), 7.45 - 7.36 (m, 6H), 4.65 - 4.49 (m, 1H), 4.47 - 4.21 (m, 1H), 4.06 - 3.80 (m, 1H), 3.57 - 3.40 (m, 1H), 3.06 - 2.88 (m, 1H), 2.31 - 2.10 (m, 2H), 1.37 (s, 9H), 1.18 (s, 9H), 1.10 (s, 9H). 35 Step e: To a stirred solution of di-tert-butyl 7,7-dibromo-2-(((tert-butyldiphenylsilyl)oxy)methyl)- 3,4-diazabicyclo[4.1.0]heptane-3,4-dicarboxylate (2.80 g) in toluene (20 mL) was added tributyltin hydride (5.62 g) and AIBN (317 mg). The resulting solution was purged with nitrogen gas and then stirred at 80°C for 2 hr. The reaction mixture was cooled to 0°C and diluted with EtOAc (20 mL), then quenched with 10% aq. potassium fluoride solution (10 mL), resulting in a fine white 40 precipitate. The resulting mixture was filtered over Celite®, and the layers of the filtrate were separated. The aq. layer was extracted with EtOAc (3 x 20 mL), and the combined organic layers PAT059646-WO-PCT 5 were washed with brine (20 mL), dried over Na2SO4, and concentrated to afford a yellow oil, which crystallized under high vacuum. The crystals were washed with deionized water and ice-cold methanol, then dried under reduced pressure to afford di-tert-butyl 2-(((tert- butyldiphenylsilyl)oxy)methyl)-3,4-diazabicyclo[4.1.0]heptane-3,4-dicarboxylate (2.10 g) as colorless crystals. LC/MS (RXNMON-Acidic-NonPolar method): M/Z = 589 [M+Na]+.1H NMR (400 10 MHz, CDCl3) 7.71 – 7.63 (m, 4H), 7.46 – 7.33 (m, 6H), 4.62 – 4.33 (m, 1H), 4.26 – 3.99 (m, 1H), 3.94 – 3.87 (m, 1H), 3.69 – 3.53 (m, 1H), 3.19 – 3.01 (m, 1H), 1.42 (s, 9H), 1.35 – 1.32 (m, 2H), 1.23 – 1.17 (m, 9H), 1.08 (s, 9H), 0.87 – 0.81 (m, 1H), 0.19 – 0.08 (m, 1H). Step f: To a stirred solution of di-tert-butyl 2-(((tert-butyldiphenylsilyl)oxy)methyl)-3,4- 15 diazabicyclo[4.1.0]heptane-3,4-dicarboxylate (2.10 g) in THF (10 mL) at 0°C was added TBAF (1.0 M in THF, 5.6 mL) and the resulting solution was stirred at 0°C for 1 hr. The reaction solution was then allowed to warm to RT and stir for 5 hr. The reaction was concentrated under reduced pressure and the crude material was purified by column chromatography (SiO2, 0 to 100% EtOAc in heptane) to afford di-tert-butyl 2-(hydroxymethyl)-3,4-diazabicyclo[4.1.0]heptane-3,4- 20 dicarboxylate (1.05 g) as a colorless oil. LC/MS (RXNMON-Acidic method): M/Z = 329 [M+H]+.1H NMR (400 MHz, CDCl3) 4.67 – 4.39 (m, 1H), 4.27 – 4.07 (m, 1H), 3.76 – 3.45 (m, 2H), 3.42 – 3.09 (m, 1H), 2.79 – 2.53 (m, 1H), 1.54 – 1.42 (m, 18H), 1.24 – 1.11 (m, 1H), 0.85 – 0.76 (m, 1H), 0.69 – 0.59 (m, 1H), 0.28 – 0.09 (m, 1H). 25 Step g: To a stirred solution of di-tert-butyl 2-(hydroxymethyl)-3,4-diazabicyclo[4.1.0]heptane-3,4- dicarboxylate (1.05 g) in acetonitrile (10 mL) at RT under a nitrogen atmosphere was added TEMPO (30.0 mg), sodium chlorite (578 mg), and pH 7 phosphate buffer (10 mL). Vigorous stirring produced a pale-yellow emulsion, to which was added sodium hypochlorite (0.74 M aq. solution, 260 L) dropwise. The resulting deep-red mixture was stirred at RT for 4 hr, then cooled 30 to 0°C, and quenched with sat. aq. sodium thiosulfate solution (10 mL), then acidified by addition of sat. citric acid solution (10 mL). The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine, dried over Na2SO4, and concentrated under reduced pressure. The resulting residue was dissolved in N,N-dimethylformamide (20 mL) under a nitrogen atmosphere, and to this solution was added cesium carbonate (1.26 g) and 35 iodomethane (684 mg). The resulting mixture was stirred at RT for 24 hr, then diluted with water (30 mL) and EtOAc (20 mL). The layers were separated, and the aq. layer was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine, dried over Na2SO4, and concentrated under reduced pressure. The resulting yellow oil was purified by column chromatography (C18, 0 to 100% acetonitrile in water + 0.1% formic acid) to afford 3,4-di-tert- 40 butyl 2-methyl 3,4-diazabicyclo[4.1.0]heptane-2,3,4-tricarboxylate (870 mg) as a yellow oil. LC/MS (RXNMON-Acidic method): M/Z = 379 [M+Na]+.1H NMR (400 MHz, CDCl3) 5.11 (s, 1H), PAT059646-WO-PCT 5 4.32 - 4.21 (m, 1H), 3.76 (s, 3H), 3.25 - 3.08 (m, 1H), 1.45 (s, 18H), 1.39 - 1.33 (m, 2H), 0.89 - 0.83 (m, 1H), 0.14 - 0.06 (m, 1H). Step h: Racemic 3,4-di-tert-butyl 2-methyl 3,4-diazabicyclo[4.1.0]heptane-2,3,4-tricarboxylate (670 mg) was purified by chiral SFC (Prep_SFC1 method) to give 3,4-di-tert-butyl 2-methyl 10 (1S,2S,6R)-3,4-diazabicyclo[4.1.0]heptane-2,3,4-tricarboxylate (260 mg) as the first eluting isomer. Intermediate 2 Note: axial chirality of compounds is shown in below scheme. 15 benzyl (S)-4-(5-(5-bromo-1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-1H-indol-2-yl)-6-(1- methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate
PAT059646-WO-PCT 5 Procedure Step a: To a mixture of 3-bromo-5-fluoropicolinonitrile (910 g) in toluene (9.1 L) was added MeMgBr (3 M in 2-MeTHF, 1.96 L) dropwise at 5°C. The mixture was stirred at 5°C for 1 hr. The 10 reaction mixture was poured into 10% aq. HCl solution (4.55 L) and stirred for 30 min. at 10°C. The reaction mixture was partitioned between water and toluene and the aq. phase was extracted with toluene (4.55 L). The combined organic layers were washed with brine (4.55 L) and concentrated under reduced pressure at 60°C to yield the desired product as an oil (890 g), which was used in the next step without further purification. 15 PAT059646-WO-PCT 5 Step b: To a mixture of 1-(3-bromo-5-fluoropyridin-2-yl)ethan-1-one (855 g) in DMF (8.55 L) was added benzyl piperazine-1-carboxylate (867 g) and K2CO3 (742 g). The reaction was stirred at 50°C for 16 hr. The reaction mixture was poured into ice/water (17 L) and stirred at RT for 3 hr. The mixture was filtered and rinsed with water. The filter cake was treated with MTBE at RT for 16 hr, filtered, and rinsed with MTBE (428 mL) to yield the desired product as a light-yellow solid 10 (1387 g).1H NMR (300 MHz, DMSO-d6) 8.37 (d, J = 2.6 Hz, 1H), 7.54 (d, J = 2.5 Hz, 1H), 7.45 – 7.29 (m, 5H), 5.12 (s, 2H), 3.61 – 3.41 (m, 8H), 2.54 (s, 3H). Step c: To a mixture of benzyl 4-(6-acetyl-5-bromopyridin-3-yl)piperazine-1-carboxylate (533 g) in DCM (5.0 L) was added DIEA (463 g) and formic acid (275 g). The headspace of the flask was 15 evacuated and backfilled with nitrogen and then RuCl (p-cymene) [(S,S)-Ts-DPEN] (37.7 g) was added. The reaction was stirred at 35°C for 16 hr. The reaction mixture was cooled down to 5°C and water (2.5 L) was added. The resulting mixture was stirred for 30 min. at 25°C. The reaction mixture was partitioned between water and DCM; the organic layer was washed with 5% NaCl (2.5 L) and concentrated under reduced pressure at 40°C. The residue was purified by column 20 chromatography (SiO2, 33% EtOAc in heptane) to afford the desired product as a brown solid (484 g).1H NMR (300 MHz, DMSO-d6) 8.30 (d, J = 2.5 Hz, 1H), 7.54 (d, J = 2.5 Hz, 1H), 7.43 – 7.28 (m, 5H), 5.10 (d, J = 6.4 Hz, 2H), 5.05 – 4.89 (m, 2H), 3.54 (s, 4H), 3.30 – 3.17 (m, 4H), 1.34 (d, J = 6.2 Hz, 3H). 25 Step d: To a mixture of benzyl (S)-4-(5-bromo-6-(1-hydroxyethyl)pyridin-3-yl)piperazine-1- carboxylate (400.0 g) in DMF (3.6 L) was added t-BuOLi (105.0 g) at 25°C. Next, MeI (248 g) was dropwise to the mixture. The resulting reaction mixture was stirred at 25°C for 16. To the reaction mixture was added water (1.8 L) and stirred 25°C for 2 hr, which was filtered and rinsed with water (360 mL). The filter cake was treated with MTBE (1.5 L) and heptane (735 mL) at 25°C for 16 hr. 30 Filtration gave the desired product as a brown solid (253 g).1H NMR (400 MHz, DMSO-d6) 8.33 (d, J = 2.5 Hz, 1H), 7.53 (d, J = 2.5 Hz, 1H), 7.44 – 7.29 (m, 5H), 5.11 (s, 2H), 4.73 (q, J = 6.4 Hz, 1H), 3.57 (d, J = 25.5 Hz, 4H), 3.30 – 3.22 (m, 4H), 3.11 (d, J = 9.7 Hz, 3H), 1.36 (d, J = 6.4 Hz, 3H). 35 Step e: A mixture of benzyl (S)-4-(5-bromo-6-(1-methoxyethyl)pyridin-3-yl)piperazine-1- carboxylate (132 g), bis(pinacolato)diboron (108 g), Pd(dppf)Cl2 (22.2 g), KOAc (74.5 g) in toluene (1.2 L) was degassed and purged with nitrogen (3x) and stirred at 100°C for 10 hr. The reaction mixture was concentrated then redissolved in EtOAc (600 mL). Sulfhydryl silica gel (150 g) was added, and the mixture was stirred at 25°C for 30 min. The reaction mixture was filtered, washed40 by EtOAc (3 x 300 mL), and concentrated to yield crude benzyl (S)-4-(6-(1-methoxyethyl)-5- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-3-yl)piperazine-1-carboxylate (227 g) as a PAT059646-WO-PCT 5 black brown oil. LC/MS (FinalAnalysis-Basic method): M/Z = 400 [M-C6H10+H]+. The product was used in the next step without further purification. Step f: To a solution of 5-bromo-3-(3-((tert-butyldiphenylsilyl)oxy)-2,2-dimethylpropyl)-2-iodo-1H- indole (141 g)) and benzyl (S)-4-(6-(1-methoxyethyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- 10 yl)pyridin-3-yl)piperazine-1-carboxylate (157 g) in dioxane (600 mL), water (200 mL) and toluene (200 mL) was added K3PO4 (115 g) and Pd(dppf)Cl2 (15.9 g). The mixture was sparged with nitrogen and stirred at 80°C for 12 hr under a nitrogen atmosphere. The reaction mixture was concentrated and purified by column chromatography (SiO2, 0 to 10% methanol in DCM). The residue was triturated with MTBE at RT for 12 hr. The desired product (66.3 g) was obtained as15 an off-white solid. for 12 hr. The mixture was filtered, concentrated and dried to yield (S)-4-(5-(5- bromo-3-(3-((tert-butyldiphenylsilyl)oxy)-2,2-dimethylpropyl)-1H-indol-2-yl)-6-(1- methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (66.3 g) as an off-white solid. 1H NMR (400 MHz, DMSO-d6) = 11.30 (s, 1H), 8.50 - 8.38 (m, 1H), 7.85 - 7.66 (m, 1H), 7.52 (br d, J = 6.4 Hz, 4H), 7.43 - 7.39 (m, 4H), 7.38 - 7.36 (m, 6H), 7.30 (br d, J = 8.4 Hz, 2H), 7.25 - 7.23 (m, 1H), 7.20 20 (dd, J = 1.6, 8.6 Hz, 1H), 5.12 - 5.10 (m, 2H), 4.12 - 4.05 (m, 1H), 3.51 (br s, 4H), 3.32 - 3.20 (m, 3H), 3.20 - 3.14 (m, 4H), 2.86 (br s, 2H), 1.33 - 1.29 (m, 3H), 1.12 - 1.00 (m, 2H), 0.98 - 0.93 (m, 9H), 0.70 - 0.59 (m, 6H) Step g: To a solution of benzyl (S)-4-(5-(5-bromo-3-(3-((tert-butyldiphenylsilyl)oxy)-2,2- 25 dimethylpropyl)-1H-indol-2-yl)-6-(1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (73.0 g) in DMF (730 mL) was added Cs2CO3 (81.6 g) and EtI (26.0 g) at 0°C. The reaction mixture stirred at 25°C for 12 hr. The reaction was stirred at 30°C for 4 hr. The resulting mixture was diluted with EtOAc (800 mL) and washed with brine (3 x 800 mL). The organic layer was dried over anhydrous Na2SO4 and concentrated under reduced pressure to yield benzyl (S)-4-(5-(5-bromo-3-(3-((tert-30 butyldiphenylsilyl)oxy)-2,2-dimethylpropyl)-1-ethyl-1H-indol-2-yl)-6-(1-methoxyethyl)pyridin-3- yl)piperazine-1-carboxylate (83.9 g) as a brown oil. Step h: A mixture of benzyl (S)-4-(5-(5-bromo-3-(3-((tert-butyldiphenylsilyl)oxy)-2,2- dimethylpropyl)-1-ethyl-1H-indol-2-yl)-6-(1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate 35 (83.9 g) and CsF (42.3 g) in DMF (800 mL) was evacuated and backfilled with nitrogen (3x). The reaction was stirred at 66°C for 24 hr. The reaction was cooled to 25°C and to it was added CsF (42.3 g) and it was stirred at 66°C for 12 hr. The resulting mixture was diluted with EtOAc (1 L) and washed with brine (3 x 1.0 L). Then the organic phase was concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, 0 to 100% EtOAc in40 petroleum ether) twice to yield benzyl (S)-4-(5-(5-bromo-1-ethyl-3-(3-hydroxy-2,2- PAT059646-WO-PCT 5 dimethylpropyl)-1H-indol-2-yl)-6-(1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (axial chirality as shown in structure) (20.2 g) as an off-white solid. m/z = 665 [M+3H]+ Intermediate 3 and Intermediate 3a Note: axial chirality of compounds is as shown in below scheme. 10 (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)-5- ((triisopropylsilyl)oxy)phenyl)propanoic acid Procedure 15 Step a: A suspension of Intermediate 2 (369 mg) and methyl (S)-2-((tert-butoxycarbonyl)amino)- 3-(3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5-((triisopropylsilyl)oxy)phenyl)propanoate (482 mg) was prepared under a nitrogen atmosphere in 5 wt% aq. Tween80:lecithin (8:2 w:w ratio, 11 mL) and isopropanol (5.5 mL) was added. The resulting solution was degassed for 5 min. via sparging with nitrogen.1,1'-Bis (di-t-butylphosphino)ferrocene palladium dichloride (18.1 20 mg) and triethylamine (282 mg) were sequentially charged to the degassed mixture and the solution was stirred at 0°C for 1 hr. Additional methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5-((triisopropylsilyl)oxy)phenyl)propanoate (161 mg) was charged to the mixture after 1 hr and stirring at 80°C continued for an additional 30 min. The reaction mixture was extracted with EtOAc (3 x 15.0 mL), and the organic extracts were 25 passed through an phase separator. The filtrate was concentrated and the remaining residue was purified by column chromatography (SiO2, 0 to 100% (3:1 EtOAc/ethanol) in heptane) to afford benzyl 4-(5-(5-(3-((S)-2-((tert-butoxycarbonyl)amino)-3-methoxy-3-oxopropyl)-5- ((triisopropylsilyl)oxy)phenyl)-1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-1H-indol-2-yl)-6-((S)-1- methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (570 mg) as a tan foam. LC/MS 30 (FinalAnalysis-Basic method): M/Z = 1035 [M+H]+. PAT059646-WO-PCT 5 Step b: To a stirred solution of benzyl 4-(5-(5-(3-((S)-2-((tert-butoxycarbonyl)amino)-3-methoxy- 3-oxopropyl)-5-((triisopropylsilyl)oxy)phenyl)-1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-1H-indol- 2-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (621 mg) in methanol (8.0 mL) under nitrogen was added Pd(OH)2 on carbon (82.8 mg, 10% wt.). The reaction vessel was 10 evacuated and backfilled with hydrogen. The reaction stirred at RT for 3 h. The reaction mixture was filtered under nitrogen by washing with MeOH, and then concentrated to afford methyl (S)-2- ((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1- methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)-5- ((triisopropylsilyl)oxy)phenyl)propanoate (475 mg) as an orange foam. LC/MS (FinalAnalysis- 15 Basic method): M/Z = 901 [M+H]+. Step c: To a stirred solution of methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3- hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)-1H-indol-5- yl)-5-((triisopropylsilyl)oxy)phenyl)propanoate (475 mg) in isopropanol (5.0 mL) under nitrogen 20 was charged aq. formaldehyde (393 μL, 37 wt%). Sodium triacetoxyborohydride (335 mg) was added and the resulting suspension stirred at RT for 2 hr. The reaction mixture was concentrated to 1/3 of initial volume, diluted with DCM (10 mL), quenched with sat. NaHCO3 (5 mL), filtered through a phase separator, and concentrated. The crude material was purified by column chromatography (C18, 20 to 70% acetonitrile in water + 0.1% formic acid), followed by25 lyophilization to afford methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)- 5-((triisopropylsilyl)oxy)phenyl)propanoate (393 mg) as a white solid. LC/MS (Peptide method): M/Z = 915 [M+H]+.1H NMR (400 MHz, DMSO-d6) 8.46 – 8.40 (m, 3H), 7.83 (d, J = 1.7 Hz, 1H), 7.55 (d, J = 8.5 Hz, 1H), 7.41 (dd, J = 8.5, 1.7 Hz, 1H), 7.29 (d, J = 8.1 Hz, 1H), 7.24 (d, J = 2.9 30 Hz, 1H), 7.14 (s, 1H), 6.98 (d, J = 1.9 Hz, 1H), 6.73 (t, J = 1.8 Hz, 1H), 4.43 (s, 1H), 4.28 – 4.19 (m, 1H), 4.04 (m, 2H), 3.86 (m, 1H), 3.63 (s, 3H), 3.25 (d, J = 4.4 Hz, 3H), 3.10 – 2.95 (m, 3H), 2.93 – 2.88 (m, 1H), 2.85 (s, 3H), 2.71 – 2.65 (m, 1H), 2.45 (t, J = 4.9 Hz, 3H), 2.21 (s, 3H), 1.35 (d, J = 6.2 Hz, 3H), 1.32 (s, 9H), 1.27 (dd, J = 8.9, 5.9 Hz, 4H), 1.15 (t, J = 7.1 Hz, 3H), 1.10 (d, J = 7.4 Hz, 18H), 0.68 (s, 3H), 0.61 (s, 3H). 35 Step d: To a solution of methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)- 5-((triisopropylsilyl)oxy)phenyl)propanoate (393 mg) in DCE (10 mL) was added trimethyltin hydroxide (389 mg) under a nitrogen atmosphere. The resulting suspension was stirred at 70°C 40 for 4 hr. The reaction mixture was cooled to RT, diluted with DCM (10 mL), and quenched with aq. sodium hydrogen sulfate (0.05M, 4.6 mL). The organic layer was then passed through a phase PAT059646-WO-PCT 5 separator and concentrated to afford (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3- hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)- 1H-indol-5-yl)-5-((triisopropylsilyl)oxy)phenyl)propanoic acid (axial chirality as shown in structure)(462 mg) as a tan solid. LC/MS (Peptide method): M/Z = 903 [M+H]+. 1H NMR (400 MHz, DMSO-d6) 8.45 (d, J = 2.8 Hz, 1H), 7.81 (s, 1H), 7.54 (d, J = 8.5 Hz, 1H), 7.39 (d, J = 8.5 10 Hz, 1H), 7.25 (d, J = 2.8 Hz, 1H), 7.08 (d, J = 11.7 Hz, 1H), 6.95 (s, 1H), 6.66 (s, 1H), 6.35 (d, J = 8.1 Hz, 1H), 4.42 (t, J = 5.3 Hz, 1H), 4.03 (m, 3H), 3.88 (dd, J = 14.5, 7.1 Hz, 1H), 3.25 (q, J = 4.3 Hz, 3H), 3.07 (dd, J = 10.3, 5.5 Hz, 1H), 3.03 – 2.96 (m, 2H), 2.86 (s, 3H), 2.68 (d, J = 6.7 Hz, 1H), 2.46 (t, J = 5.0 Hz, 3H), 2.22 (s, 3H), 1.36 (d, J = 6.3 Hz, 3H), 1.32 (s, 8H), 1.30 – 1.27 (m, 4H), 1.15 (t, J = 7.2 Hz, 4H), 1.10 (d, J = 7.4 Hz, 18H), 0.86 (t, J = 6.7 Hz, 4H), 0.69 (s, 3H), 0.62 15 (s, 3H). Intermediate 4 Note: axial chirality of compounds is as shown in below scheme. (S)-2-((tert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1- 20 methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)thiazol-2-yl)propanoic acid PAT059646-WO-PCT 5 Procedure Step a: To a solution of Intermediate 2 (28.2 g) in toluene (200 mL) was added Pin2B2 (16.1 g) and KOAc (10.3 g). To this mixture was added Pd(dppf)Cl2 (3.11 g). The reaction was stirred at 100°C for 12 hr. The reaction was concentrated under reduced pressure to yield the crude residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl10 acetate = 1:0 to 0:1) to yield benzyl (S)-4-(5-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-5-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indol-2-yl)-6-(1-methoxyethyl)pyridin-3-yl)piperazine-1- carboxylate (18.7 g) as a yellow solid. LC/MS: M/Z = 711.6 [M+H]+. Step b: A mixture of benzyl (S)-4-(5-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-5-(4,4,5,5-15 tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indol-2-yl)-6-(1-methoxyethyl)pyridin-3-yl)piperazine-1- carboxylate (270 mg), methyl (S)-3-(4-bromothiazol-2-yl)-2-((tert- butoxycarbonyl)amino)propanoate (153 mg), PdCl2(dtbpf) (24.8 mg) and potassium carbonate (105 mg) in 1,4-dioxane (2.0 mL) and water (0.4 mL) was stirred at 85°C for 3 hr under nitrogen. The reaction mixture was diluted with DCM and water and the layers were separated. The aq. 20 layer was extracted with DCM (3x), and the combined organic extracts were washed with brine, dried over MgSO4, filtered, and concentrated. The residue was purified by column chromatography (C18, 0 to 100% acetonitrile in water + 0.1% formic acid) to give benzyl 4-(5-(5- (2-((S)-2-((tert-butoxycarbonyl)amino)-3-methoxy-3-oxopropyl)thiazol-4-yl)-1-ethyl-3-(3-hydroxy- 2,2-dimethylpropyl)-1H-indol-2-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate 25 (250 mg) as a beige powder after lyophilization.1H NMR (400 MHz, DMSO-d6) 8.46 (d, J = 2.8 Hz, 1H), 8.22 (d, J = 1.7 Hz, 1H), 7.80 (s, 1H), 7.75 (dd, J = 8.6, 1.6 Hz, 1H), 7.52 (d, J = 8.6 Hz, 1H), 7.43 (d, J = 8.4 Hz, 1H), 7.39 - 7.31 (m, 6H), 5.11 (s, 2H), 4.51 (td, J = 8.8, 4.9 Hz, 1H), 4.43 (t, J = 5.3 Hz, 1H), 4.10 - 4.00 (m, 2H), 3.93 - 3.82 (m, 1H), 3.68 (s, 3H), 3.56 (s, 4H), 3.51 - 3.37 (m, 2H), 3.28 (q, J = 4.8 Hz, 4H), 3.12 - 2.97 (m, 2H), 2.84 (s, 3H), 2.71 - 2.65 (m, 1H), 2.22 (d, J 30 = 14.0 Hz, 1H), 1.37 (t, J = 2.4 Hz, 12H), 1.15 (t, J = 7.1 Hz, 3H), 0.67 (s, 3H), 0.61 (s, 3H). Step c: A flask containing benzyl 4-(5-(5-(2-((S)-2-((tert-butoxycarbonyl)amino)-3-methoxy-3- oxopropyl)thiazol-4-yl)-1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-1H-indol-2-yl)-6-((S)-1- methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (250 mg) and Pd(OH)2 on carbon (212 mg, 35 20% wt.) in MeOH (2.0 mL) under nitrogen atmosphere was evacuated and backfilled with hydrogen (2x). The reaction was stirred at RT for 2 hr. The reaction diluted with methanol, filtered. The filtrate was dried under reduced pressure to yield methyl (S)-2-((tert-butoxycarbonyl)amino)- 3-(4-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(piperazin-1- yl)pyridin-3-yl)-1H-indol-5-yl)thiazol-2-yl)propanoate (155 mg). LC/MS (RXNMON-Acidic 40 method): M/Z = 735 [M+H]+. PAT059646-WO-PCT 5 Step d: A solution of methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)thiazol-2- yl)propanoate (155 mg) and aq. formaldehyde solution (47.1 L, 37% wt.) in MeOH (2.0 mL) was stirred at RT for 20 min. before addition of sodium triacetoxyborohydride (89 mg). The reaction stirred at RT for 2 h. Additional formaldehyde solution (24 L) and sodium triacetoxyborohydride 10 (45 mg) were added and the reaction stirred at RT for 1 hr. The reaction was quenched directly with a volumetric equivalent of sat. NaHCO3 Solution. and DMSO and stirred vigorously. The mixture was then partially dried under reduced pressure to remove MeOH. The resulting slurry was purified by column chromatography (C18, 0 to 100% acetonitrile in water + 0.1% TFA) to give methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-15 ((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)thiazol-2- yl)propanoate (150 mg) as a yellow solid. LC/MS (RXNMON-Acidic method): M/Z = 750 [M+H]+. Step e: A solution of methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5- 20 yl)thiazol-2-yl)propanoate (150 mg) and trimethyltin hydroxide (181 mg) in DCE (2.0 mL) was stirred at 60°C for 16 hr under nitrogen. Additional trimethyltin hydroxide (90 mg) was added and the reaction was stirred at 60°C for 6 hr. The reaction mixture was concentrated and purified by column chromatography (C18, 0 to 100% acetonitrile in water + 0.1% TFA) to give (S)-2-((tert- butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1- 25 methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)thiazol-2-yl)propanoic acid (axial chirality as shown in structure)(140 mg) as a yellow solid. LC/MS (Peptide method): M/Z = 735 [M+H]+. Intermediate 5 30 Note: axial chirality of compounds is as shown in below scheme. tert-butyl ((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1- yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)- indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)carbamate
PAT059646-WO-PCT 5 Procedure Step a, part 1: To a stirred solution of Intermediate 1 (20.0 mg) in DCM (0.5 mL) was added trifluoroacetic acid (215 μL). The resulting solution was stirred at RT for 1 hr. The reaction mixture 10 was concentrated under reduced pressure. The resulting residue was taken up in DCM and then concentrated under reduced pressure (3x) to afford methyl (1S,2S,6R)-3,4- diazabicyclo[4.1.0]heptane-2-carboxylate bis-trifluoroacetate salt (14.2 mg) LC/MS (RXNMON- Acidic method): M/Z = 156 [M+H]+. 15 Step a, part 2: To a stirred solution of Intermediate 3 (trifluoroacetic acid salt; 25.0 mg) and methyl (1S,2S,6R)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylate bis-trifluoroacetate salt (14.2 mg) in DMF (0.50 mL) was added DIPEA (52 μL) and HATU (18.7 mg). The resulting solution was stirred at RT for 30 min. The reaction mixture was diluted with DMSO (1 mL) and purified by column chromatography (C18, 0 to 100% acetonitrile in water + 0.1% TFA) followed by lyophlization to20 afford methyl (1S,2S,6R)-4-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)- 5-((triisopropylsilyl)oxy)phenyl)propanoyl)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylate (27.0 mg) as a yellow solid. LC/MS (RXNMON-Acidic method): M/Z = 1039 [M+H]+. PAT059646-WO-PCT 5 Step b: To a stirred solution of methyl (1S,2S,6R)-4-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1- ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1- yl)pyridin-3-yl)-1H-indol-5-yl)-5-((triisopropylsilyl)oxy)phenyl)propanoyl)-3,4- diazabicyclo[4.1.0]heptane-2-carboxylate (22.0 mg) in DCE (1.0 mL) was added trimethyltin hydroxide (38.3 mg). The solution was stirred at 75°C for 24 hr. The reaction was cooled to RT 10 and quenched with water (5 mL). The resulting mixture was extracted with EtOAc (3 x 5 mL), and the combined organic layers were washed with brine, dried over Na2SO4, and concentrated to afford (1S,2S,6R)-4-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)- 5-((triisopropylsilyl)oxy)phenyl)propanoyl)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylic acid (17.0 15 mg). LC/MS (Peptide method): M/Z = 1025 [M+H]+. Step c: To a stirred solution of (1S,2S,6R)-4-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3- (3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)- 1H-indol-5-yl)-5-((triisopropylsilyl)oxy)phenyl)propanoyl)-3,4-diazabicyclo[4.1.0]heptane-2- 20 carboxylic acid (14.5 mg) in DCM (1.0 mL) was added EDC (13.6 mg) and HOAt (9.6 mg). The solution stirred at RT for 20 hr under nitrogen. The reaction mixture was concentrated and purified by column chromatography (C18, 0 to 100% acetonitrile in water + 0.1% TFA) followed by lyophilization to afford tert-butyl ((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-25 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)carbamate (axial chirality as shown in structure)(14.0 mg) as a white solid. LC/MS (Peptide method): M/Z = 1007 [M+H]+. Intermediate 6 30 Note: axial chirality of compounds is as shown in below scheme. tert-butyl ((61R,65S,66S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1- yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-2(4,2)-thiazola-1(5,3)-indola- 6(3,5)-bicyclo[4.1.0]heptanacycloundecaphane-4-yl)carbamate
PAT059646-WO-PCT 5 Procedure Step a, part 1: A solution of Intermediate 1 (111 mg) in DCM (3.5 mL) and TFA (1.2 mL) was stirred at RT for 1 hr under nitrogen. The reaction mixture was concentrated and dried under10 reduced pressure to yield methyl (1S,2S,6R)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylate bis- trifluoroacetate salt (120 mg) as a yellow oil. LC/MS (RXMON-Acidic method): M/Z = 157 [M+H]+. Step a, part 2: To a solution of Intermediate 4 (47.8 mg) in NMP (0.10 mL) was added methyl (1S,2S,6R)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylate bis-trifluoroacetate salt (27.5 mg), 15 followed by DIPEA (0.11 mL) and HATU (49.5 mg) under nitrogen. The solution was stirred at RT for 30 min. The reaction mixture was diluted with minimal DMSO and purified by column chromatography (C18, 10 to 40% acetonitrile in water + 0.1% TFA) to yield a yellow solid after lyophilization. This solid was reconstituted in EtOAc and neutralized via vigorous stirring in the presence of sat. NaHCO3 solution. The resulting organic layer was then passed through a phase20 separator, concentrated, and dried to yield methyl (1S,2S,6R)-4-((2S)-2-((tert- butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)thiazol-2-yl)propanoyl)-3,4- diazabicyclo[4.1.0]heptane-2-carboxylate (22.1 mg) as a pale yellow foam. LC/MS (Peptide method): M/Z = 895 [M+Na]+. 25 PAT059646-WO-PCT 5 Step b: To a solution of methyl (1S,2S,6R)-4-((2S)-2-((tert-butoxycarbonyl)amino)-3-(4-(1-ethyl- 3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3- yl)-1H-indol-5-yl)thiazol-2-yl)propanoyl)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylate (22.1 mg) in DCE (0.5 mL) under nitrogen was added trimethyltin hydroxide (22.9 mg). The resulting suspension was stirred at 70°C for 1.5 hr. The reaction mixture was cooled to RT, diluted with 10 DCM (5.0 mL), and quenched with aq. NaHSO4 (0.05 M, 2.5 mL). The organic layer was then passed through a phase separator and concentrated to afford crude (1S,2S,6R)-4-((S)-2-((tert- butoxycarbonyl)amino)-3-(4-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)thiazol-2-yl)propanoyl)-3,4- diazabicyclo[4.1.0]heptane-2-carboxylic acid (21.7 mg) as an off-white foam. This material was 15 used in the next step without further purification. LC/MS (Peptide method): M/Z = 860 [M+H]+. Step c: To a solution of crude (1S,2S,6R)-4-((S)-2-((tert-butoxycarbonyl)amino)-3-(4-(1-ethyl-3- (3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)- 1H-indol-5-yl)thiazol-2-yl)propanoyl)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylic acid (21.7 mg) 20 and HOAt (17.2 mg) in DCM (2.50 mL) under a nitrogen atmosphere was added a solution of EDC (24.3 mg) in DCM (0.5 mL) dropwise over the course of 20 min. The resulting suspension was stirred at RT for 16 hr. The reaction mixture was concentrated and purified by column chromatography (C18, 10 to 50% acetonitrile in water + 0.1% TFA) to yield tert-butyl ((61R,65S,66S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-25 10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-2(4,2)-thiazola-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptanacycloundecaphane-4-yl)carbamate trifluoroacetate salt (axial chirality as shown in structure) (8.3 mg) as a pale yellow solid after lyophilization. Intermediate 7 30 N-methyl-N-(morpholine-4-carbonyl)-L-valine Procedure Step a: To a solution of N-methyl-L-valine benzyl ester 4-toluenesulfonate (464 mg) in DCM (6.2 35 mL) at 0°C was added DIPEA (1.23 mL). The solution was stirred at 0°C for 10 min. Triphosgene (174.9 mg) was added and the reaction was stirred at 0°C for 10 min, then allowed to warm to RT and stirred for 30 min. Morpholine (103 mg) was added to the reaction at 0°C. The reaction mixture was warmed to RT and stirred for 1 hr. The reaction mixture was quenched with sat. NaHCO3 PAT059646-WO-PCT 5 solution and stirred vigorously. The quenched mixture was passed through a phase separator and concentrated under reduced pressure. The crude material was purified by column chromatography (SiO2, 0 to 80% EtOAc in heptane) to yield benzyl N-methyl-N-(morpholine-4- carbonyl)-L-valinate (230 mg) as a clear oil. LC/MS (FinalAnalysis-acidic method): M/Z = 335 [M+H]+. 10 Step b: A flask containing benzyl N-methyl-N-(morpholine-4-carbonyl)-L-valinate (231 mg) and Pd on carbon (732 mg, 10 wt%) in MeOH (8.1 mL) at RT was evacuated and backfilled with nitrogen. Triethylsilane (240 mg) was added over the course of 5 min. The reaction was stirred at RT for 30 min. The reaction mixture was filtered over a pad of Celite®, rinsed with methanol, and 15 then concentrated to yield N-methyl-N-(morpholine-4-carbonyl)-L-valine (171 mg) as a colorless oil that solidifies over time. LC/MS (RXNMON-Acidic method): M/Z = 245 [M+H]+. Intermediate 8 1-(6-isopropoxy-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-3-yl)-4-methylpiperazine 20 Procedure Step a: A mixture of 3-bromo-2-chloro-5-iodopyridine (25.0 g) and cesium carbonate (64.0 g) in isopropanol (250 mL) was stirred at 80°C for 17 hr. Additional cesium carbonate (12.8 g) was 25 added and the reaction mixture was stirred at 80°C for 5 hr. The reaction mixture was concentrated, diluted in EtOAc, filtered over a pad of Florisil®, concentrated, and dried under reduced pressure which yielded 3-bromo-5-iodo-2-isopropoxypyridine (26.0 g) as a dark orange oil.1H NMR (400 MHz, CDCl3) 8.22 (d, J = 2.0 Hz, 1H), 8.03 (d, J = 2.1 Hz, 1H), 5.27 (hept, J = 6.2 Hz, 1H), 1.37 (d, J = 6.2 Hz, 6H). 30 Step b: To a solution of 3-bromo-5-iodo-2-isopropoxypyridine (1.04 g) and xantphos (176 mg) in toluene (6.1 mL) was added 1-methyl piperazine (320 mg), bis(dibenzylideneacetone)dipalladium (137 mg), and sodium tert-butoxide (2M in THF, 3.80 mL). The solution was sparged with nitrogen for 5 min. then stirred at 60°C for 20 min. The reaction was cooled to RT and poured into brine. 35 The aq. layer was extracted with EtOAc (3x), and the combined organic layers were washed with PAT059646-WO-PCT 5 brine, dried over Na2SO4, filtered, and concentrated. The material was purified by column chromatography (C18, 10 to 60% acetonitrile in water + 0.1% formic acid) which yielded 1-(5- bromo-6-isopropoxypyridin-3-yl)-4-methylpiperazine (580 mg) as an orange oil. LC/MS (FinalAnalysis-acidic method): M/Z = 316 [M+H]+.1H NMR (400 MHz, CDCl3) 7.73 (d, J = 2.7 Hz, 1H), 7.49 (d, J = 2.7 Hz, 1H), 5.21 (hept, J = 6.2 Hz, 1H), 3.14 (s, 4H), 2.63 (s, 2H), 2.40 (s, 10 2H), 1.36 (d, J = 6.2 Hz, 6H). Step c: A vial containing 1-(5-bromo-6-isopropoxypyridin-3-yl)-4-methylpiperazine (209 mg), bis(pinacolato)diborane (338 mg), potassium acetate (196 mg), and PdCl2(dtbpf) (43.4 mg) was evacuated and backfilled with nitrogen. Toluene (6.7 mL) was added, and the reaction mixture 15 was sparged with nitrogen for 5 min. then stirred at 90°C for 18 hr. The reaction mixture was filtered over pad of Florisil® and Celite®, rinsed with EtOAc and MeOH, then concentrated. The crude residue was dissolved in EtOAc, filtered through phase separator, concentrated, dissolved in heptane, filtered through a phase separator, concentrated, dissolved in DCM, filtered, and concentrated to yield crude 1-(6-isopropoxy-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- 20 yl)pyridin-3-yl)-4-methylpiperazine (300 mg) as a brown oil. The material was used in the next step without further purification. LC/MS (FinalAnalysis-Acidic): M/Z = 280 [M-C6H10+H]+ (mass of boronic acid; BPin ester hydrolyzes under LCMS conditions). Intermediate 9 and Intermediate 9a 25 tert-butyl ((61R,65S,66S,4S)-11-ethyl-12-(2-isopropoxy-5-(4-methylpiperazin-1-yl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)carbamate
PAT059646-WO-PCT 5 Procedure Step a: To a solution of 3-(5-bromo-1H-indol-3-yl)-2,2-dimethylpropyl acetate (46.0 g), methyl (S)- 2-((tert-butoxycarbonyl)amino)-3-(3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5- ((triisopropylsilyl)oxy)phenyl)propanoate (62.5 g) and K2CO3 (48.0 g) in 1,4-dioxane (460 mL) and10 water (92.0 mL) that had been degassed and purged with nitrogen (3x) was added bis(di-tert- butyl(4-dimethylaminophenyl)phosphine)dichloropalladium (4.94 g). The reaction mixture was stirred at 85 C for 4 hr. The resulting mixture was combined with other two batches (5.0 g and 9.3 g of 3-(5-bromo-1H-indol-3-yl)-2,2-dimethylpropyl acetate were used in the same manner) for the subsequent workup. The reaction mixture was concentrated under reduced pressure to PAT059646-WO-PCT 5 remove dioxane and water. The residue was purified by column chromatography (SiO2, 0 to 17% EtOAc in petroleum ether) to afford methyl (S)-3-(3-(3-(3-acetoxy-2,2-dimethylpropyl)-1H-indol-5- yl)-5-((triisopropylsilyl)oxy)phenyl)-2-((tert-butoxycarbonyl)amino)propanoate (92.0 g) as a white solid.1H NMR (400 MHz, DMSO-d6) 10.98 (d, J = 1.6 Hz, 1H), 7.68 (s, 1H), 7.41 (d, J = 8.4 Hz, 1H), 7.34 – 7.26 (m, 2H), 7.15 – 7.08 (m, 2H), 6.95 (s, 1H), 6.72 (s, 1H), 4.23 (td, J = 10.2, 4.9 10 Hz, 1H), 3.74 (s, 2H), 3.61 (s, 3H), 3.08 – 2.97 (m, 1H), 2.92 – 2.81 (m, 1H), 2.70 (s, 2H), 2.03 (s, 3H), 1.35 – 1.26 (m, 12H), 1.10 (d, J = 7.3 Hz, 18H), 0.93 (s, 6H). Step b: To a solution of methyl (S)-3-(3-(3-(3-acetoxy-2,2-dimethylpropyl)-1H-indol-5-yl)-5- ((triisopropylsilyl)oxy)phenyl)-2-((tert-butoxycarbonyl)amino)propanoate (65.0 g) and NaHCO3 15 (9.3 g) in THF (650 mL) was added AgOTf (22.7 g) in THF (100 mL) and I2 (16.8 g) in THF (100 mL) at 0 C. The reaction mixture was stirred at 0 C for 2 hr. Another portion of AgOTf (3.79 g) in THF (15.0 mL) and I2 (3.74 g) in THF (15.0 mL) were then added dropwise, and the reaction mixture was stirred at 0 C for 1 hr. The reaction was stirred for a total of 3 hr at 0 C. The resulting mixture was combined with two batches (5.0 g and 20.0 g of methyl (S)-3-(3-(3-(3-acetoxy-2,2-20 dimethylpropyl)-1H-indol-5-yl)-5-((triisopropylsilyl)oxy)phenyl)-2-((tert- butoxycarbonyl)amino)propanoate were used in the same manner) for the subsequent workup. The combined crude products were diluted with aq. Na2S2O3 (2.0 L) and extracted with EtOAc (3 x 3.0 L). The combined organic layers were washed with brine (2 x 1.5 L), dried over Na2SO4 and filtered. The residue was purified by column chromatography (SiO2, 1 to 17% EtOAc in petroleum25 ether) to afford methyl (S)-3-(3-(3-(3-acetoxy-2,2-dimethylpropyl)-2-iodo-1H-indol-5-yl)-5- ((triisopropylsilyl)oxy)phenyl)-2-((tert-butoxycarbonyl)amino)propanoate (50.5 g) as a yellow solid. 1H NMR (300 MHz, DMSO-d6) 11.63 (d, J = 17.3 Hz, 1H), 7.65 (s, 1H), 7.35 (d, J = 8.4 Hz, 1H), 7.30 – 7.26 (m, 2H), 7.11 (s, 1H), 6.94 (s, 1H), 6.73 (s, 1H), 4.23 (dd, J = 13.0, 9.7 Hz, 1H), 3.84 (s, 2H), 3.63 (s, 3H), 3.03 (dd, J = 13.7, 4.5 Hz, 1H), 2.95 – 2.81 (m, 1H), 2.67 (s, 2H), 2.04 (s, 30 3H), 1.34 – 1.22 (m, 12H), 1.09 (d, J = 7.2 Hz, 18H), 0.99 (s, 6H). Step c: To a solution of methyl (S)-3-(3-(3-(3-acetoxy-2,2-dimethylpropyl)-2-iodo-1H-indol-5-yl)-5- ((triisopropylsilyl)oxy)phenylycarbo)-2-((tert-butoxnyl)amino)propanoate (750 mg) in DCE (5.0 mL) was added trimethyltin hydroxide (496 mg). The reaction was stirred at 80°C for 24 hr. 35 Additional trimethyltin hydroxide (248 mg) was added and the reaction mixture stirred at 80°C for 24 hr. The reaction was cooled to RT, diluted with water (5 mL), and stirred vigorously for 5 min. The milky mixture was diluted with DCM, passed through a phase separator, and rinsed with DCM. The pale, yellow solution was concentrated under reduced pressure to yield (S)-2-((tert- butoxycarbonyl)amino)-3-(3-(3-(3-hydroxy-2,2-dimethylpropyl)-2-iodo-1H-indol-5-yl)-5- 40 ((triisopropylsilyl)oxy)phenyl)propanoic acid (765 mg) as an off-white solid. This material was used in the next step without further purification. LC/MS (RXNMON-basic method): M/Z = 765 [M+H]+. PAT059646-WO-PCT 5 Step d: To a solution of Intermediate 1 (391 mg) in DCM (9.0 mL) was added TFA (3.0 mL). The reaction was stirred at RT for 3 hr then concentrated under reduced pressure. To a solution of the resultant yellow liquid and (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(3-(3-hydroxy-2,2- dimethylpropyl)-2-iodo-1H-indol-5-yl)-5-((triisopropylsilyl)oxy)phenyl)propanoic acid (780 mg) in 10 DMF (4.00 mL) was added DIPEA (1.07 mL) and HATU (427 mg). The reaction mixture stirred at RT for 20 min. and then purified by column chromatography (SiO2, 0 to 100% (3:1 EtOAc/EtOH) in heptane) which yielded methyl (1S,2S,6R)-4-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(3-(3- hydroxy-2,2-dimethylpropyl)-2-iodo-1H-indol-5-yl)-5-((triisopropylsilyl)oxy)phenyl)propanoyl)-3,4- diazabicyclo[4.1.0]heptane-2-carboxylate (705 mg) as a white solid. LC/MS (RXNMON-acidic 15 method): M/Z = 904 [M+H]+.1H NMR (400 MHz, DMSO-d6) 11.52 (s, 1H), 7.70 (s, 1H), 7.32 (d, J = 8.3 Hz, 1H), 7.30 – 7.19 (m, 1H), 7.06 (s, 1H), 6.95 – 6.88 (m, 1H), 6.68 (s, 1H), 6.36 (d, J = 8.7 Hz, 1H), 5.23 (d, J = 6.9 Hz, 1H), 5.10 (td, J = 8.7, 3.9 Hz, 1H), 4.61 (t, J = 5.2 Hz, 1H), 3.90 – 3.59 (m, 7H), 3.41 (d, J = 13.2 Hz, 1H), 3.32 (s, 5H), 3.24 (d, J = 5.3 Hz, 3H), 2.95 – 2.82 (m, 2H), 1.34 – 1.15 (m, 20H), 0.89 (s, 9H), 0.75 (td, J = 8.5, 4.6 Hz, 1H), 0.47 (d, J = 5.3 Hz, 1H). 20 Step e: To a solution of methyl (1S,2S,6R)-4-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(3-(3- hydroxy-2,2-dimethylpropyl)-2-iodo-1H-indol-5-yl)-5-((triisopropylsilyl)oxy)phenyl)propanoyl)-3,4- diazabicyclo[4.1.0]heptane-2-carboxylate (705 mg) in DCE (4.0 mL) was added trimethyltin hydroxide (424 mg). The reaction was stirred at 70°C for 4 hr. The reaction was cooled to RT, 25 diluted with water, stirred vigorously for 5 min, diluted with DCM, passed through a phase separator, rinsed with DCM, and concentrated under reduced pressure. The crude material was purified by column chromatography (C18, 20 to 100% acetonitrile in water + 0.1% NH4OH). Fractions containing the product were concentrated and sat. NaHCO3 solution (15 mL) was added. The mixture was extracted with DCM (3 x 15 mL) and the combined organic layers were passed30 through a phase separator and concentrated under reduced pressure to yield (1S,2S,6R)-4-((S)- 2-((tert-butoxycarbonyl)amino)-3-(3-(3-(3-hydroxy-2,2-dimethylpropyl)-2-iodo-1H-indol-5-yl)-5- ((triisopropylsilyl)oxy)phenyl)propanoyl)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylic acid (415 mg) as a white solid. LC/MS (Peptide Method): M/Z = 911 [M+H]+. 35 Step f: To a solution of (1S,2S,6R)-4-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(3-(3-hydroxy-2,2- dimethylpropyl)-2-iodo-1H-indol-5-yl)-5-((triisopropylsilyl)oxy)phenyl)propanoyl)-3,4- diazabicyclo[4.1.0]heptane-2-carboxylic acid (415 mg) in DCM (20 mL) was added 1H- benzo[d][1,2,3]triazol-1-ol hydrate (249 mg, 86% wt.) and EDC (179 mg). The reaction mixture was stirred at RT for 14 hr. The reaction was diluted with water (15 mL) and stirred vigorously for 40 5 min. The resulting biphasic mixture was passed through a phase separator and the organic layer was concentrated. The crude material was purified by column chromatography (SiO2, 0 to 100% PAT059646-WO-PCT 5 3:1 EtOAc/EtOH in heptane) to yield tert-butyl ((61R,65S,66S,4S)-12-iodo-10,10-dimethyl-5,7- dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana- 2(1,3)-benzenacycloundecaphane-4-yl)carbamate (338 mg) as a white sold. LC/MS (Peptide Method): M/Z = 893 [M+H]+. 10 Step g: To a solution of tert-butyl ((61R,65S,66S,4S)-12-iodo-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)carbamate (338 mg) in DMF (3.0 mL) was added DIPEA (206 L), cesium carbonate (506 mg), and iodoethane (62.1 L). The reaction was stirred at 35°C for 2.5 hr. The reaction was diluted with water (10 mL) and stirred vigorously. The aq. layer was 15 extracted with EtOAc (3x), and the combined organic extracts were concentrated. The crude material was purified by column chromatography (SiO2, 0 to 60% 3:1 EtOAc/EtOH in heptane) to yield tert-butyl ((61R,65S,66S,4S)-11-ethyl-12-iodo-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)carbamate (328 mg) as a white solid. LC/MS (FinalAnalysis- 20 Basic method): M/Z = 900 [M+H]+.1H NMR (400 MHz, DMSO-d6) 7.92 (d, J = 1.8 Hz, 1H), 7.58 (d, J = 8.6 Hz, 1H), 7.46 (dd, J = 8.8, 1.7 Hz, 1H), 7.26 (s, 1H), 7.13 (d, J = 8.8 Hz, 1H), 7.03 (t, J = 1.9 Hz, 1H), 6.81 (s, 1H), 4.91 (t, J = 9.1 Hz, 1H), 4.79 (d, J = 12.0 Hz, 1H), 4.38 – 4.22 (m, 3H), 4.16 (d, J = 12.8 Hz, 1H), 3.92 (d, J = 11.0 Hz, 1H), 3.51 (d, J = 11.0 Hz, 1H), 3.27 (d, J = 13.2 Hz, 1H), 3.14 (d, J = 14.4 Hz, 1H), 2.76 (dd, J = 14.0, 9.9 Hz, 1H), 2.59 (d, J = 13.8 Hz, 1H), 1.38 25 – 1.19 (m, 21H), 1.09 (dd, J = 7.4, 1.7 Hz, 18H), 1.02 – 0.91 (m, 4H), 0.40 (d, J = 5.5 Hz, 1H). Step h: To a solution of tert-butyl ((61R,65S,66S,4S)-11-ethyl-12-iodo-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)carbamate (328 mg), Intermediate 8 (crude, 198 mg), aq. K3PO4 30 solution (155 mg, 730 L, 1.0 molar), and mesylate[(di(1-adamantyl)-n-butylphosphine)-2-(2 - amino-1,1 -biphenyl)]palladium(II) (26.6 mg) were dissolved in 1,4-dioxane (3.7 mL) and sparged with nitrogen for 5 min. before heating at 85°C for 1.5 hr. The reaction mixture was poured into separatory funnel and diluted with sat. NaHCO3 solution. The aq. layer was extracted with EtOAc (3x), and the combined organic layers were washed with brine and concentrated. The crude 35 material was purified by column chromatography (C18, 50 to 100% acetonitrile in water + 0.1% TFA) to yield tert-butyl ((61R,65S,66S,4S)-11-ethyl-12-(2-isopropoxy-5-(4-methylpiperazin-1- yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)- indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)carbamate (200 mg) as a brownish orange solid. LC/MS (FinalAnalysis-Acidic method): M/Z = 1007 [M+H]+.1H NMR 40 (400 MHz, DMSO-d6) 7.99 – 7.84 (m, 2H), 7.59 – 7.52 (m, 1H), 7.51 – 7.45 (m, 1H), 7.40 – 7.31 (m, 1H), 7.27 (s, 0H), 7.23 – 7.16 (m, 1H), 7.15 – 7.08 (m, 1H), 7.05 – 6.98 (m, 1H), 6.83 – 6.77 PAT059646-WO-PCT 5 (m, 1H), 6.64 (s, 0H), 5.36 – 5.22 (m, 1H), 5.01 – 4.87 (m, 1H), 4.82 – 4.72 (m, 1H), 4.39 – 4.28 (m, 1H), 4.25 – 4.08 (m, 2H), 3.98 – 3.83 (m, 1H), 3.81 – 3.65 (m, 1H), 3.61 – 3.40 (m, 1H), 3.28 – 3.20 (m, 1H), 3.14 – 3.06 (m, 4H), 2.78 – 2.66 (m, 1H), 2.65 – 2.57 (m, 1H), 2.27 – 2.18 (m, 3H), 2.17 – 2.01 (m, 1H), 1.97 – 1.86 (m, 4H), 1.78 – 1.62 (m, 4H), 1.52 (s, 1H), 1.35 – 1.32 (m, 9H), 1.32 – 1.28 (m, 4H), 1.28 – 1.18 (m, 6H), 1.14 (s, 1H), 1.11 – 1.08 (m, 18H), 1.04 – 1.00 (m, 10 1H), 0.96 (s, 2H), 0.93 – 0.87 (m, 2H), 0.55 – 0.44 (m, 1H), 0.39 (s, 3H). Intermediate 10 (2R,3S)-3-phenyloxetane-2-carboxylic acid 15 Procedure Step a: A solution of styrene (26.0 g) and 2-oxoacetic acid (74.0 g, 50% in water) in CH3CN (1 L) was purged with nitrogen for 10 min. while the entire vessel was sonicated. The reaction mixture was transferred to a photoreactor consisting of a closed borosilicate cylinder equipped with a cold 20 finger, which was set to 5°C. The cylinder was irradiated with total 801W high power LEDs (365 nm) for 24 hr, and then the reaction mixture was concentrated at 40°C to yield 68.2 g of a viscous pale-brown oil that was dissolved in 250 mL MTBE and washed twice with 100 mL water followed by 216 mL 1 N NaOH solution. The basic aqueous solution was washed with 150 mL MTBE then acidified by slow addition of 12.8 g conc. H2SO4 to a pH of <3 and extracted with MTBE. The 25 combined organic phases were washed with brine, dried over sodium sulfate, filtered, and concentrated to yield a crude cis/trans mixture of 3-phenyloxetane-2-carboxylic acid (35.5 g) as a viscous pale brown oil. HPLC (HPLC-quant. method): Rt = 3.01 min. (cis acid); 3.42 min. (trans acid). MS (ES–): m/z 177 [M–H]. 30 Step b: To a solution of a crude cis/trans mixture of 3-phenyloxetane-2-carboxylic acid (25.9 g) in 150 mL absolute EtOH was added AmberChromTM, 50WX4 (5.9 g) and the suspension heated at 60°C for 2 hr, then at 70°C for another 4.5 hr. The reaction mixture was cooled to RT, filtered, and concentrated to yield 28.0 g of orange oil. The crude material was purified by column PAT059646-WO-PCT 5 chromatography (5 to 30% EtOAc/heptane) to afford racemic ethyl cis-3-phenyloxetane-2- carboxylate (1.76 g) as pale-yellow oil as the second eluting isomer. LC/MS (Final_Analysis_2min method): Rt = 0.71 min, m/z 207 [M+H]+.1H NMR (400 MHz, DMSO-d6) 7.33 (d, J = 3.5 Hz, 4H), 7.29 – 7.23 (m, 1H), 5.47 (d, J = 9.2 Hz, 1H), 4.91 (dd, J = 8.2, 6.0 Hz, 1H), 4.80 (t, J = 6.4 Hz, 1H), 4.57 (m, 1H), 3.70 (q, J = 7.1 Hz, 2H), 0.69 (t, J = 7.1 Hz, 3H). 10 Step c: Ethyl cis-3-phenyloxetane-2-carboxylate (1.15 g) was dissolved in EtOH + 0.05 NH3 (18 mL) and then purified by chiral SFC (Prep_SFC2 method) to give ethyl (2R,3S)-3-phenyloxetane- 2-carboxylate as the first eluting isomer as colorless oil (514 mg; 98% ee). Chiral SFC (Chiral_SFC1 method): Rt = 1.15 min. LC/MS (Final_Analysis_2min method): m/z 207 [M+H]+.1H 15 NMR (400 MHz, DMSO-d6) 7.30 – 7.36 (m, 5 H), 5.47 (d, J = 9.2 Hz, 1 H), 4.91 (dd, J = 8.1, 6.0 Hz, 1 H), 4.80 (t, J = 6.3 Hz, 1 H), 4.54 – 4.60 (m, 2 H), 3.70 (q, J = 7.1 Hz, 3 H), 0.69 (t, J = 7.1 Hz, 3 H). Step d: A mixture of (2R,3S)-3-phenyloxetane-2-carboxylate (496 mg) in THF (2.5 mL) and MeOH 20 (2.5 mL) was cooled in an ice bath. LiOH monohydrate (151 mg) was added, and the resulting mixture stirred at 0°C for 3 h. The reaction mixture was diluted with DCM (10 mL) and the biphasic mixture was acidified to be a pH of 3.5 with citric acid (4 mL, 10% aqueous solution), followed by the addition of solid citric acid (0.2 g). The phases were separated, and the aqueous layer extracted 6 times with DCM. The combined organic phases were dried over sodium sulfate, 25 filtered, and concentrated to yield (2R,3S)-3-phenyloxetane-2-carboxylic acid (399 mg; >99% ee) as a white powder. Chiral UPLC (Chiral_UPLC1 method): Rt = 5.71 min. LC/MS (Final_Analysis_2min): m/z 177 [M–H]–.1H NMR (400 MHz, DMSO-d6) 12.45 (s, 1H), 7.39 – 7.30 (m, 4H), 7.29 – 7.23 (m, 1H), 5.39 (d, J = 9.1 Hz, 1H), 4.90 (dd, J = 8.1, 5.9 Hz, 1H), 4.70 (t, J = 6.3 Hz, 1H), 4.50 (td, J = 8.6, 6.6 Hz, 1H). 30 Intermediate 11 (S)-4-(6-(1-methoxyethyl)-5-(4,4,5,5-tetraethyl-1,3,2-dioxaborolan-2-yl)pyridin-3-yl)morpholine 35 PAT059646-WO-PCT 5 Step a: To a mixture of (S)-3-bromo-5-iodo-2-(1-methoxyethyl)pyridine (3.05 g), Pd(OAc)2 (100 mg), rac-BINAP (278 mg) and Cs2CO3 (5.812 g) in toluene (30 mL) at RT was added morpholine (855 mg, 846 L) under nitrogen. The reaction mixture was allowed to stir at 90°C for 16 hr, cooled to RT, filtered, and concentrated. The residue was purified by flash column chromatography on (SiO2, 0 to 30% 3:1 EtOAc/EtOH in heptane) to afford (S)-4-(5-bromo-6-(1-methoxyethyl)pyridin- 10 3-yl)morpholine (2.35 g) as an orange oil. LC/MS (RXNMON-Acidic): M/Z = 301.0 [M+H]+. 1H NMR (400 MHz, CDCl3) 8.30 (d, J = 2.6 Hz, 1H), 7.30 (s, 1H), 4.86 (q, J = 6.4 Hz, 1H), 3.91 – 3.82 (m, 4H), 3.29 (s, 3H), 3.24 – 3.15 (m, 4H), 1.47 (d, J = 6.4 Hz, 3H). Step b: To a solution of (S)-4-(5-bromo-6-(1-methoxyethyl)pyridin-3-yl)morpholine (1.88 g) in15 DMSO (25 mL), 4,4,5,5-tetraethyl-2-(4,4,5,5,-tetraethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (5.71 g), Pd(dppf)Cl2-DCM adduct (127 mg) and potassium phenoxide (1.90 g) were added. The mixture was allowed to stir at 60°C for 1 hr under nitrogen, after which time it was cooled to RT and diluted with water and EtOAc. The layers were separated. The aq. layer was extracted with EtOAc (2x). The combined organic extracts were washed with brine, dried 20 over Na2SO4, filtered, and concentrated. The residue was purified by column chromatography (SiO2, 0 to 60% 3:1 EtOAc/EtOH in heptane) to afford (S)-4-(6-(1-methoxyethyl)-5-(4,4,5,5- tetraethyl-1,3,2-dioxaborolan-2-yl)pyridin-3-yl)morpholine (1.35 g) along with a small amount of impurities as orange oil. LC/MS (RXNMON-Basic): M/Z = 405.0 [M+H]+. 1H NMR (400 MHz, CDCl3) 8.35 (d, J = 3.1 Hz, 1H), 7.48 (d, J = 3.1 Hz, 1H), 4.95 (q, J = 6.4 Hz, 1H), 3.93 – 3.82 25 (m, 4H), 3.24 (s, 3H), 3.22 – 3.16 (m, 4H), 1.92 – 1.70 (m, 8H), 1.46 (d, J = 6.4 Hz, 3H), 1.02 – 0.95 (m, 12H). The following intermediates of Table 1 were synthesized using the above procedure or modifications of the above procedure using the corresponding amine coupling partner. 30 Table 1: 4,4,5,5-tetraethyl-1,3,2-dioxaborolane intermediates PAT059646-WO-PCT 5 Intermediate 13 (S)-8-(6-((S)-1-methoxyethyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-3- yl)octahydropyrazino[2,1-c][1,4]oxazine 10 Procedure Step a: To a solution of (S)-octahydropyrazino[2,1-c][1,4]oxazine (328 mg) and (S)-3-bromo-5- iodo-2-(1-methoxyethyl)pyridine (745 mg) in toluene (11.2 mL) was added xantphos (126 mg), KOtBu (489 mg), and Pd2dba3 (39.9 mg). The reaction was purged with nitrogen (3x) and stirred at 100°C under nitrogen for 2 hr. The reaction was diluted with EtOAc, filtered through Celite®, 15 and concentrated. The residue was purified by column chromatography (SiO2, 0 to 100% 3:1 EtOAc/EtOH in heptane) to afford (S)-8-(5-bromo-6-((S)-1-methoxyethyl)pyridin-3- yl)octahydropyrazino[2,1-c][1,4]oxazine (281.1 mg). LC/MS (RXNMON-Acidic method): M/Z = 356.4 [M+H]+. 20 Step b: To a solution of (S)-8-(5-bromo-6-((S)-1-methoxyethyl)pyridin-3-yl)octahydropyrazino[2,1- c][1,4]oxazine (281 mg) and bis(pinacolato)diboron (301 mg) in 2-MeTHF (7.7 mL) was added potassium 2-ethylhexanoate (316 mg) and Pd(dppf)Cl2 (86.6 mg). The reaction was purged with nitrogen (3x) and heated at 80°C for 2 hr. The reaction was partitioned with 2% wt. aq. NaHCO3. The aq. layer was extracted with EtOAc. The organic extract was dried with Na2SO4 and 25 concentrated. The residue was purified by column chromatography (propylamine SiO2, 0 to 11% EtOH in heptane) to afford (S)-8-(6-((S)-1-methoxyethyl)-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyridin-3-yl)octahydropyrazino[2,1-c][1,4]oxazine (271 mg). LC/MS (RXNMON-Acidic method): M/Z = 322.4 [M-C6H10]+.1H NMR (400 MHz, CDCl3) 8.30 (d, J = 3.0 Hz, 1H), 7.40 (d, J = 3.1 Hz, 1H), 4.81 (q, J = 6.5 Hz, 1H), 3.89 (m, 1H), 3.84 – 3.71 (m, 2H), 3.59 30 (m, 1H), 3.37 (m, 2H), 3.29 – 3.19 (m, 3H), 3.02 (m, 1H), 2.91 (d, J = 11.1 Hz, 1H), 2.75 (d, J = 11.6 Hz, 1H), 2.56 – 2.39 (m, 4H), 1.45 (m, 3H), 1.36 (d, J = 1.3 Hz, 12H). PAT059646-WO-PCT 5 Intermediate 14 and Intermediate 14a (3S,4S)-1-(methoxycarbonyl)-4-phenylpyrrolidine-3-carboxylic acid Procedure Step a: To a suspension of racemic cis-methyl 4-phenylpyrrolidine-3-carboxylate hydrochloride 10 (2 g) and NaHCO3 (2.09 g) in DCM (30 mL) and water (3 mL) was added methyl carbonochloridate (1.28 mL). The reaction was stirred at RT for 30 min. The reaction mixture was diluted with water, extracted with DCM (2x), passed through a phase separator, and concentrated. The residue was purified by column chromatography (SiO2, 0 to 55% EtOAc in heptane) to afford racemic cis- dimethyl 4-phenylpyrrolidine-1,3-dicarboxylate (1.68 g) as an orange oil. LC/MS (FinalAnalysis- 15 Basic) M/Z = 264.3 [M+H]+. Step b: Racemic cis-dimethyl 4-phenylpyrrolidine-1,3-dicarboxylate (1.68 g) was purified by chiral SFC (Prep_SFC3 method) to yield dimethyl (3S,4S)-4-phenylpyrrolidine-1,3-dicarboxylate (0.766 g) as the first eluting isomer, a pale orange solid.1H NMR (400 MHz, CDCl3) 7.33 – 7.22 (m, 20 3H), 7.18 – 7.13 (m, 2H), 3.94 – 3.80 (m, 3H), 3.76 (s, 3H), 3.72 (q, J = 6.9 Hz, 2H), 3.41 (s, 4H). LC/MS (FinalAnalysis-Basic) M/Z = 264.3 [M+H]+. Step c: A mixture of dimethyl (3S,4S)-4-phenylpyrrolidine-1,3-dicarboxylate (150 mg) and trimethyltin hydroxide (155 mg) in DCE (5.6 mL) was stirred at 85°C. After stirring for 24 hr, 25 additional trimethyltin hydroxide (52 mg) was added. After stirring for 6 hr, additional trimethyltin hydroxide (103 mg) was added. The reaction stirred for a total of 4 days. The reaction mixture was concentrated, suspended in EtOAc and 1N aq. HCl, and the phases were separated. The aq. layer was extracted with EtOAc (3x). The combined organic layers were passed through a phase separator and concentrated. The residue was purified by column chromatography [SiO2, 030 to 60% 3:1 EtOAc/EtOH (containing 1% AcOH) in heptane] to yield (3S,4S)-1-(methoxycarbonyl)- 4-phenylpyrrolidine-3-carboxylic acid (130 mg) as a white solid. 1H NMR (400 MHz, DMSO) 12.25 (d, J = 2.4 Hz, 1H), 7.30 (td, J = 7.4, 2.2 Hz, 2H), 7.26 – 7.21 (m, 1H), 7.20 (s, 2H), 3.72 (dq, J = 11.6, 6.0 Hz, 2H), 3.67 – 3.61 (m, 4H), 3.59 (d, J = 7.0 Hz, 2H), 3.43 (dd, J = 14.5, 6.9 Hz, 1H). LC/MS (FinalAnalysis-Basic) M/Z = 250.3 [M+H]+. 35 PAT059646-WO-PCT 5 The following intermediates of Table 2 were synthesized using the above procedure or modifications of the above procedure. Table 2: Phenylpyrrolidine intermediates 10 Intermediate 16 dimethyl (3S,4S)-4-(4-(4-(tert-butoxycarbonyl)piperazin-1-yl)phenyl)pyrrolidine-1,3-dicarboxylate Procedure Step a: To a mixture of dimethyl (3S,4S)-4-phenylpyrrolidine-1,3-dicarboxylate (400 mg) and 15 AgOTf (781 mg) in DCM (13 mL) was added AcOH (46 mg, 4 L) and I2 (771 mg) at 0 °C. After 1.5 hr, the mixture was allowed to warm to RT and stir for 2 hr. After this time, the mixture was poured into dilute, aq. NH4OH (50 mL) and sat. aq. sodium thiosulfate (10 mL). The mixture was filtered, and the filtrate was extracted with DCM (3x). The combined organic extracts were washed with brine, dried over Na2SO4, filtered, and concentrated. The residue was purified by column20 chromatography (SiO2; 10 to 50% EtOAc in heptane) to afford dimethyl (3S,4S)-4-(4- PAT059646-WO-PCT 5 iodophenyl)pyrrolidine-1,3-dicarboxylate (465 mg) as a colorless syrup. 1H NMR (400 MHz, CDCl3) 7.65 – 7.59 (m, 2H), 6.94 – 6.87 (m, 2H), 3.90 – 3.77 (m, 3H), 3.75 (s, 4H), 3.65 (q, J = 7.0 Hz, 1H), 3.45 (s, 3H), 3.43 – 3.35 (m, 1H). Step b: Dimethyl (3S,4S)-4-(4-iodophenyl)pyrrolidine-1,3-dicarboxylate (105 mg), tert-butyl 10 piperazine-1-carboxylate (53 mg), Cs2CO3 (219 mg), Pd2(dba)3 (7.4 mg), and XPhos (7.7 mg) were dissolved in toluene (2.7 mL) and sparged with nitrogen for 5 min. The mixture was then stirred at 110°C for 20 h, after which time it was filtered and concentrated. The residue was purified by column chromatography (SiO2; 30 to 60% EtOAc in heptane) to afford dimethyl (3S,4S)-4-(4-(4-(tert-butoxycarbonyl)piperazin-1-yl)phenyl)pyrrolidine-1,3-dicarboxylate (74 mg) 15 as a yellow foam. LC/MS (FinalAnalysis-Basic): M/Z = 448.5 [M+H]+. 1H NMR (400 MHz, CDCl3) 7.07 (d, J = 8.4 Hz, 2H), 6.93 – 6.78 (m, 2H), 3.86 – 3.80 (m, 1H), 3.79 – 3.71 (m, 5H), 3.70 – 3.62 (m, 2H), 3.58 (s, 4H), 3.45 – 3.40 (m, 3H), 3.40 – 3.32 (m, 1H), 3.15 – 3.05 (m, 4H), 1.48 (s, 9H). 20 Intermediate 17 (3S,4S)-1-(methoxycarbonyl)-4-(4-(4-methylpiperazin-1-yl)phenyl)pyrrolidine-3-carboxylic acid Procedure 25 Step a: A solution of dimethyl (3S,4S)-4-(4-(4-(tert-butoxycarbonyl)piperazin-1- yl)phenyl)pyrrolidine-1,3-dicarboxylate (74 mg) and trimethyltin hydroxide (60 mg) in DCE (1.1 mL) was stirred at 85 °C for 5 hr. After this time, additional trimethyltin hydroxide (30 mg) was added and stirring continued for 17 hr. After this time, additional trimethyltin hydroxide (15 mg) was added and the reaction stirred for an additional 8 hr, for a total of 30 hr at 85 °C. The mixture 30 was then concentrated and diluted with 1N aq. HCl and EtOAc. The aq. layer was extracted with EtOAc (3x), and the combined organic extracts were washed with brine, passed through a phase separator and concentrated. The residue was purified by column chromatography (SiO2; 0 to 75% 3:1 EtOAc/EtOH in heptane) to afford (3S,4S)-4-(4-(4-(tert-butoxycarbonyl)piperazin-1- yl)phenyl)-1-(methoxycarbonyl)pyrrolidine-3-carboxylic acid (53 mg) as a white solid. LC/MS 35 (FinalAnalysis-Basic): M/Z = 434.4 [M+H]+.1H NMR (400 MHz, DMSO) 12.20 (s, 1H), 7.03 (d, PAT059646-WO-PCT 5 J = 8.4 Hz, 2H), 6.91 – 6.82 (m, 2H), 3.73 – 3.50 (m, 9H), 3.43 (t, J = 5.1 Hz, 4H), 3.05 (t, J = 5.3 Hz, 4H), 1.41 (s, 9H). Intermediate 18 (3S,4S)-4-(4-(4-(2-hydroxyacetyl)piperazin-1-yl)phenyl)-1-(methoxycarbonyl)pyrrolidine-3- 10 carboxylic acid Procedure Step a: To a solution of Intermediate 17 (76 mg) in DCM (1.3 mL) was added TFA (0.4 mL) and the solution was allowed to react at RT for 2 hr. The reaction mixture was concentrated, and the 15 resulting residue was redissolved in DCM and concentrated to dryness; this process was repeated (3x). The resulting material was treated with sat. aq. NaHCO3 and extracted with DCM (3x). The combined organic extracts were passed through a phase separator and concentrated to afford dimethyl (3S,4S)-4-(4-(piperazin-1-yl)phenyl)pyrrolidine-1,3-dicarboxylate (68 mg) as a yellow residue. LC/MS (RXNMON-Basic) M/Z = 348.5 [M+H]+. 20 Step b: To solution of dimethyl (3S,4S)-4-(4-(piperazin-1-yl)phenyl)pyrrolidine-1,3-dicarboxylate (30 mg), 2-hydroxyacetic acid (6.8 mg), and DIPEA (32.9 mg, 44.4 L) in acetonitrile (800 μL) was added HATU (35.5 mg). The reaction stirred at RT for 45 min., after which time the reaction was diluted with sat. aq. NaHCO3 and EtOAc. The aq. layer was extracted with EtOAc (3x) and 25 the combined organic extracts were washed with brine, passed through a phase separator and concentrated to afford crude dimethyl (3S,4S)-4-(4-(4-(2-hydroxyacetyl)piperazin-1- yl)phenyl)pyrrolidine-1,3-dicarboxylate (37 mg) as a pale yellow solid, which was taken as-is to the next step. LC/MS (RXNMON-Basic) M/Z = 406.5 [M+H]+. 30 Step c: To a solution of crude dimethyl (3S,4S)-4-(4-(4-(2-hydroxyacetyl)piperazin-1- yl)phenyl)pyrrolidine-1,3-dicarboxylate (37 mg) was added trimethyltin hydroxide (61 mg) in DCE (800 μL). The reaction was stirred at 85°C for 3 days, after which time the reaction mixture was concentrated and the residue treated with aq.1N HCl and EtOAc. The aq. layer was extracted PAT059646-WO-PCT 5 with EtOAc, and the combined organic extracts were washed with brine, passed through a phase separator, and concentrated. The residue was purified by preparatory HPLC (C18, 10 to 30% acetonitrile in water (+ 0.1% formic acid)) to afford (3S,4S)-4-(4-(4-(2-hydroxyacetyl)piperazin-1- yl)phenyl)-1-(methoxycarbonyl)pyrrolidine-3-carboxylic acid (12 mg) as a white solid. LC/MS (FinalAnalysis-Basic) M/Z = 392.5 [M+H]+.1H NMR (400 MHz, DMSO) 7.14 (s, 4H), 4.15 – 4.13 10 (m, 3H), 3.72 – 3.65 (m, 4H), 3.62 (d, J = 3.9 Hz, 4H), 3.58 – 3.54 (m, 4H), 3.45 – 3.33 (m, 2H), 3.22 (m, 2H), 2.07 (s, 2H). Intermediate 19 (2R,3R)-3-(4-(4-methylpiperazin-1-yl)phenyl)tetrahydrofuran-2-carboxylic acid 15 Procedure Step a: To a solution of (2R,3R)-3-phenyltetrahydrofuran-2-carboxylic acid (10 g) in EtOH (260 mL) was added conc. H2SO4 (226 mg, 416 L). The reaction was heated at 90°C for 5 hr. The reaction mixture was concentrated and diluted in EtOAc and sat. NaHCO3. The aq. layer was 20 extracted with EtOAc (2x), and the combined organic extracts were washed with brine, dried over Na2SO4, filtered, and concentrated to afford ethyl (2R,3R)-3-phenyltetrahydrofuran-2-carboxylate (11.2 g) as a clear, yellow oil.1H NMR (400 MHz, CDCl3) 7.31 – 7.19 (m, 5H), 4.65 (d, J = 8.1 Hz, 1H), 4.43 (ddd, J = 8.5, 7.4, 5.0 Hz, 1H), 4.03 (q, J = 8.0 Hz, 1H), 3.86 – 3.64 (m, 3H), 2.45 – 2.33 (m, 2H), 0.82 (t, J = 7.1 Hz, 3H). LC/MS (FinalAnalysis-Basic) M/Z = 221.1 [M+H]+. 25 Step b: To a solution of ethyl (2R,3R)-3-phenyltetrahydrofuran-2-carboxylate (989 mg) and Ag(OTf) (2.31 g) in DCM (40 mL) and AcOH (135 mg, 129 L) at 0 °C was added I2 (2.28 g). The reaction mixture was allowed to warm to RT and stir for 1.5 hr, after which time it was poured into PAT059646-WO-PCT 5 dilute aq. NH4OH and sat. aq. sodium thiosulfate. The resulting precipitate was filtered, and the filtrate was extracted with DCM (3x). The combined organic extracts were washed with brine, dried over Na2SO4, filtered, and concentrated. The residue was purified by column chromatography (SiO2; 0-15% EtOAc in heptane) to afford ethyl (2R,3R)-3-(4- iodophenyl)tetrahydrofuran-2-carboxylate (909 mg) as a clear oil.1H NMR (400 MHz, CDCl3) 10 7.63 – 7.58 (m, 2H), 7.00 – 6.95 (m, 2H), 4.62 (d, J = 7.8 Hz, 1H), 4.40 (td, J = 8.4, 4.2 Hz, 1H), 4.05 – 3.98 (m, 1H), 3.86 (dq, J = 10.8, 7.2 Hz, 1H), 3.75 (dq, J = 10.8, 7.1 Hz, 1H), 3.67 (q, J = 7.9 Hz, 1H), 2.43 – 2.23 (m, 2H), 0.90 (s, 3H). LC/MS (RXNMON-basic) M/Z = 347.1 [M+H]+. Step c: A mixture of (2R,3R)-3-(4-iodophenyl)tetrahydrofuran-2-carboxylate (300 mg), tert-butyl 15 piperazine-1-carboxylate (254 mg), Cs2CO3 (1.11 g), Pd2dba3 (31.2 mg), and XPhos (32.5 mg) in toluene (12 mL) was sparged with nitrogen for 5 min. and heated at 110°C for 20 hr. The reaction mixture was filtered over a pad of Celite® and concentrated. The residue was purified by column chromatography (SiO2; 25-45% EtOAc in hepane) to afford tert-butyl 4-(4-((2R,3R)-2- (ethoxycarbonyl)tetrahydrofuran-3-yl)phenyl)piperazine-1-carboxylate (109 mg) as an off-white 20 solid.1H NMR (400 MHz, MeOD) 7.15 – 7.10 (m, 2H), 6.94 – 6.89 (m, 2H), 4.60 (d, J = 8.1 Hz, 1H), 4.32 (td, J = 7.9, 4.6 Hz, 1H), 3.98 (td, J = 8.2, 7.4 Hz, 1H), 3.84 – 3.64 (m, 3H), 3.55 (t, J = 5.2 Hz, 4H), 3.08 (dd, J = 6.3, 4.1 Hz, 4H), 2.37 – 2.24 (m, 2H), 1.48 (s, 9H), 0.83 (t, J = 7.1 Hz, 3H). LC/MS (FinalAnalysis-Basic) M/Z = 405.5 [M+H]+. 25 Step d: To a solution of tert-butyl 4-(4-((2R,3R)-2-(ethoxycarbonyl)tetrahydrofuran-3- yl)phenyl)piperazine-1-carboxylate (109 mg) in DCM (2 mL) was added TFA (500 μL). The reaction was stirred at RT for 30 min. The reaction mixture was then concentrated, diluted in DCM, and concentrated again; this process was repeated (3x). The remaining residue was dissolved in DCM and washed with sat. NaHCO3. The aq. layer was extracted with DCM (3x), and 30 the combined organic extracts were passed through a phase separator, and concentrated to afford ethyl (2R,3R)-3-(4-(piperazin-1-yl)phenyl)tetrahydrofuran-2-carboxylate (93 mg). LC/MS (RXMON-Basic) M/Z = 305.4 [M+H]+. Step e: To a solution of ethyl (2R,3R)-3-(4-(piperazin-1-yl)phenyl)tetrahydrofuran-2-carboxylate 35 (93 mg) in MeOH (2.5 mL) was added aq. formaldehyde (65.4 mg, 60 L, 37% wt). The reaction was stirred for 30 min. and then sodium triacetoxyhydroborate (114 mg) was added. The reaction was stirred at RT for 30 min. and was then diluted with brine and sat. NaHCO3 and the aq. layer was extracted with DCM (3x). The combined organic layers were passed through a phase separator and concentrated to afford ethyl (2R,3R)-3-(4-(4-methylpiperazin-1- 40 yl)phenyl)tetrahydrofuran-2-carboxylate (87 mg). PAT059646-WO-PCT 5 Step f: To a solution ethyl (2R,3R)-3-(4-(4-methylpiperazin-1-yl)phenyl)tetrahydrofuran-2- carboxylate (87 mg) in THF (800 μL), MeOH (800 μL), and water (800 μL) was added aq. 1N LiOH (23 mg, 1 mL). The reaction stirred at RT for 24 hr, after which time additional aq.1N LiOH (5.7 mg, 240 μL) was added, and the reaction stirred overnight for 16 hr. The reaction stirred for a total of 40 hr. The volatiles were removed and the mixture was purified by column10 chromatography (C18; 0 to 15% acetonitrile in water (+0.1% NH4OH)) to afford (2R,3R)-3-(4-(4- methylpiperazin-1-yl)phenyl)tetrahydrofuran-2-carboxylic acid (72 mg) as a white solid after lyophilization.1H NMR (400 MHz, DMSO) 7.12 – 7.06 (m, 2H), 6.76 – 6.69 (m, 2H), 4.17 – 4.11 (m, 1H), 4.11 – 4.00 (m, 1H), 3.79 – 3.69 (m, 1H), 3.36 – 3.32 (m, 3H), 3.08 – 2.99 (m, 4H), 2.43 (t, J = 5.0 Hz, 4H), 2.20 (s, 3H), 1.99 – 1.87 (m, 1H). LC/MS (RXNMON-basic) M/Z = 291.5 [M+H]+. 15 Intermediate 20 (2R,3S)-1-(tert-butoxycarbonyl)-3-phenylazetidine-2-carboxylic acid Procedure 20 Step a: (2RS,3SR)-1-(tert-butoxycarbonyl)-3-phenylazetidine-2-carboxylic acid (820 mg) was purified by chiral SFC (Prep_SFC6 method) to yield (2R,3S)-1-(tert-butoxycarbonyl)-3- phenylazetidine-2-carboxylic acid as the second eluting isomer. LC/MS (RXNMON-Acidic): M/Z = 276.3 [M–H] 25 Intermediate 21 (1R,2S)-2-phenylcyclopropane-1-carboxylic acid Procedure Step a: Racemic cis 2-phenylcyclopropane-1-carboxylic acid (243 mg) was purified by SFC 30 (Prep_SFC4 method) to yield (1R,2S)-2-phenylcyclopropane-1-carboxylic acid (111 mg) as the first eluting isomer, a white solid. LC/MS (FinalAnalysis-Basic) M/Z = 163.0 [M+H]+.1H NMR (400 PAT059646-WO-PCT 5 MHz, CDCl3) 7.29 – 7.18 (m, 5H), 2.69 – 2.59 (m, 1H), 2.06 (ddd, J = 9.2, 7.7, 5.6 Hz, 1H), 1.68 (ddd, J = 7.7, 5.6, 5.1 Hz, 1H), 1.38 (ddd, J = 8.7, 7.7, 5.1 Hz, 1H). Intermediate 22 (1R,2S)-2-phenylcyclobutane-1-carboxylic acid 10 Procedure Step a: Racemic cis 2-phenylcyclobutane-1-carboxylic acid (250 mg) was purified by SFC (Prep_SFC5 method) to yield (1R,2S)-2-phenylcyclobutane-1-carboxylic acid (124 mg) as the second eluting isomer, a yellow oil. LC/MS (FinalAnalysis-Basic) M/Z = 175.1 [M+H]+.1H NMR 15 (400 MHz, CDCl3) 7.31 – 7.13 (m, 5H), 4.03 – 3.88 (m, 1H), 3.58 – 3.41 (m, 1H), 2.71 – 2.53 (m, 1H), 2.32 (m, 2H), 2.26 – 2.13 (m, 1H). Intermediate 23 onyl)(methyl)amino)-3-methylpent-4-enoic acid 20 Procedure Step a: A solution of (2S,3R)-2-(((S)tert-butylsulfinyl)amino)-3-methylpent-4-enoic acid (250 mg) in DCM (3.6 mL) was cooled to 0 °C and 4N HCl in dioxane (195 mg, 1.34 mL) was added. The solution was stirred at RT for 50 min. The mixture was then concentrated to afford (2S,3R)-2- 25 amino-3-methylpent-4-enoic acid hydrochloride (165.0 mg) as a white solid. 1H NMR (400 MHz, DMSO) 13.81 (s, 1H), 8.35 (s, 2H), 5.81 (ddd, J = 17.4, 10.4, 7.2 Hz, 1H), 5.32 – 5.02 (m, 2H), 3.87 (d, J = 4.5 Hz, 1H), 2.89 – 2.72 (m, 1H), 1.10 (d, J = 6.9 Hz, 3H). PAT059646-WO-PCT 5 Step b: A solution of (2S,3R)-2-amino-3-methylpent-4-enoic acid hydrochloride (158 mg) in 1,4- dioxane (1.91 mL) was cooled to 0 °C and 1N aq. NaOH (95 mg, 2.4 mL) was added. A solution of Boc2O (245 mg, 263 L) was added to the reaction at 0 °C and the reaction was allowed to warm to RT and stir for 42 hr. The reaction pH was then adjusted to 10 with aq.1N NaOH. The resulting basic aq. mixture was washed with Et2O. The remaining aq. layer was acidified to pH 2 10 with aqueous 1N HCl and extracted with 15 mL portions of EtOAc (3x). The combined organic extracts were dried over Na2SO4, filtered, and concentrated to afford (2S,3R)-2-((tert- butoxycarbonyl)amino)-3-methylpent-4-enoic acid (210 mg) as a colorless oil. 1H NMR (400 MHz, DMSO) 12.50 (s, 1H), 6.94 (d, J = 8.7 Hz, 1H), 5.75 (ddd, J = 17.5, 10.4, 7.3 Hz, 1H), 5.08 – 4.96 (m, 2H), 3.93 (dd, J = 8.7, 6.3 Hz, 1H), 2.59 (p, J = 6.9 Hz, 1H), 1.38 (s, 9H), 0.96 (d, 15 J = 6.9 Hz, 3H). Step c: A solution of (2S,3R)-2-((tert-butoxycarbonyl)amino)-3-methylpent-4-enoic acid (210 mg) in THF (5.9 mL) was cooled to 0 °C. NaH (110 mg, 60% wt.) was added in portions. Following complete addition, MeI (1.04 g) was added and the reaction was allowed to warm to RT and stir 20 for 22 hr. Additional NaH (44 mg, 60% wt.) and MeI (520 mg, 229 L) were added and stirring at RT was continued for 16 hr. Et2O was added and the reaction was quenched with water (15 mL). The resulting organic phase was washed with additional water (15 mL). The combined aq. extracts were acidified to a pH of 3 with aq. citric acid and extracted with 15 mL portions of EtOAc (3x). Combined organic extracts were washed with aq. sodium thiosulfate solution, dried over25 Na2SO4, filtered, and concentrated to afford (2S,3R)-2-((tert-butoxycarbonyl)(methyl)amino)-3- methylpent-4-enoic acid (150 mg) as an oil.1H NMR (400 MHz, DMSO) 12.60 (s, 1H), 5.80 (m, 1H), 5.17 – 4.96 (m, 2H), 4.26 (dd, J = 97.9, 10.1 Hz, 1H), 2.76 (s, 3H), 2.74 – 2.63 (m, 1H), 1.40 (d, J = 8.7 Hz, 9H), 0.91 (dd, J = 13.2, 6.8 Hz, 3H). 30 Intermediate 24 N-(tert-butoxycarbonyl)-N-methyl-L-alloisoleucine PAT059646-WO-PCT 5 Procedure Step a: To a solution of (2S,3R)-2-(((S)-tert-butylsulfinyl)amino)-3-methylpent-4-enoic acid (250 mg) in MeOH (5 mL) was charged Pd(OH)2 on carbon (151 mg, 20% wt.). The reaction vessel was evacuated and backfilled with H2. The reaction was stirred under an atmosphere of H2 at RT for 22 hr. The reaction was filtered and concentrated to afford ((S)-tert-butylsulfinyl)-L- 10 alloisoleucine (252 mg) as a reddish-brown oil. LC/MS (RXMON-Acidic method): M/Z = 236.0 [M+H]+. Step b: To a solution of ((S)-tert-butylsulfinyl)-L-alloisoleucine (85.6 mg) in DCM (1.2 mL). The solution was cooled to 0 °C and 4N HCl in 1,4-dioxane (455 L) was added. The reaction was 15 stirred at RT for 30 min., after which time the solution was concentrated to afford L-alloisoleucine hydrochloride (67 mg) as a white solid.1H NMR (400 MHz, DMSO) 13.79 (s, 1H), 8.25 (s, 2H), 3.83 (s, 1H), 1.94 (ddd, J = 13.0, 9.2, 5.6 Hz, 1H), 1.52 (dq, J = 13.8, 6.8 Hz, 1H), 1.25 – 1.08 (m, 1H), 0.97 – 0.85 (m, 6H). 20 Step c: To a vial charged with L-alloisoleucine hydrochloride (67 mg) was added 1,4-dioxane (759 L). The reaction solution was cooled to 0 °C and aq.1N NaOH (949 L) was added. Following this, Boc2O (99 mg) was added, and the reaction was allowed to warm to RT. Additional aq.1N NaOH (949 L) and Boc2O (99 mg) were added. The reaction stirred for total of 29 hr, after which time the pH was adjusted to 10 with aq.1N NaOH. The mixture was washed with Et2O and the 25 aq. layer was then acidified to pH 2 with aq.1N HCl and extracted with EtOAc (3x). The combined organic extracts were dried over Na2SO4, filtered, and concentrated to afford (tert- butoxycarbonyl)-L-alloisoleucine (85.6 mg) as a white solid.1H NMR (400 MHz, DMSO) 12.45 (s, 1H), 6.85 (d, J = 8.9 Hz, 1H), 4.00 (dd, J = 8.9, 5.0 Hz, 1H), 1.86 – 1.76 (m, 1H), 1.39 (s, 9H), 1.36 – 1.29 (m, 1H), 1.14 (dt, J = 14.2, 7.3 Hz, 1H), 0.88 – 0.82 (m, 6H). 30 Step d: To a solution of (tert-butoxycarbonyl)-L-alloisoleucine (70 mg) in THF (2.0 mL) at 0 °C was sequentially added MeI (344 mg, 151 L) and NaH (73 mg, 60% wt.). The reaction was allowed to warm to RT and stir for 28 hr. The reaction was then diluted with Et2O and washed with water (15 mL, 2x). Combined aq. extracts were acidified with citric acid (pH 3) and extracted 35 with EtOAc. Combined organic extracts were dried over Na2SO4, filtered, and concentrated. This residue was purified by column chromatography (C18; 10 to 100% acetonitrile in water + 0.1% formic acid) to afford N-(tert-butoxycarbonyl)-N-methyl-L-alloisoleucine (40 mg) as a colorless oil after lyophilization.1H NMR (400 MHz, DMSO) 12.64 (s, 1H), 4.20 (dd, J = 100.9, 10.0 Hz, 1H), 2.73 (d, J = 2.2 Hz, 3H), 2.01 – 1.83 (m, 1H), 1.44 (d, J = 7.7 Hz, 1H), 1.39 (d, J = 9.2 Hz, 9H), 40 1.09 (dq, J = 14.9, 7.6 Hz, 1H), 0.88 (t, J = 7.4 Hz, 3H), 0.78 (dd, J = 9.6, 6.7 Hz, 3H). PAT059646-WO-PCT 5 Intermediate 25 N-(tert-butoxycarbonyl)-N,O-dimethyl-L-threonine Procedure Step a: To a solution of methyl (tert-butoxycarbonyl)-L-threoninate (3 g) in acetonitrile (130 mL) 10 at 0 °C under nitrogen was added Ag2O (13.4 g) and MeI (14.1 g, 6.19 mL). The reaction allowed to warm to RT and stir for 4 days. After this time, the reaction was filtered, rinsed with acetonitrile, and concentrated. The residue was purified by column chromatography (SiO2; 5 to 30% EtOAc in heptane) to afford methyl N-(tert-butoxycarbonyl)-O-methyl-L-threoninate (2.39 g) as a colorless oil.1H NMR (400 MHz, CDCl3) 5.21 (d, J = 9.5 Hz, 1H), 4.27 (dd, J = 9.5, 2.4 Hz, 1H), 3.90 (qd, 15 J = 6.3, 2.4 Hz, 1H), 3.76 (s, 3H), 3.28 (s, 3H), 1.45 (s, 9H), 1.20 (d, J = 6.3 Hz, 3H). Step b: To a solution of N-(tert-butoxycarbonyl)-O-methyl-L-threoninate (2.39 g) in THF (16 mL) and MeOH (16 mL) at 0 °C was added aq.1.0N LiOH (810 mg, 34 mL). The reaction was allowed to warm to RT and stir for 2.5 hr. After this time, the mixture was cooled to 0 °C, 1N aq. HCl (21 20 mL) was added, and the mixture was concentrated. The remaining aq. mixture was extracted with EtOAc (3x), the organic layers were passed through a phase separator. Filtrate was concentrated to afford N-(tert-butoxycarbonyl)-O-methyl-L-threonine (2.18 g) as a white solid. 1H NMR (400 MHz, CDCl3) 5.36 – 5.25 (m, 1H), 4.41 – 4.31 (m, 1H), 4.02 – 3.91 (m, 1H), 3.46 – 3.37 (m, 3H), 1.46 (s, 9H), 1.21 (d, J = 6.4 Hz, 3H). 25 Step c: To a solution of N-(tert-butoxycarbonyl)-O-methyl-L-threonine (500 mg) in THF (20 mL) at 0 °C under a nitrogen atmosphere was added NaH (176 mg, 60% wt.) in one portion. After 30 min., MeI (1.53 g, 670 L) and DMF (332 L) were added. The solution stirred at 0 °C for 2 hr then was allowed to warm to RT and stir for 20 hr. The reaction mixture was quenched with water, 30 diluted with aq. HCl and brine. The aq. layer was extracted with EtOAc (3x). The combined organic extracts were passed through a phase separator and concentrated. The residue was purified by column chromatography (SiO2; 5 to 35% 3:1 EtOAc/EtOH (containing 1% AcOH) in heptanes) to afford N-(tert-butoxycarbonyl)-N,O-dimethyl-L-threonine (278 mg) as white solid.1H NMR (400 MHz, CDCl3) 4.80 – 4.73 (m, 0.7H), 4.71 – 4.63 (m, 0.3H), 4.08 – 4.02 (m, 0.7H), 4.02 – 3.93 PAT059646-WO-PCT 5 (m, 0.3H), 3.33 (dm, J = 1.1 Hz, 3H), 2.96 (dm, J = 4.5 Hz, 3H), 1.51 – 1.42 (m, 9H), 1.23 – 1.15 (m, 3H). Intermediate 26 benzyl 4-(5-((61R,65S,66S,4S)-4-((tert-butoxycarbonyl)amino)-11-ethyl-10,10-dimethyl-5,7-dioxo-10 25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana- 2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1- carboxylate 15 Procedure Step a: To a solution of Intermediate 3a (11.83 g) in acetonitrile (45.8 mL) under nitrogen atmosphere was added water (900 mg, 900 μL), Et3N (2.9 g, 4 mL), and LiBr (5 g). The resulting 20 mixture was stirred at RT for 16 hr, upon which time additional LiBr (1 g) was added, and the PAT059646-WO-PCT 5 mixture was then heated to 50 °C for 4 hr. After this time, the reaction was cooled to RT and stirred for 48 hr, during which time a precipitate formed. The first crop of solids was collected via vacuum filtration and rinsed with cold acetonitrile. The filtrate was concentrated to half of its volume and cooled to 5 °C to precipitate additional product. This second crop was isolated via vacuum filtration and combined with the first crop to afford (2S)-3-(3-(2-(5-(4-10 ((benzyloxy)carbonyl)piperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-1-ethyl-3-(3-hydroxy- 2,2-dimethylpropyl)-1H-indol-5-yl)-5-((triisopropylsilyl)oxy)phenyl)-2-((tert- butoxycarbonyl)amino)propanoic acid (10.5 g) as a white solid after drying. LC/MS (RXMON acidic_nonpolar): M/Z = 1020.6 [M+H]+. 15 Step b, part 1: To Intermediate 1 (220 mg) in DCM (35 mL) was added TFA (35 mL) and the solution was stirred at RT for 2 hr, after which time, the reaction mixture was concentrated and the residue was azeotropically dried with heptane and left to dry in vacuo for 16 hr to afford methyl (1S,2S,6R)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylate, trifluoroacetate salt (3.35 g) which was advanced directly to subsequent amide coupling. 20 Step b, part 2: Crude methyl (1S,2S,6R)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylate, trifluoroacetate salt was reconstituted in DMF (30 mL) and (2S)-3-(3-(2-(5-(4- ((benzyloxy)carbonyl)piperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-1-ethyl-3-(3-hydroxy- 2,2-dimethylpropyl)-1H-indol-5-yl)-5-((triisopropylsilyl)oxy)phenyl)-2-((tert- 25 butoxycarbonyl)amino)propanoic acid (10.5 g) was added. The reaction apparatus was purged with nitrogen (3x) and the solution was cooled to 0 °C. DIPEA (4.25 g, 5.73 mL) was added, and the mixture was stirred for 5 min., after which time HATU (3.93 g) was added, and the solution was allowed to stir for 5 min. at °C and then warm to RT. After stirring at RT for 1 hr, ice (75 mL) was added to triturate the product. After melting, the reaction suspension was filtered and the30 retained solids were washed with water (75 mL) to afford methyl (1S,2S,6R)-4-((2S)-3-(3-(2-(5- (4-((benzyloxy)carbonyl)piperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-1-ethyl-3-(3-hydroxy- 2,2-dimethylpropyl)-1H-indol-5-yl)-5-((triisopropylsilyl)oxy)phenyl)-2-((tert- butoxycarbonyl)amino)propanoyl)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylate (11.36 g) as an off-white solid after drying. LC/MS (FinalAnalysis-Acidic): M/Z = 1159.9 [M+H]+. 35 Step c: To a solution of methyl (1S,2S,6R)-4-((2S)-3-(3-(2-(5-(4-((benzyloxy)carbonyl)piperazin- 1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-1H-indol-5-yl)- 5-((triisopropylsilyl)oxy)phenyl)-2-((tert-butoxycarbonyl)amino)propanoyl)-3,4- diazabicyclo[4.1.0]heptane-2-carboxylate (10.82 g) in acetonitrile (200 mL) under nitrogen 40 atmosphere was added water (3 g, 3 mL), Et3N (2.363 g, 3.25 mL), and LiBr (4.055 g). The solution was then stirred at 50 °C for 16 hr, after which time additional LiBr (2 g) and water (1 g, PAT059646-WO-PCT 5 1 mL) were added, and the mixture was allowed to cool to RT and stir for 56 hr. After 72 hr, the reaction was concentrated to half of its original volume and EtOAc (300 mL) was added. The resulting mixture was washed with aq.0.5N HCl and the organic phase was dried over Na2SO4 and filtered. The filtrate was concentrated to afford (1S,2S,6R)-4-((2S)-3-(3-(2-(5-(4- ((benzyloxy)carbonyl)piperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-1-ethyl-3-(3-hydroxy-10 2,2-dimethylpropyl)-1H-indol-5-yl)-5-((triisopropylsilyl)oxy)phenyl)-2-((tert- butoxycarbonyl)amino)propanoyl)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylic acid (11.5 g) as a yellow solid, containing some impurities, which was carried forward to the next step as-is. LC/MS (FinalAnalysis-Basic-NonPolar): M/Z = 1144.7 [M+H]+. 15 Step d: To a solution of EDC (2.60 g), HOAt (1.85 g) and DMAP (664 mg) in DCM (500 mL) was added, via mechanical syringe driver, a solution of (1S,2S,6R)-4-((2S)-3-(3-(2-(5-(4- ((benzyloxy)carbonyl)piperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-1-ethyl-3-(3-hydroxy- 2,2-dimethylpropyl)-1H-indol-5-yl)-5-((triisopropylsilyl)oxy)phenyl)-2-((tert- butoxycarbonyl)amino)propanoyl)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylic acid (7.5 g) in 20 DMF (20 mL) at a rate of 3 mL / hr. Following complete addition, the reaction was washed with sat. aq. NaHCO3 solution (100 mL), water (100 mL, 3x) and the resulting organic phase was dried over Na2SO4. Filtration and concentration afforded a crude semi-solid which was purified by column chromatography (SiO2; 0 to 15% EtOAc/EtOH 3:1 in DCM) to afford benzyl 4-(5- ((61R,65S,66S,4S)-4-((tert-butoxycarbonyl)amino)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-25 ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (4.33 g) as a white solid.1H NMR (400 MHz, DMSO) 8.47 (d, J = 2.9 Hz, 1H), 7.95 (s, 1H), 7.61 (d, J = 8.6 Hz, 1H), 7.56 – 7.49 (m, 1H), 7.40 – 7.34 (m, 4H), 7.35 – 7.31 (m, 1H), 7.31 (s, 1H), 7.28 – 7.24 (m, 1H), 7.10 (d, J = 8.7 Hz, 1H), 7.06 – 7.01 (m, 1H), 6.79 (s, 1H), 5.11 (s, 2H), 5.00 30 – 4.91 (m, 1H), 4.91 – 4.84 (m, 1H), 4.28 – 4.08 (m, 4H), 3.75 (d, J = 10.9 Hz, 1H), 3.65 (d, J = 10.8 Hz, 1H), 3.57 (s, 4H), 3.31 – 3.21 (m, 5H), 3.14 (s, 3H), 2.93 – 2.81 (m, 1H), 2.77 – 2.68 (m, 1H), 2.67 – 2.56 (m, 2H), 1.38 – 1.34 (m, 3H), 1.33 (s, 9H), 1.29 – 1.27 (m, 2H), 1.26 – 1.18 (m, 5H), 1.13 – 1.06 (m, 18H), 0.97 – 0.91 (m, 3H), 0.88 – 0.83 (m, 4H), 0.44 (s, 2H), 0.36 – 0.28 (m, 1H). LC/MS (Peptide method): M/Z = 1126.6500 [M+H]+. 35 The following intermediates of Table 3 were synthesized using the above procedure or modifications of the above procedure using the corresponding hydropyridazine building block and Intermediate 3. Note: Axial chirality of compounds is as shown in structures in Table 3. 40 Table 3: Macrocycle intermediates PAT059646-WO-PCT 5 Intermediate 27 benzyl 4-(5-((61R,65S,66S,4S)-11-ethyl-10,10-dimethyl-4-((S)-3-methyl-2-((2R,3R)-N-methyl-3- phenyltetrahydrofuran-2-carboxamido)butanamido)-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-10 oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12- yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate
PAT059646-WO-PCT 5 Step a: To a solution of benzyl 4-(5-((61R,65S,66S,4S)-4-((tert-butoxycarbonyl)amino)-11-ethyl- 10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3- 10 yl)piperazine-1-carboxylate (248 mg) in DCM (1.6 mL) was added TFA (627 mg, 421 μL). The solution was stirred at RT for 1 hr. The reaction mixture was concentrated and the residue was triturated with Et2O to afford benzyl 4-(5-((61R,65S,66S,4S)-4-amino-11-ethyl-10,10-dimethyl-5,7- dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana- 2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1- 15 carboxylate trifluoroacetate salt (370 mg) as a yellow foam. LC/MS (RXNMON-Acidic): M/Z = 1027.6 [M+H]+. Step b: To a solution of benzyl 4-(5-((61R,65S,66S,4S)-4-amino-11-ethyl-10,10-dimethyl-5,7- dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-20 2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1- carboxylate trifluoroacetate salt (370 mg) in DMF (2 mL) a 0 °C under nitrogen, was added Boc- N-Me-L-valine (101 mg), DIPEA (285 mg, 384 μL) and COMU (189 mg). The reaction mixture was allowed to warm to RT and stir for 15 min. After this time, the reaction mixture was diluted with water and extracted with EtOAc. The EtOAc extract was washed with sat. aq. NaHCO3, brine, 25 dried over Na2SO4, filtered, and concentrated. The residue was purified by column PAT059646-WO-PCT 5 chromatography (SiO2; 0 to 100% EtOAc in heptane) to afford benzyl 4-(5-((61R,65S,66S,4S)-4- ((S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-methylbutanamido)-11-ethyl-10,10-dimethyl-5,7- dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana- 2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1- carboxylate (280 mg) as a foamy solid. LC/MS (RXNMON-Acidic): M/Z = 1240.4 [M+H]+. 10 Step c: To a solution of benzyl 4-(5-((61R,65S,66S,4S)-4-((S)-2-((tert- butoxycarbonyl)(methyl)amino)-3-methylbutanamido)-11-ethyl-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate 15 (280 mg) in DCM (2 mL) was added TFA (411 μL). The reaction solution was stirred at RT for 1 hr, after which time it was concentrated to dryness. The residue was re-dissolved in DCE, concentrated, and the remaining residue was triturated with Et2O to afford benzyl 4-(5- ((61R,65S,66S,4S)-11-ethyl-10,10-dimethyl-4-((S)-3-methyl-2-(methylamino)butanamido)-5,7- dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-20 2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1- carboxylate trifluoroacetate salt (300 mg) as a yellow solid. LC/MS (RXNMON-Acidic): M/Z = 1140.6 [M+H]+. Step d: To a solution of benzyl 4-(5-((61R,65S,66S,4S)-11-ethyl-10,10-dimethyl-4-((S)-3-methyl-2-25 (methylamino)butanamido)-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)- indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1- methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate trifluoroacetate salt (270 mg) in DMF (1.5 mL) under nitrogen was added (2R,3R)-3-phenyltetrahydrofuran-2-carboxylic acid (62 mg) followed by DIPEA (276 mg, 372 μL) and HATU (162 mg). After stirring at RT for 1 hr, the reaction mixture 30 was diluted with MeOH and purified by column chromatography (C18; 10 to 100% acetonitrile in water + 0.1% formic acid) to afford benzyl 4-(5-((61R,65S,66S,4S)-11-ethyl-10,10-dimethyl-4-((S)- 3-methyl-2-((2R,3R)-N-methyl-3-phenyltetrahydrofuran-2-carboxamido)butanamido)-5,7-dioxo- 25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana- 2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1- 35 carboxylate (231 mg). HRMS: M/Z = 1314.7500 [M+H]+. The following intermediates of Table 4 were synthesized using the above procedure or modifications of the above procedure. 40 Table 4: Cbz-protected macrocycle intermediates PAT059646-WO-PCT 5 Intermediate 29 (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(piperazin-1- yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)- indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- 10 oxobutan-2-yl)-N-methyl-3-phenyltetrahydrofuran-2-carboxamide Step a: To a solution of Intermediate 27 (130 mg) in THF (8 mL) and EtOH (3 mL) was added 15 Pd(OH)2 on carbon (69 mg, 10% wt.). The reaction mixture was purged with H2 and stirred under an H2 atmosphere at RT for 30 min., after which time Celite® and water were added. The mixture was filtered, and the filter cake was washed with methanol (10 mL). The combined filtrate was concentrated, and the residue was purified by column chromatography (C18; 10 to 100% acetonitrile in water + 0.1% TFA). The product fraction was concentrated, naturalized with sat. PAT059646-WO-PCT 5 aq. NaHCO3 and extracted with EtOAc. The EtOAc extract was concentrated to afford (2R,3R)- N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)- N-methyl-3-phenyltetrahydrofuran-2-carboxamide (110 mg) as a white foam. LC/MS 10 (FinalAnalysis-Acidic): M/Z = 1180.4 [M+H]+. The following intermediates of Table 5 were synthesized using the above procedure or modifications of the above procedure. 15 Table 5: Cbz-protected macrocycle intermediates Intermediate 31 (S)-2-((tert-butoxycarbonyl)amino)-3-(2-fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5- ((triisopropylsilyl)oxy)phenyl)propanoate 20 PAT059646-WO-PCT 5 Step a: To a suspension of zinc dust (1.21 g) in DMF (20 mL) under nitrogen atmosphere was added I2 (141 mg). The mixture was stirred for 15 min., after which time methyl (R)-2-((tert- butoxycarbonyl)amino)-3-iodopropanoate (1.83 g) and I2 (141 mg) were sequentially added. The mixture was allowed to stir at RT for 1 hr. After this time, a solution of 4-(benzyloxy)-1-fluoro-2- iodobenzene (1.22 g) in DMF (5 mL), Pd2(dba)3 (85 mg), and SPhos (153 mg) were sequentially 10 added. The mixture was allowed to stir at RT for 15 min. and then 50 °C for 1 hr. The reaction was diluted with water (150 mL) and EtOAc (50 mL) and the suspension was filtered. The EtOAc extract was washed with water (15 mL, 3x), brine, passed through a phase separator, and concentrated. The resulting residue was purified by column chromatography (SiO2; 0 to 20% EtOAc in heptane) to afford methyl (S)-3-(5-(benzyloxy)-2-fluorophenyl)-2-((tert- 15 butoxycarbonyl)amino)propanoate (1.23 g) as a yellow oil. LC/MS (FinalAnalysis-Acidic): M/Z = 304.1 [M-Boc+H]+. Step b: To a solution of methyl (S)-3-(5-(benzyloxy)-2-fluorophenyl)-2-((tert- butoxycarbonyl)amino)propanoate (1.23 g) in MeOH (15 mL) under nitrogen atmosphere was 20 charged Pd(OH)2 on carbon (2.14 g, 10% wt.). The mixture was purged with H2 and allowed to stir under a H2 atmosphere for 2 hr, after which time the mixture was filtered and concentrated to afford crude methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(2-fluoro-5-hydroxyphenyl)propanoate (1.014 g) which was used without further purification. LC/MS (RXMON-Acidic): M/Z = 214.2 [M- Boc+H]+. 25 Step c: To a solution of crude methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(2-fluoro-5- hydroxyphenyl)propanoate (1.01 g) in 2-MeTHF (10 mL) under nitrogen atmosphere was sequentially charged imidazole (621 mg) and TIPS-Cl (880 mg, 976 μL). The reaction mixture was left to stir at RT for 16 hr. After this time, the mixture was concentrated and the resulting 30 residue purified by column chromatography (SiO2; 0 to 20% EtOAc in heptane) to afford methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(2-fluoro-5-((triisopropylsilyl)oxy)phenyl)propanoate (1.745 g), with some impurities, as a colorless oil.1H-NMR (400 MHz, CDCl3) 6.86 (t, J = 9.1 Hz, 1H), 6.75 – 6.68 (m, 1H), 6.64 (dd, J = 6.2, 3.0 Hz, 1H), 5.01 (d, J = 8.3 Hz, 1H), 4.56 (q, J = 6.6 Hz, 1H), 3.72 (s, 3H), 3.10 – 3.00 (m, 2H), 1.41 (s, 9H), 1.25 – 1.18 (m, 3H), 1.08 (d, J = 7.4 Hz, 18H). 35 Step d: 4,4,4’,4’,5,5,5’,5’-octamethyl-2,2’-bi(1,3,2-dioxaborolane) (1.179 g) and 4,4’-di-tert-butyl- 2,2’-dipyridine (299.2 mg) were suspended in heptane (12 mL) and placed under nitrogen atmosphere. (1,5-cylooctadiene)(methoxy)iridium(I) dimer (370 mg) was added and the mixture was stirred at RT for 5 min., after which time a solution of methyl (S)-2-((tert- 40 butoxycarbonyl)amino)-3-(2-fluoro-5-((triisopropylsilyl)oxy)phenyl)propanoate (1.75 g) in heptane (12 mL) was added. The reaction mixture was stirred at 75 °C for 1 hr and then concentrated. The PAT059646-WO-PCT 5 resulting residue was purified by column chromatography (SiO2, 0 to 20% EtOAc in heptane) to afford methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(2-fluoro-3-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-5-((triisopropylsilyl)oxy)phenyl)propanoate (1.071 g) as a colorless syrup.1H NMR (400 MHz, CDCl3) 7.07 (t, J = 3.7 Hz, 1H), 6.73 (dd, J = 6.1, 3.2 Hz, 1H), 4.98 (d, J = 8.3 Hz, 1H), 4.52 (q, J = 6.9 Hz, 1H), 3.71 (s, 3H), 3.12 – 2.94 (m, 2H), 1.41 (s, 9H), 1.34 (s, 11H), 10 1.25 – 1.18 (m, 3H), 1.08 (s, 7H), 1.10 (s, 10H).19F NMR (376 MHz, CDCl3) -117.93. Intermediate 32 tert-butyl ((61R,65S,66S,4S)-11-ethyl-22-fluoro-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1- yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)- 15 indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)carbamate Procedure Step a: Intermediate 2 (552 mg), Intermediate 31 (450 mg), Pd(dtbpf)Cl2 (49 mg) and K2CO3 (209 mg) in 1,4-dioxane (4 mL) and water (800 L), was flushed with nitrogen and stirred at 70°C for 20 1 hr. The reaction was diluted with water and EtOAc. The layers were separated. The aq. layer was extracted with EtOAc (3x), and the combined organic extracts were washed with brine, dried over Na2SO4, filtered, and concentrated onto Celite® (15 g). The adsorbed material was purified by column chromatography (SiO2; 0 to 100 EtOAc/EtOH 3:1 in heptane) to afford benzyl 4-(5-(5- PAT059646-WO-PCT 5 (3-((S)-2-((tert-butoxycarbonyl)amino)-3-methoxy-3-oxopropyl)-2-fluoro-5- ((triisopropylsilyl)oxy)phenyl)-1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-1H-indol-2-yl)-6-((S)-1- methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (760 mg) as a colorless oil. LC/MS (FinalAnalysis-Acidic): M/Z = 1053.0 [M+H]+. 10 Step b: A solution of benzyl 4-(5-(5-(3-((S)-2-((tert-butoxycarbonyl)amino)-3-methoxy-3- oxopropyl)-2-fluoro-5-((triisopropylsilyl)oxy)phenyl)-1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-1H- indol-2-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (760 mg) in MeOH (10 mL) was purged with nitrogen, then Pd(OH)2 on carbon (532 mg, 20% wt.) was added. The mixture was purged again with nitrogen, then H2 via balloon. The reaction was stirred at RT for 2 15 hr under H2. After this time, the reaction was flushed with nitrogen prior to workup, then diluted with excess MeOH. The slurry was filtered under nitrogen protection and the filtrate concentrated to afford methyl (S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)-2-fluoro- 5-((triisopropylsilyl)oxy)phenyl)propanoate, which was carried forward to the next step without 20 further purification. LC/MS (RXMON-Acidic): M/Z = 919.0 [M+H]+. Step c: To a solution of methyl (2S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy- 2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)-2- fluoro-5-((triisopropylsilyl)oxy)phenyl)propanoate (80 mg) in MeOH (3 mL) was added aq. 25 formaldehyde (21 mg, 20 L, 37% wt.) and the solution was allowed to stir at RT for 30 min., after which time sodium triacetoxyhydroborate (37 mg) was added and the mixture stirred for 2 hr. The reaction was partially concentrated and purified by column chromatography (SiO2; 0 to 50% MeOH in DCM) to afford methyl (2S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy- 2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5- 30 yl)-2-fluoro-5-((triisopropylsilyl)oxy)phenyl)propanoate (70 mg) as a colorless oil. LC/MS (RXMON-Basic): M/Z = 933.2 [M+H]+. Step d: To a solution of methyl (2S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy- 2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5- 35 yl)-2-fluoro-5-((triisopropylsilyl)oxy)phenyl)propanoate (350 mg) in DCE (3 mL) was added trimethyltin hydroxide (339 mg) and the mixture was purged with nitrogen and stirred at 75°C for 4 hr. After this time, the reaction was cooled to RT and diluted with 10 volumetric equivalents of DCM. The mixture was quenched with aq.0.05N sodium bisulfite (37.54 mL) with vigorous stirring for 5 min. The layers were separated. The aq. layer was extracted with DCM (3x), and the 40 combined organic extracts were washed with brine, dried over Na2SO4, filtered, and concentrated to afford (2S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)-2-(2- PAT059646-WO-PCT 5 ((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)-2-fluoro-5- ((triisopropylsilyl)oxy)phenyl)propanoic acid (340 mg). LC/MS (Peptide method): M/Z = 919.0 [M+H]+. Step e, part 1: To Intermediate 1 (220 mg) in DCM (2.5 mL) was added TFA (2.4 mL) and the 10 solution was stirred at RT for 1 hr, after which time, the reaction mixture was concentrated. Additional DCM was added and concentrated and this process was repeated 3x to afford methyl (1S,2S,6R)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylate, trifluoroacetate salt (157 mg) which was carried forward without further purification. 15 Step e, part 2: To methyl (1S,2S,6R)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylate (157 mg) at RT, a solution of (2S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)- 2-fluoro-5-((triisopropylsilyl)oxy)phenyl)propanoic acid (340 mg), DIPEA (574 mg, 774 L), and HATU (282 mg) in DMF (3 mL) was added. The reaction was stirred at RT for 3 hr. Purification 20 by column chromatography (C18; 10 to 100% acetonitrile in water (+0.1% NH4OH)) afforded methyl (1S,2S,6R)-4-((2S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)- 2-fluoro-5-((triisopropylsilyl)oxy)phenyl)propanoyl)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylate (260 mg) as a white solid after lyophilization. LC/MS (Peptide method): M/Z = 1057.0 [M+H]+. 25 Step f: To a solution of methyl (1S,2S,6R)-4-((2S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3- (3-hydroxy-2,2-dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)- 1H-indol-5-yl)-2-fluoro-5-((triisopropylsilyl)oxy)phenyl)propanoyl)-3,4- diazabicyclo[4.1.0]heptane-2-carboxylate (260 mg) in DCE (2 mL) was added trimethyltin 30 hydroxide (223 mg) and the mixture was purged with nitrogen and stirred at 75°C for 4 hr. The reaction was cooled to ambient temperature and diluted with 10 volumetric equivalents of DCM. The mixture was quenched with aq.0.05N sodium bisulfite (25 mL) with vigorous stirring for 5 min. The layers were separated. The aq. layer was extracted with DCM (3x), and the combined organic extracts were washed with brine, dried over Na2SO4, filtered, and concentrated to afford35 (1S,2S,6R)-4-((S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy-2,2-dimethylpropyl)- 2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)-2-fluoro-5- ((triisopropylsilyl)oxy)phenyl)propanoyl)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylic acid, which was forwarded to the next step. LC/MS (Peptide method): M/Z = 1042.0 [M+H]+. 40 Step g: To (1S,2S,6R)-4-((2S)-2-((tert-butoxycarbonyl)amino)-3-(3-(1-ethyl-3-(3-hydroxy-2,2- dimethylpropyl)-2-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indol-5-yl)- PAT059646-WO-PCT 5 2-fluoro-5-((triisopropylsilyl)oxy)phenyl)propanoyl)-3,4-diazabicyclo[4.1.0]heptane-2-carboxylic acid (260 mg) was charged DCM (25 mL) and HOAt (170 mg) sequentially and the mixture was purged with nitrogen (3x). A solution of EDC (239 mg) in DCM (2.5 mL) was added to the mixture and the reaction was stirred at RT for 72 hr, after which time it was concentrated and purified by column chromatography (C18; 0 to 100% acetonitrile in water (+0.1% TFA)) to afford tert-butyl10 ((61R,65S,66S,4S)-11-ethyl-22-fluoro-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin- 3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola- 6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)carbamate, trifluoroacetate salt (145 mg) as a yellow powder after lyophilization. LC/MS (Peptide method): M/Z = 1025.0 [M+H]+. 15 Intermediate 33 tert-butyl ((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)carbamate 20 Procedure Step a: Intermediate 26 (250 mg) and Pd(OH)2 on carbon (22 mg, 10% wt.) were suspended in MeOH (2.2 mL). The flask was purged with nitrogen (2x) and then with hydrogen (2x). The 25 reaction was stirred at RT under an atmosphere of hydrogen for 2 hr. The solution was filtered and concentrated to afford tert-butyl ((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5- (piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64- diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)carbamate (212 mg) as a grey solid. LC/MS (Peptide Method) M/Z = 994.5900 [M+H]+. 30 Intermediate 34 4-(5-((61R,65S,66S,4S)-11-ethyl-10,10-dimethyl-4-((2S,3R)-3-methyl-2-(methylamino)pent-4- enamido)-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- PAT059646-WO-PCT 5 bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3- yl)piperazine-1-carboxylate Procedure Step a: To a solution of Intermediate 33 (213 mg) and NEt3 (67.5 mg, 93 L) in DCM (2.2 mL) 10 was added methyl carbonochloridate (41.5 mg, 34 L). The reaction was stirred for 1 hr and additional methyl carbonochloridate (11.0 mg, 9 L) and NEt3 (67.5 mg, 93 L) were added. The reaction stirred at RT for a total of 2 hr. The reaction mixture was diluted with DCM and washed with brine. The aq. layer was extracted with DCM (3x), and the combined organic extracts were passed through a phase separator and concentrated. The residue was purified by column15 chromatography (SiO2; 0 to 10% MeOH (containing 0.2% NH4OH) in DCM) to afford methyl 4-(5- ((61R,65S,66S,4S)-4-((tert-butoxycarbonyl)amino)-11-ethyl-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (190 mg) as a white solid. LC/MS (Peptide Method) M/Z = 1052.6100 [M+H]+.1H NMR (400 MHz, 20 DMSO) 8.47 (d, J = 2.9 Hz, 1H), 7.99 – 7.93 (m, 1H), 7.61 (d, J = 8.6 Hz, 1H), 7.52 (dd, J = 8.7, 1.7 Hz, 1H), 7.30 (s, 1H), 7.27 (d, J = 2.8 Hz, 1H), 7.10 (d, J = 8.7 Hz, 1H), 7.04 (t, J = 1.9 Hz, 1H), 6.79 (s, 1H), 4.95 (t, J = 9.2 Hz, 1H), 4.89 (d, J = 11.9 Hz, 1H), 4.29 – 4.19 (m, 2H), 4.19 – 4.07 (m, 2H), 3.76 (d, J = 10.8 Hz, 1H), 3.64 – 3.60 (m, 3H), 3.56 – 3.47 (m, 4H), 3.30 – 3.22 (m, 5H), 3.15 (s, 3H), 2.92 – 2.82 (m, 1H), 2.77 – 2.70 (m, 1H), 2.69 (s, 3H), 2.67 – 2.57 (m, 2H), 1.36 25 (d, J = 6.2 Hz, 3H), 1.33 (s, 9H), 1.28 (d, J = 7.4 Hz, 2H), 1.23 (q, J = 7.3 Hz, 4H), 1.12 – 1.08 (m, 18H), 0.97 – 0.91 (m, 3H), 0.85 (s, 3H), 0.44 (s, 3H), 0.36 – 0.30 (m, 1H). PAT059646-WO-PCT 5 Step b: To a solution of methyl 4-(5-((61R,65S,66S,4S)-4-((tert-butoxycarbonyl)amino)-11-ethyl- 10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3- yl)piperazine-1-carboxylate (190 mg) in DCM (1.5 mL) was charged TFA (0.37 mL). The reaction was stirred at RT for 45 min. and then concentrated. The remaining residue was dissolved in DCM 10 and concentrated again; this process was repeated (3x). The resulting solid was dissolved in DCM and neutralized with sat. aq. NaHCO3. The aq. layer was extracted with DCM (3x). The combined organic extracts were passed through a phase separator and concentrated to afford methyl 4-(5- ((61R,65S,66S,4S)-4-amino-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8- oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12- 15 yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (171 mg) as a pale yellow solid. LC/MS (Peptide Method) M/Z = 951.6500 [M+H]+. Step c: To a solution of methyl 4-(5-((61R,65S,66S,4S)-4-amino-11-ethyl-10,10-dimethyl-5,7-dioxo- 25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-20 2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1- carboxylate (86 mg), Intermediate 23 (29 mg), and DIPEA (46.7 mg, 63 L) in NMP (900 L) at 0 °C was added COMU (50 mg). The reaction was allowed to warm to RT and stirred at RT for 40 min. The reaction mixture was diluted with EtOAc and washed with sat. aq. NaHCO3 and brine. The aq. layer was extracted with EtOAc (3x). The combined organic extracts were washed with25 water (2x), brine (2x), passed through a phase separator, and concentrated to afford methyl 4-(5- ((61R,65S,66S,4S)-4-((2S,3R)-2-((tert-butoxycarbonyl)(methyl)amino)-3-methylpent-4-enamido)- 11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola- 6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1- methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (96 mg) as an orange solid. LC/MS (Peptide 30 Method) M/Z = 1177.6801 [M+H]+.1H NMR (400 MHz, DMSO) 8.48 (d, J = 2.9 Hz, 1H), 7.96 – 7.87 (m, 1H), 7.65 – 7.59 (m, 1H), 7.48 – 7.41 (m, 1H), 7.34 – 7.24 (m, 2H), 6.98 – 6.94 (m, 1H), 6.67 – 6.57 (m, 1H), 5.88 – 5.73 (m, 1H), 5.44 – 5.31 (m, 1H), 5.04 – 4.90 (m, 2H), 4.32 – 3.99 (m, 5H), 3.95 (d, J = 11.7 Hz, 1H), 3.71 (d, J = 10.8 Hz, 1H), 3.68 – 3.60 (m, 4H), 3.56 – 3.49 (m, 4H), 3.30 – 3.22 (m, 5H), 3.13 – 3.05 (m, 3H), 2.78 – 2.72 (m, 3H), 2.72 – 2.64 (m, 5H), 1.46 – 35 1.40 (m, 9H), 1.37 (d, J = 6.2 Hz, 3H), 1.31 – 1.22 (m, 5H), 1.19 – 1.13 (m, 2H), 1.12 – 1.06 (m, 18H), 1.04 – 0.95 (m, 3H), 0.86 (s, 4H), 0.80 (s, 3H), 0.49 (s, 3H), 0.15 (s, 1H). Step d: A solution of 4-(5-((61R,65S,66S,4S)-4-((2S,3R)-2-((tert-butoxycarbonyl)(methyl)amino)-3- methylpent-4-enamido)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-40 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)- 6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (95 mg) in DCM (650 L) was PAT059646-WO-PCT 5 charged TFA (160 L). The reaction was stirred at RT for 45 min. and then concentrated. The remaining residue was dissolved in DCM and concentrated again; this process was repeated (3x). The resulting solid was dissolved in DCM and neutralized with sat. aq. NaHCO3. The aq. layer was extracted with DCM (3x). The combined organic extracts were passed through a phase separator and concentrated to afford methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-10,10-dimethyl-4-10 ((2S,3R)-3-methyl-2-(methylamino)pent-4-enamido)-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8- oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12- yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (93 mg) as a yellow solid. LC/MS (Peptide Method) M/Z = 1076.6200 [M+H]+. 15 Intermediate 35 methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-10,10-dimethyl-4-((2S,3R)-3-methyl-2-((3S,4S)-N-methyl- 4-phenylpyrrolidine-3-carboxamido)pent-4-enamido)-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8- oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12- yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate 20 Procedure Step a: To a solution of Intermediate 34 (199 mg), Intermediate 15 (59 mg), and DIPEA (60 mg, 81 L) in 1,4-dioxane (740 L) was added PyBroP (95 mg). The reaction stirred for 3 hr and additional Intermediate 15 (27 mg), DIPEA (24 mg, 32 L) and PyBroP (43 mg) were added. The 25 reaction stirred for 21 hr and additional PyBroP (43 mg) and DIPEA (24 mg, 32 L) were added. 166 PAT059646-WO-PCT 5 The reaction stirred at 40 °C for an additional 24 hr. The reaction stirred for a total of 48 h. The reaction mixture was concentrated, then was diluted in EtOAc and washed with sat. aq. NaHCO3 and Rochelle's salt. The aq. layer was extracted with EtOAc (3x), and the combined organic extracts were passed through a phase separator and concentrated. The residue was purified by column chromatography (SiO2; 0 to 10% MeOH (+0.2% NH4OH) in DCM) to afford methyl 4-(5-10 ((61R,65S,66S,4S)-4-((2S,3R)-2-((3S,4S)-1-(tert-butoxycarbonyl)-N-methyl-4-phenylpyrrolidine-3- carboxamido)-3-methylpent-4-enamido)-11-ethyl-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (218 mg) as an off-white solid.1H NMR (400 MHz, DMSO) 8.48 (m, 1H), 7.92 – 7.82 (m, 1H), 15 7.74 – 7.41 (m, 2H), 7.35 – 7.13 (m, 8H), 7.00 – 6.84 (m, 2H), 6.70 – 6.59 (m, 1H), 6.59 – 6.52 (m, 1H), 5.64 – 5.48 (m, 1H), 5.47 – 5.36 (m, 1H), 5.17 – 4.89 (m, 1H), 4.86 – 4.75 (m, 1H), 4.52 (d, J = 9.3 Hz, 1H), 4.28 – 4.01 (m, 4H), 3.90 – 3.82 (m, 1H), 3.70 (s, 4H), 3.66 – 3.60 (m, 5H), 3.56 – 3.47 (m, 5H), 3.31 – 3.20 (m, 5H), 3.10 – 3.06 (m, 2H), 2.75 – 2.61 (m, 6H), 2.38 (s, 1H), 1.66 – 1.53 (m, 1H), 1.49 – 1.35 (m, 17H), 1.34 – 1.22 (m, 4H), 1.20 – 1.15 (m, 2H), 1.14 – 1.08 20 (m, 13H), 1.05 – 0.98 (m, 6H), 0.86 (d, J = 6.7 Hz, 1H), 0.77 (s, 3H), 0.51 (s, 2H), 0.41 – 0.28 (m, 2H). LC/MS (Peptide method) M/Z = 1349.8000 [M+H]+. Step b: To a solution of methyl 4-(5-((61R,65S,66S,4S)-4-((2S,3R)-2-((3S,4S)-1-(tert- butoxycarbonyl)-N-methyl-4-phenylpyrrolidine-3-carboxamido)-3-methylpent-4-enamido)-11-25 ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola- 6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1- methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (218 mg) in DCM (1.8 mL) was added TFA (4 μL). The reaction stirred at RT for 45 min. The reaction mixture was concentrated, and the remaining residue was dissolved in DCM and concentrated again; this process was repeated (3x). 30 The resulting yellow solid was dissolved in DCM and washed with sat. aq. NaHCO3. The aq. layer of extracted with DCM (3x), and the organic extracts were passed through a phase separator and concentrated to afford methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-10,10-dimethyl-4-((2S,3R)-3- methyl-2-((3S,4S)-N-methyl-4-phenylpyrrolidine-3-carboxamido)pent-4-enamido)-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- 35 benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (206 mg) as an off-white solid. LC/MS (Peptide method) M/Z = 1249.7400 [M+H]+. Intermediate 36 (2R,3S)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-40 1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza- PAT059646-WO-PCT 5 1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3- methyl-1-oxobutan-2-yl)-N-methyl-3-phenylazetidine-2-carboxamide Procedure 10 Step a: A solution of Intermediate 5 (890 mg) in DCM (7 mL) and TFA (1.8 mL) was stirred at RT for 50 min. The solution was concentrated, and the resulting yellow foam was dissolved in DCM and concentrated again. This process was repeated (3x). Et2O was added, the mixture sonicated, and the liquid was decanted away (2x). The remaining solids were dried to afford 61R,65S,66S,4S)- 4-amino-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-15 dimethyl-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-5,7-dione, trifluoroacetate (1.15 g) as a yellow foam. LC/MS (FinalAnalysis-Basic): M/Z = 907.2 [M+H]+. Step b: To a solution of (61R,65S,66S,4S)-4-amino-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-20 methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64- diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-5,7-dione trifluoroacetate (1.45 g), N-(tert-butoxycarbonyl)-N-methyl-L-valine (515 mg), and DIPEA (1.00 mg, 1.16 mL) in NMP (10 mL) was added COMU (953 mg) at 0 °C under a nitrogen atmosphere. The reaction warmed to RT and stir for 45 min. Water was added, the mixture was filtered, and 25 retained solids were rinsed with water. The retained material was purified by column chromatography (SiO2; 0 to 10% MeOH (containing 1% NH4OH) in DCM) to afford tert-butyl ((2S)- 1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2- 168 PAT059646-WO-PCT 5 yl)(methyl)carbamate (1.16 g) as a pale orange solid. LC/MS (FinalAnalysis-Acidic) M/Z = 1120.0 [M+H]+.1H NMR (400 MHz, DMSO) 8.46 (d, J = 2.9 Hz, 1H), 8.04 (s, 1H), 7.93 – 7.72 (m, 1H), 7.61 (d, J = 8.6 Hz, 1H), 7.44 (dd, J = 8.8, 1.7 Hz, 1H), 7.31 (s, 1H), 7.23 (d, J = 2.9 Hz, 1H), 7.00 – 6.91 (m, 1H), 6.73 – 6.61 (m, 1H), 5.34 (s, 1H), 4.93 (s, 1H), 4.29 – 4.07 (m, 4H), 4.06 – 3.95 (m, 2H), 3.75 – 3.68 (m, 1H), 3.65 (s, 1H), 3.34 – 3.20 (m, 6H), 3.10 (s, 2H), 2.90 – 2.72 (m, 4H), 10 2.72 – 2.65 (m, 4H), 2.47 (s, 1H), 2.26 – 2.21 (m, 3H), 2.07 – 2.00 (m, 1H), 1.99 (s, 1H), 1.40 (s, 9H), 1.38 – 1.34 (m, 3H), 1.32 – 1.22 (m, 4H), 1.21 – 1.05 (m, 20H), 1.02 – 0.95 (m, 3H), 0.92 – 0.87 (m, 3H), 0.87 – 0.83 (m, 1H), 0.83 – 0.72 (m, 6H), 0.49 (s, 3H), 0.22 – 0.10 (m, 1H). Step c: A solution of tert-butyl ((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-15 (4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8- oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)amino)-3-methyl-1-oxobutan-2-yl)(methyl)carbamate (657 mg) in DCM (4.8 mL) and TFA (1.2 mL) was stirred at RT for 1 hr. The solution was concentrated, and the residue was dissolved in DCM and concentrated again. This process was repeated (3x). The remaining residue was 20 dissolved in EtOAc and neutralized with sat. aq. NaHCO3. The aq. layer was extracted with EtOAc (3x). The combined organic extracts were washed with sat. aq. NaHCO3, brine, dried over Na2SO4, and passed through a phase separator. The filtrate was concentrated to afford (2S)-N- ((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-25 bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)-3-methyl-2- (methylamino)butanamide (580 mg) as an orange solid. LC/MS (Peptide Method) M/Z = 1019.6800 [M+H]+. Step d: To a solution of (2S)-N-((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-30 methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa- 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)-3- methyl-2-(methylamino)butanamide (128 mg), Intermediate 20 (45 mg), and PyBroP (76 mg) in 1,4-dioxane (1.2 mL) was added DIPEA (59 mg, 79 L). The reaction stirred at RT for 20 hr and additional PyBroP (29 mg) and DIPEA (16 mg, 22 L) were added. The reaction was stirred for 35 an additional 18 hr after which time additional Intermediate 20 (8.7 mg), PyBroP (15 mg), and DIPEA (8.2 mg, 11 L) were added. The reaction stirred for an additional 6 hr, representing a total of 48 hr. The mixture was concentrated, diluted with EtOAc, and washed with sat. aq. NaHCO3 and sat. Rochelle's salt solution. The aq. layer was extracted with EtOAc (3x), and the combined organic extracts were passed through a phase separator and concentrated to afford tert-butyl40 (2R,3S)-2-(((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin- 1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza- PAT059646-WO-PCT 5 1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3- methyl-1-oxobutan-2-yl)(methyl)carbamoyl)-3-phenylazetidine-1-carboxylate (270 mg). LC/MS (Peptide Method) M/Z = 1279.2900 [M+H]+. Step e: To a solution of crude tert-butyl (2R,3S)-2-(((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-10 1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)(methyl)carbamoyl)-3- phenylazetidine-1-carboxylate (270 mg) in DCM (1.1 mL) was added TFA (0.3 mL). The reaction was stirred at RT for 2 hr and concentrated. The remaining residue was dissolved in DCM and 15 concentrated again; this process was repeated (3x). The resulting solid was dissolved in DCM and neutralized with sat. aq. NaHCO3. The aq. layer was extracted with DCM (3x). The combined organic extracts were passed through a phase separator and concentrated to afford (2R,3S)-N- ((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin- 3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-20 6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxobutan-2-yl)-N-methyl-3-phenylazetidine-2-carboxamide (133 mg) as a yellow solid. LC/MS (FinalAnalysis-Basic) M/Z = 1179.7 [M+H]+. Intermediate 37 25 methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-4-((2S,3R)-2-((3S,4S)-1-(methoxycarbonyl)-N-methyl-4- (4-(piperazin-1-yl)phenyl)pyrrolidine-3-carboxamido)-3-methylpent-4-enamido)-10,10-dimethyl- 5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3- yl)piperazine-1-carboxylate 30
PAT059646-WO-PCT 5 Procedure Step a: To a solution of Intermediate 34 (49.4 mg), Intermediate 17 (25.9 mg), and DIPEA (18 mg, 24 L) in 1,4-dioxane (200 μL) was added PyBroP (32.1 mg) and the reaction was heated to 40°C. After stirring for 5 hr additional PyBroP (10.7 mg) and DIPEA (12 mg, 16 L) were added. 10 The reaction was stirred for 6 hr and additional Intermediate 17 (10 mg), PyBroP (21.4 mg), and DIPEA (11.9 mg, 16.0 L) were added, and the reaction was stirred at 40 °C for 16 hr. After this time, additional PyBroP (10.7 mg) was added, and the reaction mixture stirred for 5 additional hr. The reaction mixture stirred for a total of 30 hr. The reaction mixture was concentrated and purified by column chromatography (SiO2; 0 to 10% MeOH (+0.2% NH4OH) in DCM) to afford tert-butyl15 4-(4-((3S,4S)-4-(((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-12-(5-(4-(methoxycarbonyl)piperazin-1- yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8- oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)amino)-3-methyl-1-oxopent-4-en-2-yl)(methyl)carbamoyl)-1-(methoxycarbonyl)pyrrolidin-3- yl)phenyl)piperazine-1-carboxylate (89 mg) as an off white-solid. LC/MS (Peptide method) M/Z = 20 1491.8500 [M+H]+. Step b: To a solution of tert-butyl 4-(4-((3S,4S)-4-(((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-12-(5- (4-(methoxycarbonyl)piperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-25 2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopent-4-en-2- yl)(methyl)carbamoyl)-1-(methoxycarbonyl)pyrrolidin-3-yl)phenyl)piperazine-1-carboxylate (89 mg) in DCM (200 μL) was added TFA (100 μL) and the mixture was stirred at RT for 2 hr. The PAT059646-WO-PCT 5 reaction mixture was concentrated and the resulting residue was dissolved in DCM and concentrated; this process was repeated (3x). The resulting yellow solid was dissolved in DCM and washed with sat. aq. NaHCO3. The aq. layer of extracted with DCM (3x), and the organic layers were passed through a phase separator and concentrated to afford methyl 4-(5- ((61R,65S,66S,4S)-11-ethyl-4-((2S,3R)-2-((3S,4S)-1-(methoxycarbonyl)-N-methyl-4-(4-(piperazin-10 1-yl)phenyl)pyrrolidine-3-carboxamido)-3-methylpent-4-enamido)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (61 mg) as a pale yellow solid. LC/MS (Peptide method) M/Z 1390.8101 [M+H]+. 15 Intermediate 38 (2R,3S)-N-((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(piperazin-1- yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)- indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxopent-4-en-2-yl)-N-methyl-3-phenyloxetane-2-carboxamide 20 Procedure Step a: To a solution of Intermediate 26 (867 mg) in MeOH (15 mL) under nitrogen atmosphere was charged Pd(OH)2 on carbon (540 mg, 10% wt.). The reaction apparatus was evacuated and backfilled with hydrogen (3x) and the suspension was left stirring at RT under hydrogen 25 atmosphere for 2 hr. After this time, the apparatus was purged with nitrogen and the reaction mixture was filtered through a pad of Celite® topped with Na2SO4. The filter cake was thoroughly conditioned with MeOH. The filtrate was concentrated to afford tert-butyl ((61R,65S,66S,4S)-11- ethyl-12-(2-((S)-1-methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- PAT059646-WO-PCT 5 ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)carbamate (726 mg) as an off-white solid. LC/MS (Peptide method): M/Z = 1014.5900 [M+H]+. Step b: To a solution of tert-butyl ((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-10 (piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64- diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)carbamate (706 mg) in 2-MeTHF (10 mL) under nitrogen atmosphere was charged Et3N (180 mg, 248 μL). After stirring for about 5 min., Fmoc-Oxyma (453.6 mg) was added in one portion. The reaction mixture was stirred at RT for 1 hr, upon which time the reaction was quenched with 15 water (5 mL) with vigorous stirring. The aq. phase was back-extracted with EtOAc (10 mL, 3x) and combined organic extracts were passed through a phase separator. Crude filtrate was concentrated directly onto Celite® and purified by column chromatography (SiO2; 0 to 100% EtOAc in heptane) to afford (9H-fluoren-9-yl)methyl 4-(5-((61R,65S,66S,4S)-4-((tert- butoxycarbonyl)amino)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-20 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)- 6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (790 mg) as a white solid. LC/MS (Peptide method): M/Z = 1214.6899 [M+H]+. Step c: To a solution of (9H-fluoren-9-yl)methyl 4-(5-((61R,65S,66S,4S)-4-((tert-25 butoxycarbonyl)amino)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa- 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)- 6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (790 mg) in DCM (10 mL) was charged TFA (3.71 g, 2.51 mL). The resulting solution was stirred at RT for 2 hr, after which time the reaction was concentrated. The remaining residue was reconstituted in DCM and re- 30 concentrated to drive off TFA, this process was repeated a total of 3 times. The remaining residue was again taken up in DCM and the organic phase was neutralized with sat. aq. Na2CO3 solution. The aq. phase was back-extracted with DCM (10 mL, 3x) and combined organic extracts were passed through a phase separator. The filtrate was concentrated to afford (9H-fluoren-9-yl)methyl 4-(5-((61R,65S,66S,4S)-4-amino-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-35 8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane- 12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (725 mg) as a cream-colored solid. LC/MS (Peptide method): M/Z = 1114.6200 [M+H]+. Step d: To a solution of (9H-fluoren-9-yl)methyl 4-(5-((61R,65S,66S,4S)-4-amino-11-ethyl-10,10-40 dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3- PAT059646-WO-PCT 5 yl)piperazine-1-carboxylate (725 mg) in acetonitrile (10.4 mL) was charged a solution of Intermediate 23 (206 mg) in 2-MeTHF (2.6 mL). The resulting solution was cooled to 0 °C and purged with nitrogen. DIPEA (420 mg, 566 μL) and COMU (362 mg) were sequentially added, and reaction was left to stir at 0 °C for 1 hr. After this time, the reaction was concentrated, and the remaining residue was partitioned between EtOAc (50.0 mL) and water (10 mL). The organic 10 phase was taken and concentrated directly onto Celite® and purified by column chromatography (SiO2; 0 to 100% EtOAc in heptane) to afford (9H-fluoren-9-yl)methyl 4-(5-((61R,65S,66S,4S)-4- ((2S,3R)-2-((tert-butoxycarbonyl)(methyl)amino)-3-methylpent-4-enamido)-11-ethyl-10,10- dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3- 15 yl)piperazine-1-carboxylate (870 mg) as a pale pink solid. LC/MS (Peptide method): M/Z = 1340.1700 [M+H]+. Step e: To a solution of (9H-fluoren-9-yl)methyl 4-(5-((61R,65S,66S,4S)-4-((2S,3R)-2-((tert- butoxycarbonyl)(methyl)amino)-3-methylpent-4-enamido)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-20 ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (870 mg) in DCM (10 mL) was charged TFA (3.70 g, 2.50 mL) ). The resulting solution was stirred at RT for 1.5 hr, after which time the reaction was concentrated. The remaining residue was reconstituted in DCM and re-concentrated to drive off TFA, this process was repeated a total of 3 25 times. The remaining residue was again taken up in DCM and the organic phase was neutralized with sat. aq. Na2CO3 solution. The aq. phase was back-extracted with DCM (10 mL, 3x) and combined organic extracts were passed through a phase separator. The filtrate was concentrated to afford (9H-fluoren-9-yl)methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-10,10-dimethyl-4-((2S,3R)-3- methyl-2-(methylamino)pent-4-enamido)-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-30 diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)- 1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (805 mg) as a cream-colored solid. LC/MS (Peptide method): M/Z = 1239.7100 [M+H]+. 35 Step f: To a solution of (9H-fluoren-9-yl)methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-10,10-dimethyl-4- ((2S,3R)-3-methyl-2-(methylamino)pent-4-enamido)-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8- oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12- yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (805 mg) and Intermediate 10 in acetonitrile (5.2 mL) was added 2-MeTHF (1.3 mL). The solution was purged with nitrogen and 40 cooled to 0 °C. DIPEA (420 mg, 566 μL) and HATU (296 mg) were sequentially added, and the reaction solution was stirred at 0 °C for 4.5 hr. After this time, the reaction mixture was PAT059646-WO-PCT 5 concentrated directly onto silica and purified by column chromatography (SiO2; 0 to 100 EtOAc/EtOH 3:1 in heptane) to afford (9H-fluoren-9-yl)methyl 4-(5-((61R,65S,66S,4S)-11-ethyl- 10,10-dimethyl-4-((2S,3R)-3-methyl-2-((2R,3S)-N-methyl-3-phenyloxetane-2-carboxamido)pent- 4-enamido)-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3- 10 yl)piperazine-1-carboxylate (830 mg) as a cream-colored solid. LC/MS (Peptide method): M/Z = 1399.7700 [M+H]+. Step g: To a solution of (9H-fluoren-9-yl)methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-10,10-dimethyl-4- ((2S,3R)-3-methyl-2-((2R,3S)-N-methyl-3-phenyloxetane-2-carboxamido)pent-4-enamido)-5,7-15 dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana- 2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1- carboxylate (830 mg) in acetonitrile (11.9 mL) under nitrogen atmosphere was charged 4- methylpiperidine (1.18 g, 1.4 mL). The reaction solution was stirred at RT for 30 min. After this time, the solution was concentrated directly onto Celite® and purified by column chromatography 20 (SiO2; 0 to 100% EtOAc in heptane and then solvent swapping to 0 to 50% MeOH in DCM) to afford (2R,3S)-N-((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(piperazin- 1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza- 1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3- methyl-1-oxopent-4-en-2-yl)-N-methyl-3-phenyloxetane-2-carboxamide (601 mg) as a white 25 powder. LC/MS (Peptide method): M/Z = 1178.7000 [M+H]+. Compound 101 Note: axial chirality of compounds is as shown in below scheme. (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(2-((S)-1-methoxyethyl)-5-(4-30 methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola- 6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxobutan-2-yl)-N-methyl-3-phenyltetrahydrofuran-2-carboxamide
PAT059646-WO-PCT 5 Procedure Step a: To a stirred solution of Intermediate 5 (30.0 mg) in DCM (1.0 mL) was added TFA (195 μL). The reaction solution was stirred at RT for 1 hr, then concentrated under reduced pressure. 10 The resulting residue was taken up in DCM (1 mL) and concentrated under reduced pressure (3x). The residue obtained was dissolved in DMF (0.50 mL), and N-(tert-butoxycarbonyl)-N- methyl-L-valine (13.6 mg), HATU (22.3 mg) and DIPEA (50.7 μL) were added. The resulting mixture was stirred at RT for 30 min, then the reaction mixture was purified by column chromatography (C18, 0 to 100% acetonitrile in water + 0.1% TFA) followed by lyophilization15 afforded tert-butyl ((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa- 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)amino)-3-methyl-1-oxobutan-2-yl)(methyl)carbamate (23.0 mg) as a yellow solid. LC/MS (Peptide method): M/Z = 1120 [M+H]+. 20 Step b: To a stirred solution of tert-butyl ((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)(methyl)carbamate (23.0 mg) 25 in DCM (0.50 mL) was added TFA (158 μL) and the resulting solution was stirred at RT for 1 hr, then concentrated under reduced pressure. The resulting residue was taken up in DCM (1 mL) PAT059646-WO-PCT 5 and concentrated under reduced pressure (3x). The residue obtained was dissolved in DMF (0.30 mL), and (2R,3R)-3-phenyltetrahydrofuran-2-carboxylic acid (5.9 mg), HATU (15.5 mg) and N,N- diisopropylethylamine (35.1 μL) were added. The resulting mixture was stirred at RT for 3 hr then purified by column chromatography (C18, 0 to 100% acetonitrile in water + 0.1% TFA) followed by lyophilization afforded (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-10 methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3- phenyltetrahydrofuran-2-carboxamide trifluoroacetic acid salt (22.0 mg) as a yellow solid. LC/MS (Peptide method): M/Z = 1194 [M+H]+. 15 Step c: To a stirred solution of (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3- 20 phenyltetrahydrofuran-2-carboxamide trifluoroacetic acid salt (22.0 mg) in tetrahydrofuran (0.25 mL) under a nitrogen atmosphere was added TBAF (1 M solution in THF, 66.7 μL). The resulting solution was stirred at RT for 1 hr. The reaction mixture was diluted with EtOAc (5 mL) and water (5 mL) and stirred vigorously. The layers were separated, and the aq. layer was extracted with EtOAc (3 x 5 mL). The combined organic layers were washed with brine (5 mL), dried over 25 Na2SO4, and concentrated. The crude material was purified by column chromatography (C18, 0 to 100% acetonitrile in water + 0.1% NH4OH) followed by lyophilization to afford (2R,3R)-N-((2S)- 1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1- yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)- 30 N-methyl-3-phenyltetrahydrofuran-2-carboxamide (axial chirality as shown in structure)(9.5 mg) as a white solid. LC/MS (Peptide method): M/Z = 1038 [M+H]+.1H NMR (600 MHz, DMSO-d6) 9.15 (s, 1H), 8.45 (d, J = 3.0 Hz, 1H), 7.86 (s, 1H), 7.73 (d, J = 8.0 Hz, 1H), 7.57 (d, J = 8.6 Hz, 1H), 7.46 (d, J = 1.8 Hz, 1H), 7.28 – 7.27 (m, 2H), 7.25 – 7.23 (m, 1H), 7.22 (s, 2H), 7.14 (t, J = 7.3 Hz, 1H), 7.08 (s, 1H), 6.94 (s, 1H), 6.53 (s, 1H), 5.33 – 5.28 (m, 1H), 5.08 (d, J = 7.7 Hz, 1H), 35 4.79 (d, J = 11.7 Hz, 1H), 4.26 (d, J = 4.7 Hz, 1H), 4.26 – 4.25 (m, 1H), 4.24 – 4.20 (m, 1H), 4.16 – 4.14 (m, 1H), 4.13 – 4.12 (m, 1H), 4.10 – 4.06 (m, 1H), 3.88 – 3.85 (m, 1H), 3.85 – 3.82 (m, 1H), 3.79 – 3.74 (m, 1H), 3.73 – 3.70 (m, 1H), 3.59 – 3.56 (m, 1H), 3.27 – 3.26 (m, 4H), 3.18 – 3.15 (m, 1H), 3.11 (s, 3H), 2.80 – 2.76 (m, 1H), 2.70 – 2.67 (m, 1H), 2.66 (s, 3H), 2.66 – 2.62 (m, 2H), 2.46 – 2.45 (m, 4H), 2.37 – 2.34 (m, 1H), 2.29 – 2.24 (m, 1H), 2.21 (s, 3H), 1.73 – 1.66 (m, 40 1H), 1.35 (d, J = 6.1 Hz, 3H), 1.18 – 1.15 (m, 2H), 0.97 (t, J = 7.2 Hz, 3H), 0.81 (s, 3H), 0.78 (s, PAT059646-WO-PCT 5 1H), 0.66 (d, J = 6.5 Hz, 3H), 0.46 (s, 3H), 0.10 (s, 1H), -0.07 (d, J = 6.7 Hz, 3H). HRMS m/z [M+H]+ = 1037.5881. The following compounds of Table 6 were synthesized using the above procedure or modifications to the above procedure using the corresponding macrocycle, aminoacid, 10 and acid intermediates. Note: axial chirality of compounds is as shown in structures in Table 6. Table 6: PanRAS inhibitor compounds 178 PAT059646-WO-PCT 179 PAT059646-WO-PCT PAT059646-WO-PCT 181 PAT059646-WO-PCT 5 Compound 108 Note: axial chirality of compounds is as shown in below scheme. (2R,3R)-N-((1S)-1-cyclopentyl-2-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-10 diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)- 2-oxoethyl)-N-methyl-3-phenyltetrahydrofuran-2-carboxamide Procedure Step a: To a solution of Intermediate 5 (15.0 mg) in DCM (0.5 mL) at RT was added TFA (100 15 L). The resulting solution was stirred at RT for 30 min., and then concentrated under reduced pressure. The resulting residue was azeotroped three time with DCM under reduced pressure. To the residue, a solution containing DMF (0.5 mL), (S)-2-(((benzyloxy)carbonyl)(methyl)amino)- PAT059646-WO-PCT 5 2-cyclopentylacetic acid (5.1 mg), HATU (8.4 mg) and DIPEA (25 L) was added. The reaction mixture was stirred at RT for 1 hr, and then directly purified by column chromatography (C18, 0 to 100% acetonitrile in water + 0.1% TFA) to give benzyl ((1S)-1-cyclopentyl-2-(((61R,65S,66S,4S)- 11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana- 10 2(1,3)-benzenacycloundecaphane-4-yl)amino)-2-oxoethyl)(methyl)carbamate (16 mg) as a yellow solid after lyophilization. LC/MS (Peptide method): M/Z = 1180 [M+H]+. Step b: A mixture of benzyl ((1S)-1-cyclopentyl-2-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-15 ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-2-oxoethyl)(methyl)carbamate (16 mg) and Pd on carbon (7.2 mg, 10%) in MeOH (1.0 mL) was stirred at RT for 16 hr under hydrogen. The slurry was diluted with methanol and filtered. The filtrate was concentrated and then dissolved in DMF (0.5 mL), and (2R,3R)-3-phenyltetrahydrofuran-2-carboxylic acid (6.6 mg), HATU (17.5 mg), and 20 DIPEA (40 L) were added. The solution was stirred at RT for 5 hr. The reaction was purified directly by column chromatography (C18, 0 to 100% acetonitrile in water + 0.1% TFA) to yield (2R,3R)-N-((1S)-1-cyclopentyl-2-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa- 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- 25 yl)amino)-2-oxoethyl)-N-methyl-3-phenyltetrahydrofuran-2-carboxamide (23.0 mg) as a yellow solid after lyophilization. LC/MS (Peptide method): M/Z = 1221 [M+H]+. Step c: To a solution of (2R,3R)-N-((1S)-1-cyclopentyl-2-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-30 ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-2-oxoethyl)-N-methyl-3-phenyltetrahydrofuran-2- carboxamide (23.0 mg) in THF (0.25 mL) at RT was added TBAF in THF (686.2 L, 0.10 molar) dropwise. The reaction mixture was allowed to stir at RT for 2 hr under nitrogen. The reaction was diluted with EtOAc and water and stirred vigorously. The layers were separated. The aq. layer 35 was extracted with EtOAc (3x), and the combined organic extracts were washed with brine, dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography (C18, 0 to 100% acetonitrile in water + 0.1% NH4OH) to give (2R,3R)-N-((1S)-1-cyclopentyl-2- (((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1- yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-40 bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-2-oxoethyl)-N-methyl-3- phenyltetrahydrofuran-2-carboxamide (axial chirality as shown in structure)(6.3 mg) as a white PAT059646-WO-PCT 5 solid. LC/MS (Peptide method): M/Z = 1064 [M+H]+.1H NMR (400 MHz, DMSO-d6) 9.13 (s, 1H), 8.45 (d, J = 2.9 Hz, 1H), 7.87 (s, 1H), 7.67 (d, J = 8.4 Hz, 1H), 7.58 (d, J = 8.6 Hz, 1H), 7.49 - 7.45 (m, 1H), 7.28 - 7.26 (m, 2H), 7.25 - 7.24 (m, 1H), 7.24 - 7.21 (m, 2H), 7.16 (t, J = 7.1 Hz, 1H), 7.08 (s, 1H), 6.94 (s, 1H), 6.54 (s, 1H), 5.36 - 5.28 (m, 1H), 5.09 (d, J = 7.7 Hz, 1H), 4.81 (d, J = 11.7 Hz, 1H), 4.32 (d, J = 11.0 Hz, 1H), 4.29 - 4.25 (m, 1H), 4.24 - 4.21 (m, 1H), 4.17 - 4.14 10 (m, 1H), 4.14 - 4.12 (m, 1H), 4.11 - 4.06 (m, 1H), 3.90 - 3.83 (m, 2H), 3.81 - 3.76 (m, 1H), 3.73 (d, J = 11.3 Hz, 1H), 3.58 (d, J = 10.9 Hz, 1H), 3.27 - 3.25 (m, 4H), 3.18 (d, J = 12.0 Hz, 1H), 3.11 (s, 3H), 2.81 - 2.76 (m, 1H), 2.67 (s, 3H), 2.66 - 2.61 (m, 3H), 2.46 - 2.44 (m, 4H), 2.39 - 2.35 (m, 1H), 2.28 - 2.23 (m, 1H), 2.21 (s, 3H), 1.93 - 1.86 (m, 1H), 1.35 (d, J = 6.3 Hz, 3H), 1.20 - 1.14 (m, 6H), 0.99 - 0.95 (m, 3H), 0.82 - 0.79 (m, 4H), 0.73 - 0.63 (m, 2H), 0.45 (s, 3H), 0.12 (s, 1H), - 15 0.02 (s, 2H). HRMS m/z [M+H]+ = 1063.6038. The following compounds of Table 7 were synthesized using the above procedure or modifications to the above procedure using the corresponding macrocycle, aminoacid, and acid intermediates. Note: axial chirality of compounds is as shown in the structures in Table 7. 20 Table 7: PanRAS inhibitor compounds 184 PAT059646-WO-PCT 5 Compound 110 Note: axial chirality of compounds is as shown in below scheme. (1r,2R,3S)-N-((61R,65S,66S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1- yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-2(4,2)-thiazola-1(5,3)-indola- 10 6(3,5)-bicyclo[4.1.0]heptanacycloundecaphane-4-yl)-2,3-dimethylcyclopropane-1-carboxamide Procedure Step a: To a solution of Intermediate 6 (trifluoroacetate salt; 8.3 mg) in DCM (1.0 mL) was added TFA (49.5 mg) under a nitrogen atmosphere. The reaction mixture stirred at RT for 2 hr. The 15 reaction mixture was concentrated under reduced pressure to yield a crude yellow oil, which was triturated with Et2O and dried to yield (61R,65S,66S,4S,Z)-4-amino-11-ethyl-12-(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-11H-8-oxa-63,64-diaza- 2(4,2)-thiazola-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptanacycloundecaphane-5,7-dione bis- trifluoroacetate salt (8.4 mg) as yellow solid. LC/MS (Peptide method): M/Z = 763 [M+Na]+. 20 PAT059646-WO-PCT 5 Step b: To a solution of (61R,65S,66S,4S,Z)-4-amino-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-11H-8-oxa-63,64-diaza-2(4,2)-thiazola-1(5,3)- indola-6(3,5)-bicyclo[4.1.0]heptanacycloundecaphane-5,7-dione bis-trifluoroacetate salt (8.4 mg) and (1r,2R,3S)-2,3-dimethylcyclopropane-1-carboxylic acid (2.0 mg) in NMP (0.5 mL) under a nitrogen atmosphere were added DIPEA (15 μL) and COMU (7.4 mg) at 0°C. The solution was 10 stirred at 0°C for 1 hr, and then purified by column chromatography (C18, 10 to 45% acetonitrile in water + formic acid) to yield (1r,2R,3S)-N-((61R,65S,66S,4S,Z)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1- yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-2(4,2)-thiazola-1(5,3)-indola- 6(3,5)-bicyclo[4.1.0]heptanacycloundecaphane-4-yl)-2,3-dimethylcyclopropane-1-carboxamide 15 (axial chirality as shown in structure) (4.7 mg) as a white solid after lyophilization.1H NMR (400 MHz, DMSO-d6) 8.46 (dd, J = 14.9, 2.3 Hz, 2H), 8.39 (d, J = 9.0 Hz, 1H), 7.81 (s, 1H), 7.72 (dd, J = 8.7, 1.7 Hz, 1H), 7.56 (d, J = 8.7 Hz, 1H), 7.23 (d, J = 2.9 Hz, 1H), 5.46 (d, J = 6.4 Hz, 1H), 4.80 (d, J = 11.8 Hz, 1H), 4.63 (d, J = 11.8 Hz, 1H), 4.34 – 4.09 (m, 4H), 3.73 (d, J = 10.7 Hz, 1H), 3.58 (d, J = 10.7 Hz, 1H), 3.27 (d, J 20 = 4.9 Hz, 5H), 3.19 (s, 4H), 3.15 (d, J = 5.4 Hz, 2H), 2.95 (d, J = 14.4 Hz, 1H), 2.22 (s, 3H), 2.04 – 1.95 (m, 1H), 1.34 (d, J = 6.1 Hz, 3H), 1.24 (s, 6H), 1.17 – 1.11 (m, 3H), 1.06 (dd, J = 8.7, 5.3 Hz, 6H), 0.95 – 0.88 (m, 7H), 0.36 (s, 3H). LC/MS (Peptide method): M/Z = 837 [M+H]+. Compound 111 25 Note: axial chirality of compounds is as shown in below scheme. N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola- 6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxobutan-2-yl)-N-methylmorpholine-4-carboxamide 30 PAT059646-WO-PCT 5 Step a: To a solution of Intermediate 5 (16.0 mg) in DCM (0.50 mL) at RT was added TFA (104 L). The resulting solution was stirred at RT for 30 min, and then concentrated under reduced pressure. The resulting residue was azeotroped with DCM under reduced pressure. To the residue, a solution of Intermediate 7, DIPEA (16.9 L) and HATU (14.8 mg) in NMP (85 L) and DMF (0.250 mL) was added. The reaction mixture was stirred at RT for 3 hr, and then TBAF in 10 THF (40 L, 1.0 molar) was added dropwise. After stirring for 16 hr at RT, the reaction mixture was diluted with EtOAc and water and stirred vigorously. The layers were separated. The aq. layer was extracted with EtOAc (3x), and the combined organic extracts were washed with brine, dried over Na2SO4, filtered, and concentrated. The residue was purified by column chromatography (C18, 0 to 100% acetonitrile in water + 0.1% NH4OH) to yield N-((2S)-1-15 (((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1- yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)- N-methylmorpholine-4-carboxamide (axial chirality as shown in structure)(5.7 mg) as a white solid.1H NMR (400 MHz, DMSO-d6) 9.33 (s, 1H), 8.45 (d, J = 2.9 Hz, 1H), 8.06 (d, J = 8.6 Hz, 20 1H), 7.89 (s, 1H), 7.59 (d, J = 8.5 Hz, 1H), 7.46 (d, J = 8.6 Hz, 1H), 7.25 (s, 1H), 7.13 (s, 1H), 6.95 (s, 1H), 6.38 (s, 1H), 5.49 – 5.43 (m, 1H), 4.95 (d, J = 11.5 Hz, 1H), 4.32 – 4.01 (m, 4H), 3.93 (d, J = 11.7 Hz, 1H), 3.81 (d, J = 11.0 Hz, 1H), 3.79 – 3.72 (m, 1H), 3.68 – 3.56 (m, 5H), 3.29 – 3.26 (m, 4H), 3.25 – 3.19 (m, 4H), 3.13 – 3.11 (m, 4H), 2.71 – 2.66 (m, 4H), 2.62 (s, 3H), 2.34 – 2.32 (m, 2H), 2.24 – 2.20 (m, 2H), 2.09 – 1.99 (m, 4H), 1.36 (d, J = 6.2 Hz, 3H), 1.23 – 1.23 (m, 2H), 25 0.97 (t, J = 7.1 Hz, 3H), 0.85 – 0.81 (m, 7H), 0.77 (d, J = 6.6 Hz, 3H), 0.47 (s, 3H), 0.18 (s, 1H). HRMS m/z [M+H]+ : 976.5706. LC/MS (Peptide method): M/Z = 977 [M+H]+. Compound 112 Note: axial chirality of compounds is as shown in below scheme. 30 (2R,3S)-N-((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola- 6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopent- 4-en-2-yl)-N-methyl-3-phenyloxetane-2-carboxamide
PAT059646-WO-PCT 5 Step a: To a solution of Intermediate 5 (60 mg) in DCM (994 L) was added TFA (199 L). The reaction was stirred at RT for 40 min., after which time it was concentrated. The resulting yellow foam was dissolved in DCM and concentrated to dryness; this process was repeated (3x). To the 10 remaining residue was triturated with Et2O. The resulting mixture was concentrated to afford (61R,65S,66S,4S)-4-amino-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin- 3-yl)-10,10-dimethyl-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-5,7-dione, trifluoroacetate salt (73 mg) as a yellow foam. LC/MS (Peptide method): M/Z = 906.5700 [M+H]+. 15 Step b: To a vial charged with Intermediate 23 (15.6 mg) and HATU (24.4 mg) was charged DMF (641 L) and DIPEA (41.4 mg, 55.8 L) and the mixture was stirred at RT for 2 min. (61R,65S,66S,4S)-4-amino-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin- 3-yl)-10,10-dimethyl-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- 20 bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-5,7-dione trifluoroacetate salt (40 mg) was then added and the resulting solution was stirred at RT for 35 min. Water was added to reaction mixture and precipitate was filtered off. The precipitate was re-dissolved in DCM, dried on Na2SO4, filtered, and concentrated to afford crude tert-butyl ((2S,3R)-1-(((61R,65S,66S,4S)-11- ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-25 dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana- 2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopent-4-en-2-yl)(methyl)carbamate (32 mg) which was carried forward to next step. LC/MS (Peptide Method): M/Z = 1131.7100 [M+H]+. PAT059646-WO-PCT 5 Step c: To a solution of tert-butyl ((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopent-4-en-2-yl)(methyl)carbamate (32 10 mg) in DCM (471 L) was added TFA (94.3 L). The reaction solution was stirred at RT for 2 hr. After this time, the solution was diluted with DCM and concentrated to dryness; this process was repeated (3x). The resulting residue was triturated with Et2O and the mixture was concentrated again to afford (2S,3R)-N-((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-15 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)-3- methyl-2-(methylamino)pent-4-enamide, trifluoroacetate salt (38.8 mg). LC/MS (Peptide Method): M/Z = 1031.6500 [M+H]+. Step d: To a vial containing Intermediate 10 (10.1 mg) and HATU (21.5 mg) was charged DMF 20 (400 L) and DIPEA (36.5 mg, 49.2 L) was added. The solution was allowed to stir for 5 min., after which time it was charged to a vial containing (2S,3R)-N-((61R,65S,66S,4S)-11-ethyl-12-(2- ((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)pent-4-enamide, trifluoroacetate salt 25 (38.8 mg). The solution was then stirred at RT for 3 hr. After this time, TMAF (7.9 mg) was charged to the reaction and stirring continued for 3 hr, upon which time the reaction mixture was triturated with water and the precipitate was collected via vacuum filtration. The collected solids were dissolved in DCM and the aq. filtrate was extracted with DCM (20 mL, 3x). Organic extracts were combined and dried over Na2SO4. Filtration and concentration afforded a residue which was 30 reconstituted in DMSO and purified by column chromatography (C18; 10 to 100% acetonitrile + 0.1% formic acid) to afford (2R,3S)-N-((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(2- ((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8- oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)amino)-3-methyl-1-oxopent-4-en-2-yl)-N-methyl-3-phenyloxetane-2-carboxamide (13 mg) as a 35 white solid after lyophilization.1H NMR (400 MHz, DMSO) 9.36 (s, 1H), 8.46 (d, J = 2.9 Hz, 1H), 7.87 (s, 1H), 7.72 (d, J = 8.5 Hz, 1H), 7.58 (d, J = 8.6 Hz, 1H), 7.52 – 7.45 (m, 3H), 7.34 – 7.22 (m, 5H), 7.09 (s, 1H), 6.95 (s, 1H), 6.49 (d, J = 26.6 Hz, 2H), 5.97 (d, J = 7.7 Hz, 1H), 5.58 – 5.44 (m, 1H), 5.36 – 5.30 (m, 1H), 5.08 – 4.97 (m, 1H), 4.85 (d, J = 11.6 Hz, 1H), 4.73 (dd, J = 18.3, 13.8 Hz, 2H), 4.55 – 4.38 (m, 4H), 4.29 – 4.18 (m, 1H), 4.17 – 4.04 (m, 3H), 3.88 (d, J = 11.6 Hz, 40 1H), 3.72 (d, J = 10.9 Hz, 1H), 3.59 (d, J = 10.8 Hz, 1H), 3.19 (d, J = 12.3 Hz, 2H), 3.11 (s, 4H), 2.71 – 2.57 (m, 5H), 2.43 – 2.28 (m, 7H), 2.20 – 2.12 (m, 1H), 1.36 (d, J = 6.1 Hz, 4H), 1.16 (d, J PAT059646-WO-PCT 5 = 8.0 Hz, 2H), 0.97 (t, J = 7.1 Hz, 3H), 0.80 (s, 4H), 0.46 (s, 3H), 0.13 (d, J = 5.2 Hz, 1H), -0.09 (d, J = 6.8 Hz, 3H). LC/MS (Peptide Method): M/Z = 1035.5699 [M+H]+. Compound 113 Note: axial chirality of compounds is as shown in below scheme. 10 (2R,3R)-N-((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola- 6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxopentan-2-yl)-N-methyl-3-phenyltetrahydrofuran-2-carboxamide 15 Procedure Step a: To a solution of ((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-20 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)carbamate (90 mg) in DCM (715 L) was added TFA (179 L). The reaction was stirred at RT for approximately 1.5 hr, after which time it was concentrated. The resulting yellow foam was dissolved in DCM and concentrated to dryness; this process was repeated (3x). To the remaining residue was added Et2O. The mixture was sonicated and decanted (2x). The remaining solid was25 dried to afford (61R,65S,66S,4S)-4-amino-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64- diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-5,7-dione, PAT059646-WO-PCT 5 trifluoroacetate salt (116 mg) as a yellow foam. LC/MS (RXMON-Acidic method): M/Z = 906.7 [M+H]+. Step b: To a vial charged with Intermediate 24 (14 mg) and HATU (20 mg) was charged DMF (513 L) and DIPEA (27 mg, 35.8 L) and the mixture was stirred at RT for 2 min. 10 (61R,65S,66S,4S)-4-amino-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin- 3-yl)-10,10-dimethyl-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-5,7-dione trifluoroacetate salt (33.5 mg) was then added and the resulting solution was stirred at RT for 30 min. Water was added to reaction mixture and precipitate was filtered off. The precipitate was re-dissolved in DCM and15 passed through a filter to afford crude tert-butyl ((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)- 1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopentan-2-yl)(methyl)carbamate (30 mg) as a beige solid which was carried forward to next step. LC/MS (Peptide Method): M/Z = 20 1133.7100 [M+H]+. Step c: To a solution of tert-butyl ((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- 25 benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopentan-2-yl)(methyl)carbamate (30 mg) in DCM (441 L) was added TFA (88 L). The reaction solution was stirred at RT for 40 min. After this time, the solution was diluted with DCM and concentrated to dryness; this process was repeated (3x). Et2O was then added and the material was concentrated again to afford (2S,3R)- N-((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-30 10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)-3-methyl-2- (methylamino)pentanamide trifluoroacetate salt (33 mg) as a yellow powder. LC/MS (Peptide Method): M/Z = 1033.6700 [M+H]+. 35 Step d: To a vial containing (2R,3R)-3-phenyltetrahydrofuran-2-carboxylic acid (8.5 mg) and HATU (17 mg) was charged NMP (400 L) and DIPEA (29 mg, 39 L) was added. The solution was allowed to stand for 2 min., after which time it was charged to a vial containing (2S,3R)-N-((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-40 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)-3- methyl-2-(methylamino)pentanamide trifluoroacetate salt (33 mg). The solution was then stirred PAT059646-WO-PCT 5 at RT for 18 hr. After this time, the solution containing crude (2R,3R)-N-((2S,3R)-1- (((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopentan-2- yl)-N-methyl-3-phenyltetrahydrofuran-2-carboxamide was directly subjected to the next step. 10 LC/MS (Peptide Method): M/Z = 1208.7400 [M+H]+. Step e: To a solution of crude (2R,3R)-N-((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-15 benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopentan-2-yl)-N-methyl-3- phenyltetrahydrofuran-2-carboxamide in (400 L) was added TMAF (6.2 mg). The reaction was stirred at RT for 5 hr, after which time DCM and water were added. The aq. layer was then extracted with DCM (4x). Organic extracts were combined, dried, filtered and concentrated. The residue was purified by column chromatography (C18; 10 to 100% acetonitrile in water + 0.1%20 formic acid) to afford (2R,3R)-N-((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64- diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)- 3-methyl-1-oxopentan-2-yl)-N-methyl-3-phenyltetrahydrofuran-2-carboxamide (7 mg) as a white solid after lyophilization.1H NMR (400 MHz, DMSO) 9.17 (s, 1H), 8.45 (d, J = 2.9 Hz, 1H), 7.87 25 (s, 1H), 7.63 (d, J = 8.3 Hz, 1H), 7.58 (d, J = 8.6 Hz, 1H), 7.47 (dd, J = 8.6, 1.7 Hz, 1H), 7.28 (d, J = 7.5 Hz, 3H), 7.25 – 7.20 (m, 4H), 7.16 (d, J = 7.1 Hz, 1H), 7.09 (s, 1H), 6.92 (d, J = 14.1 Hz, 2H), 6.60 (s, 1H), 6.49 (s, 1H), 5.32 (d, J = 5.9 Hz, 1H), 5.10 (d, J = 7.7 Hz, 1H), 4.81 (d, J = 11.7 Hz, 1H), 4.37 (d, J = 10.5 Hz, 1H), 4.30 – 4.18 (m, 3H), 4.18 – 4.05 (m, 4H), 3.87 (q, J = 7.5 Hz, 2H), 3.81 – 3.68 (m, 2H), 3.58 (d, J = 10.3 Hz, 1H), 3.18 (d, J = 12.1 Hz, 2H), 3.11 (s, 3H), 2.78 30 (d, J = 9.1 Hz, 2H), 2.64 (s, 3H), 2.38 – 2.28 (m, 3H), 2.21 (s, 3H), 1.51 (s, 1H), 1.35 (d, J = 6.2 Hz, 4H), 1.17 (s, 3H), 0.97 (t, J = 7.1 Hz, 4H), 0.81 (s, 3H), 0.67 (d, J = 5.4 Hz, 4H), 0.46 (s, 3H), 0.09 (d, J = 22.5 Hz, 2H), -0.10 (d, J = 6.7 Hz, 3H. LC/MS (Peptide Method): M/Z = 1051.600 [M+H]+. 35 The following compounds of Table 8 were synthesized using the above procedure or modifications of the above procedure using the corresponding macrocycle, amino acid, and carboxylic acid. Note: axial chirality of compounds is as shown in structures in Table 8. For Table 8, an asterisk, *, indicates that PyBroP/DIPEA/1,4-dioxane was used for acylation of the corresponding N-methylated amino acid. Unless otherwise noted, HRMS was measured via the 40 Peptide Method with a MS detector. PAT059646-WO-PCT 5 Table 8: PanRAS inhibitor compounds PAT059646-WO-PCT 194 PAT059646-WO-PCT PAT059646-WO-PCT 196 PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT 5 Compound 123 Note: axial chirality of compounds is as shown in below scheme. (2R,3S)-N-((2S,3R)-1-(((63S,4S)-11-ethyl-25-hydroxy-12-((M)-2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-61,62,63,64,65,66-hexahydro-11H-8-10 oxa-1(5,3)-indola-6(1,3)-pyridazina-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxopent-4-en-2-yl)-N-methyl-3-phenyloxetane-2-carboxamide PAT059646-WO-PCT 5 Procedure Step a: MeOH (80 mL) was carefully added to a mixture of Intermediate 26a (9.5 g) and Pd(OH)2 on carbon (602 mg, 10% wt). The mixture was sparged with nitrogen for 3 min. and stirred under H2 (balloon) for 2 hr. The mixture was filtered through a pad of Celite®. The filtrate was10 concentrated to afford tert-butyl ((12R,63S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(piperazin-1- yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-61,62,63,64,65,66-hexahydro- 11H-8-oxa-1(5,3)-indola-6(1,3)-pyridazina-2(1,3)-benzenacycloundecaphane-4-yl)carbamate (8.4 g). LC/MS (RXNMON-Acidic method): M/Z = 980.9 [M+H]+. 15 Step b: To a solution of tert-butyl ((12R,63S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(piperazin- 1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-61,62,63,64,65,66-hexahydro- 11H-8-oxa-1(5,3)-indola-6(1,3)-pyridazina-2(1,3)-benzenacycloundecaphane-4-yl)carbamate (3.0 g) in MeOH (29 mL) was added formaldehyde (701 mg, 643 L, 37 wt% in water). The reaction was stirred for 10 min. and sodium triacetoxyborohydride (1.83 g) was added. The 20 reaction was stirred 30 min. Sat. aq. NaHCO3 solution was slowly added to the reaction. The aq. layer was extracted with DCM (2x). The combined DCM extracts were dried over Na2SO4 and concentrated to afford tert-butyl ((12R,63S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)- 61,62,63,64,65,66-hexahydro-11H-8-oxa-1(5,3)-indola-6(1,3)-pyridazina-2(1,3)- 25 benzenacycloundecaphane-4-yl)carbamate (2.86 g). LC/MS (peptide): M/Z = 994.9 [M+H]+. Step c: To a solution of tert-butyl ((12R,63S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)- PAT059646-WO-PCT 5 61,62,63,64,65,66-hexahydro-11H-8-oxa-1(5,3)-indola-6(1,3)-pyridazina-2(1,3)- benzenacycloundecaphane-4-yl)carbamate (3.57 g) in DCM (36 mL) and triethylsilane (292 mg, 401 L) was added TFA (13.1 g, 8.85 mL) at 0°C. The reaction was stirred at RT for 2 hr. The reaction was concentrated, and the residue was dissolved in DCM. The DCM solution was washed with sat. aq. NaHCO3 solution (0°C). The aq. layer was extracted with DCM (2x). The10 combined DCM extracts were dried over Na2SO4 and concentrated to afford (12R,63S,4S)-4- amino-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl- 25-((triisopropylsilyl)oxy)-61,62,63,64,65,66-hexahydro-11H-8-oxa-1(5,3)-indola-6(1,3)-pyridazina- 2(1,3)-benzenacycloundecaphane-5,7-dione trifluoroacetate salt (3.33 g). LC/MS (peptide): M/Z = 894.5600 [M+H]+. 15 Step d: To a solution of (12R,63S,4S)-4-amino-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-25-((triisopropylsilyl)oxy)-61,62,63,64,65,66- hexahydro-11H-8-oxa-1(5,3)-indola-6(1,3)-pyridazina-2(1,3)-benzenacycloundecaphane-5,7- dione trifluoroacetate salt (3.33 g) and Intermediate 23 (1.08 g) in MeCN (33 mL) and 2-methyl 20 THF (8.3 mL) at 0°C was added DIPEA (2.74 g, 3.7 mL) and COMU (1.97 g). The reaction was warmed up to RT and stirred for 30 min. The reaction was concentrated, dissolved in EtOAc, and washed with water, sat. aq. NaHCO3 and brine. The EtOAc layer was dried over Na2SO4 and concentrated. The residue was purified by column chromatography (SiO2; 0 to 20% MeOH in DCM) to afford tert-butyl ((2S,3R)-1-(((12R,63S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-25 methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)- 61,62,63,64,65,66-hexahydro-11H-8-oxa-1(5,3)-indola-6(1,3)-pyridazina-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopent-4-en-2-yl)(methyl)carbamate (3.62 g). LC/MS (peptide): M/Z = 1119.7200 [M+H]+. 30 Step e: To a solution of tert-butyl ((2S,3R)-1-(((12R,63S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5- (4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)- 61,62,63,64,65,66-hexahydro-11H-8-oxa-1(5,3)-indola-6(1,3)-pyridazina-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopent-4-en-2-yl)(methyl)carbamate (3.62 g) in DCM (31 mL) and anisole (332 mg, 333 L) at was added TFA (11.2 g, 7.56 mL) at 0°C. The 35 reaction was stirred at RT for 30 min. The reaction was concentrated, and the residue was dissolved in DCM. The DCM solution was washed with sat. aq. NaHCO3 solution. The aq. layer was extracted with DCM (3x). The combined DCM extracts were passed through a phase separator and concentrated to afford (2S,3R)-N-((12R,63S,4S)-11-ethyl-12-(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-40 ((triisopropylsilyl)oxy)-61,62,63,64,65,66-hexahydro-11H-8-oxa-1(5,3)-indola-6(1,3)-pyridazina- PAT059646-WO-PCT 5 2(1,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)pent-4-enamide trifluoroacetate salt (3.27 g). LC/MS (peptide): M/Z = 1019.7200 [M+H]+. Step f: To a solution of (2S,3R)-N-((12R,63S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-10 61,62,63,64,65,66-hexahydro-11H-8-oxa-1(5,3)-indola-6(1,3)-pyridazina-2(1,3)- benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)pent-4-enamide trifluoroacetate salt (3.27 g) and Intermediate 10 (oxetane) (868 mg) in MeCN (24 mL) and 2-methyl THF (6.1 mL) at 0°C was added DIPEA (2.36 g, 3.2 mL) and HATU (1.8 g). The reaction was warmed up to RT and stirred for 20 hr. The reaction was concentrated, dissolved in EtOAc and filtered. The filtrate 15 was washed with water, sat. aq. NaHCO3 and brine. The EtOAc layer was dried over Na2SO4 and concentrated. The residue was purified by column chromatography (SiO2; 0 to 20% MeOH in DCM) to afford (2R,3S)-N-((2S,3R)-1-(((12R,63S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)- 61,62,63,64,65,66-hexahydro-11H-8-oxa-1(5,3)-indola-6(1,3)-pyridazina-2(1,3)-20 benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopent-4-en-2-yl)-N-methyl-3- phenyloxetane-2-carboxamide (3.09 g).1H NMR (400 MHz, DMSO) 8.46 (d, J = 2.8 Hz, 1H), 7.90 (s, 1H), 7.76 (d, J = 8.3 Hz, 1H), 7.61 (d, J = 8.6 Hz, 1H), 7.52 – 7.41 (m, 4H), 7.36 – 7.21 (m, 4H), 7.02 (s, 1H), 6.62 (s, 1H), 5.83 (d, J = 8.1 Hz, 1H), 5.56 – 5.33 (m, 3H), 5.05 – 4.94 (m, 1H), 4.77 – 4.42 (m, 4H), 4.42 – 3.91 (m, 5H), 3.84 – 3.47 (m, 3H), 3.29 – 3.22 (m, 4H), 3.02 (d, 25 J = 3.0 Hz, 3H), 2.86 – 2.57 (m, 5H), 2.48 – 2.43 (m, 4H), 2.31 (s, 2H), 2.22 (s, 3H), 2.13 – 1.90 (m, 2H), 1.78 (d, J = 11.9 Hz, 1H), 1.65 – 1.46 (m, 2H), 1.34 (p, J = 7.7 Hz, 7H), 1.20 – 1.11 (m, 16H), 1.11 – 1.08 (m, 2H), 1.07 – 0.99 (m, 3H), 0.67 (d, J = 40.8 Hz, 6H), -0.11 (d, 3H). LC/MS (FinalAnalysis-Basic): M/Z = 1179.8 [M+H]+. 30 Step g: To a solution of (2R,3S)-N-((2S,3R)-1-(((12R,63S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)- 5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)- 61,62,63,64,65,66-hexahydro-11H-8-oxa-1(5,3)-indola-6(1,3)-pyridazina-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopent-4-en-2-yl)-N-methyl-3- phenyloxetane-2-carboxamide (109 mg) in DMF (2.0 mL) at 0°C open to air was 35 added tetramethylammonium fluoride (12.9 mg). The reaction was stirred for 45 min, upon which time the reaction was diluted with DMSO and purified via ISCO (30 g C18 gold column, 10%-80% CH3CN and water, 0.1% formic acid). Pure fractions were combined, frozen, and lyophilized to yield (2R,3S)-N-((2S,3R)-1-(((63S,4S)-11-ethyl-25-hydroxy-12-((M)-2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-61,62,63,64,65,66-hexahydro-11H-8-40 oxa-1(5,3)-indola-6(1,3)-pyridazina-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxopent-4-en-2-yl)-N-methyl-3-phenyloxetane-2-carboxamide (62.5 mg) as a white powder. 1H PAT059646-WO-PCT 5 NMR (400 MHz, DMSO) 9.33 (s, 1H), 8.45 (d, J = 2.8 Hz, 1H), 8.22 (s, 1H), 7.90 (s, 1H), 7.75 (d, J = 8.6 Hz, 1H), 7.58 (d, J = 8.6 Hz, 1H), 7.55 – 7.46 (m, 3H), 7.32 (t, J = 7.5 Hz, 2H), 7.24 (t, J = 4.9 Hz, 3H), 7.01 (s, 1H), 6.47 (s, 1H), 5.99 (d, J = 7.8 Hz, 1H), 5.53 (m, 1H), 5.40 – 5.27 (m, 2H), 5.02 (t, J = 6.4 Hz, 1H), 4.79 – 4.65 (m, 2H), 4.53 (d, J = 9.6 Hz, 1H), 4.43 (m, 2H), 4.29 – 4.14 (m, 2H), 4.08 (m, 2H), 3.80 (t, J = 11.9 Hz, 1H), 3.63 (d, J = 10.9 Hz, 1H), 3.55 (d, J = 10.9 10 Hz, 1H), 3.27 (d, J = 4.0 Hz, 4H), 3.06 (s, 3H), 2.77 – 2.65 (m, 5H), 2.46 (m, 4H), 2.35 (s, 3H), 2.21 (s, 3H), 2.14 (m, 1H), 1.98 (d, J = 11.5 Hz, 1H), 1.79 (d, J = 12.0 Hz, 1H), 1.62 (d, J = 12.9 Hz, 1H), 1.51 (m, 1H), 1.34 (d, J = 6.1 Hz, 3H), 1.00 (t, J = 7.0 Hz, 3H), 0.76 (s, 3H), 0.55 (s, 2H), -0.08 (d, J = 6.9 Hz, 3H). LC/MS (Peptide Method) M/Z = 1023.5800 [M+H]+. 15 Compound 124 Note: axial chirality of compounds is as shown in below scheme. (2R,3S)-1-(cyanomethyl)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64- diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)- 20 3-methyl-1-oxobutan-2-yl)-N-methyl-3-phenylazetidine-2-carboxamide Procedure Step a: To a solution of Intermediate 36 (30 mg) and DIPEA (13 mg, 18 L) in DMF (0.25 mL) was added 2-bromoacetonitrile (3.5 L). The reaction was stirred at RT for 30 min. The reaction 25 mixture was diluted with water and EtOAc. The aq. layer was extracted with EtOAc (3x), and the combined organic layers were passed through a phase separator and concentrated to afford ((2R,3S)-1-(cyanomethyl)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa- 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- 30 yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3-phenylazetidine-2-carboxamide as a yellow solid. LC/MS (Peptide Method) M/Z = 1217.7500 [M+H]+. Step b: To a solution of (2R,3S)-1-(cyanomethyl)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2- ((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-35 benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3-phenylazetidine- PAT059646-WO-PCT 5 2-carboxamide (18.1 mg) in DMF (0.5 mL) was charged with TMAF (7 mg). The mixture was briefly sonicated then stirred at for 15 min. The reaction was filtered, diluted with DMSO, and purified by preparatory HPLC (C18; 45 to 70% acetonitrile in water + 0.1% NH4OH). Desired fractions were concentrated, and the remaining residue was dissolved in DCM and washed with water. The aq. layer was extracted with DCM (3x), and combined organic extracts were passed 10 through a phase separator and concentrated. The resulting residue was dissolved in 1:1 acetonitrile : water and lyophilized to afford (2R,3S)-1-(cyanomethyl)-N-((2S)-1- (((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1- yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)- 15 N-methyl-3-phenylazetidine-2-carboxamide (4.4 mg) as a yellow solid. LC/MS (Peptide Method) M/Z = 1061.6100 [M+H]+.1H NMR (400 MHz, DMSO) 9.37 (s, 1H), 8.45 (d, J = 2.8 Hz, 1H), 7.92 – 7.85 (m, 2H), 7.61 – 7.52 (m, 3H), 7.49 – 7.44 (m, 1H), 7.29 – 7.22 (m, 3H), 7.22 – 7.16 (m, 1H), 7.09 (s, 1H), 7.01 – 6.97 (m, 1H), 6.54 – 6.49 (m, 1H), 5.29 – 5.19 (m, 1H), 4.78 (d, J = 11.8 Hz, 1H), 4.44 (d, J = 8.0 Hz, 1H), 4.30 – 4.20 (m, 2H), 4.16 – 4.08 (m, 2H), 3.97 – 3.84 (m, 20 3H), 3.84 – 3.76 (m, 1H), 3.76 – 3.70 (m, 1H), 3.66 – 3.57 (m, 2H), 3.26 (t, J = 5.4 Hz, 4H), 3.20 – 3.14 (m, 2H), 3.11 (d, J = 11.4 Hz, 4H), 2.85 – 2.72 (m, 2H), 2.67 – 2.61 (m, 1H), 2.59 – 2.55 (m, 3H), 2.45 (t, J = 5.0 Hz, 4H), 2.21 (s, 3H), 1.70 – 1.57 (m, 1H), 1.36 (d, J = 6.2 Hz, 3H), 1.26 – 1.22 (m, 2H), 1.21 – 1.15 (m, 2H), 0.97 (t, J = 7.1 Hz, 3H), 0.82 (s, 4H), 0.66 (d, J = 6.5 Hz, 3H), 0.45 (s, 2H), 0.19 – 0.12 (m, 1H), -0.27 (d, J = 6.6 Hz, 3H). 25 The following compounds of Table 9 were synthesized using the above procedure or modifications of the above procedure using the corresponding macrocycle, amino acid, and alkylation reagent. Note: axial chirality of compounds is as shown in structures in Table 9. Unless otherwise noted, HRMS was measured via the Peptide Method with a MS detector. 30 Table 9: PanRAS inhibitor compounds PAT059646-WO-PCT 5 Compound 126 Note: axial chirality of compounds is as shown in below scheme. methyl (2R,3S)-2-(((2S)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(2-((S)-1-methoxyethyl)-5- (4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-10 indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxobutan-2-yl)(methyl)carbamoyl)-3-phenylazetidine-1-carboxylate PAT059646-WO-PCT 5 Step a: To a solution of Intermediate 36 (77.6 mg) and NEt3 (34.7 mg, 48 L) in DCM (400 μL) at 0 °C was added methyl carbonochloridate (9.3 mg, 7.6 L). The reaction was allowed to warm to RT and stir for 1.5 hr. The reaction mixture was quenched with sat. aq. NaHCO3 solution and the 10 aq. layer was extracted with DCM (3x), and the combined organic extracts were passed through a phase separator and concentrated to afford methyl (2R,3S)-2-(((2S)-1-(((61R,65S,66S,4S)-11- ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana- 2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)(methyl)carbamoyl)-3- 15 phenylazetidine-1-carboxylate which was used directly in the next step. LC/MS (Peptide Method) M/Z = 1236.7500 [M+H]+. Step b: To a solution of (2R,3S)-1-(cyanomethyl)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1- methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-20 ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3-phenylazetidine-2- carboxamide (18 mg) in DMF (200 μL) was charged TMAF (3.2 mg). The mixture was briefly sonicated then stirred at for 15 min. The reaction was filtered, diluted with DMSO, and purified by preparatory HPLC (C18; 50 to 70% acetonitrile in water + 0.1% NH4OH) to afford methyl (2R,3S)-2-(((2S)-1-25 (((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(2-((S)-1-methoxyethyl)-5-(4-methylpiperazin-1-yl)pyridin- 3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana- 2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)(methyl)carbamoyl)-3- phenylazetidine-1-carboxylate (7.2 mg) as a white solid after lyophilization. LC/MS (Peptide Method) M/Z = 1080.6100 [M+H]+.1H NMR (400 MHz, DMSO) 9.44 – 9.16 (m, 1H), 8.51 – 8.21 (m, 1H), 7.93 – 7.79 30 (m, 1H), 7.62 – 7.50 (m, 1H), 7.45 (d, J = 8.6 Hz, 1H), 7.36 (d, J = 7.5 Hz, 2H), 7.32 – 7.24 (m, 3H), 7.24 – 7.17 (m, 1H), 7.16 – 7.09 (m, 1H), 6.97 – 6.84 (m, 1H), 6.84 – 6.62 (m, 1H), 6.58 – 6.42 (m, 1H), 5.49 (d, J = 8.6 Hz, 1H), 5.46 – 5.29 (m, 1H), 4.93 – 4.79 (m, 1H), 4.33 – 3.93 (m, 8H), 3.80 (s, 1H), 3.70 (d, J = 10.7 Hz, 1H), 3.59 (d, J = 10.8 Hz, 2H), 3.53 – 3.40 (m, 1H), 3.30 – 3.22 (m, 4H), 3.22 – 3.15 (m, 1H), 3.08 (s, 3H), 2.74 – 2.57 (m, 3H), 2.54 (s, 1H), 2.48 – 2.42 (m, 5H), 2.21 (s, 3H), 1.68 – 1.53 (m, 1H), 1.36 PAT059646-WO-PCT 5 (d, J = 6.2 Hz, 3H), 1.23 (s, 2H), 1.20 – 1.11 (m, 2H), 0.99 (t, J = 7.1 Hz, 3H), 0.78 (s, 5H), 0.64 (d, J = 6.4 Hz, 3H), 0.50 (s, 3H), 0.14 – 0.02 (m, 1H), -0.33 (d, J = 6.6 Hz, 3H). Compound 127 Note: axial chirality of compounds is as shown in below scheme. 10 (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(5-(4-(3-hydroxypropyl)piperazin- 1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)- indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxobutan-2-yl)-N-methyl-3-phenyltetrahydrofuran-2-carboxamide and (2R,3R)-N-((2S)-1- (((61R,65S,66S,4S)-11-ethyl-12-(5-(4-ethylpiperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-25-15 hydroxy-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)- N-methyl-3-phenyltetrahydrofuran-2-carboxamide 20 Procedure Step a: To a solution of Intermediate 27 (91 mg) in EtOH (2 mL) was added Pd(OH)2 on carbon (15 mg, 10% wt.). The reaction vessel was purged with hydrogen (3x). The mixture was stirred under hydrogen atmosphere at RT for 1.5 hr, after which time additional Pd(OH)2 on carbon (15 mg, 10% wt.) was added and the reaction resumed for 50 min. The reaction mixture was filtered25 and washed with DCM. Filtrate was concentrated to provide a mixture of (2R,3R)-N-((2S)-1- (((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)- N-methyl-3-phenyltetrahydrofuran-2-carboxamide and (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-11- PAT059646-WO-PCT 5 ethyl-12-(5-(4-ethylpiperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo- 25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana- 2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3- phenyltetrahydrofuran-2-carboxamide as a grey solid (79 mg). LC/MS (Peptide Method) M/Z = 1180.6801 [M+H]+ and M/Z = 1207.7400 [M+H]+, respectively. This mixture was carried forward 10 as-is to the next step. Step b: To the crude mixture of (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1- methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)- 11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-15 benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3- phenyltetrahydrofuran-2-carboxamide and (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12- (5-(4-ethylpiperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3-20 phenyltetrahydrofuran-2-carboxamide (27 mg) in DCE (1.1 mL) was added 3-((tert- butyldimethylsilyl)oxy)propanal (43 mg, 44 L). The mixture was stirred at RT for 10 min., after which time sodium triacetoxyhydroborate (49 mg) was added. The resulting mixture was stirred at RT for 1 hr, upon which time the reaction was diluted with DCM and quenched with brine and sat. aq. NaHCO3 (1:1). The aq. layer was extracted with DCM (3x). The combined organic extracts 25 were dried over Na2SO4, filtered, and concentrated. The remaining material was dissolved in DMF (750 μL) and treated with TMAF. The mixture was stirred at RT for 45 min., and then directly purified by preparatory HPLC (C4; 10 to 60% acetonitrile in water (+0.1% NH4OH)) to afford (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(5-(4-(3-hydroxypropyl)piperazin- 1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-30 indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxobutan-2-yl)-N-methyl-3-phenyltetrahydrofuran-2-carboxamide (8.7 mg) after lyophilization as a white solid.1H NMR (400 MHz, DMSO) 9.08 (s, 1H), 8.43 – 8.30 (m, 1H), 7.80 (s, 1H), 7.65 (d, J = 8.4 Hz, 1H), 7.51 (d, J = 8.8 Hz, 1H), 7.40 (d, J = 8.6 Hz, 1H), 7.31 – 7.12 (m, 5H), 7.12 – 6.98 (m, 2H), 6.91 – 6.68 (m, 2H), 6.52 (m, 1H), 5.30 (m, 1H), 5.08 (m, 1H), 4.77 (m, 1H), 4.36 35 (s, 1H), 4.22 – 4.01 (m, 5H), 3.88 – 3.60 (m, 4H), 3.51 (d, J = 10.7 Hz, 1H), 3.38 (m, 2H), 3.18 (d, J = 6.2 Hz, 4H), 3.12 – 2.97 (m, 4H), 2.81 – 2.67 (m, 2H), 2.60 (s, 5H), 2.34 – 2.16 (m, 4H), 1.69 – 1.46 (m, 3H), 1.29 (d, J = 6.2 Hz, 3H), 1.13 (m, 3H), 0.90 (m, 5H), 0.82 – 0.54 (m, 8H), 0.43 (m, 3H), 0.03 (s, 1H), -0.13 (d, J = 6.6 Hz, 3H). LC/MS (HRMS_2min) M/Z = 1081.6126 [M+H]+. 40 The following compounds of Table 10 were synthesized using the above procedure or modifications of the above procedure using the corresponding macrocycle and aldehyde with PAT059646-WO-PCT 5 subsequent isolation from the corresponding mixture. Compound 128 was isolated in the same reaction that generated compound 129. Note: axial chirality of compounds is as shown in structures in Table 10. Table 10: PanRAS inhibitor compounds PAT059646-WO-PCT 5 Compound 130 Note: axial chirality of compounds is as shown in below scheme. (2R,3S)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(5-(4-(4-hydroxybutyl)piperazin-1-10 yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)- indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxobutan-2-yl)-N-methyl-3-phenyloxetane-2-carboxamide 1 PAT059646-WO-PCT 5 Procedure Step a: To a suspension of Intermediate 28 (97 mg) in THF (2 ml) and EtOH (2 ml) was added Pd(OH)2 on carbon (50 mg, 10% wt.). The reaction vessel was evacuated and backfilled with hydrogen. After stirring under an atmosphere of H2 at RT for 2 hr, the reaction mixture was filtered10 and concentrated to afford a mixture containing (2R,3S)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12- (5-(4-(4-hydroxybutyl)piperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7- dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana- 2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3- phenyloxetane-2-carboxamide (80 mg) and (2R,3S)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-15 ((S)-1-methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3-phenyloxetane- 2-carboxamide. This material was carried forward directly to the next step. HRMS m/z = 1038.7500 [M+H]+. 20 Step b: To a mixture containing (2R,3S)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(5-(4-(4- hydroxybutyl)piperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3-phenyloxetane- 25 2-carboxamide (48 mg) in THF (1 mL) was added 1N TBAF in THF (21 mg, 80 μL). After stirring at RT for 1 hr, the reaction mixture was diluted with EtOAc and washed with water and brine. The PAT059646-WO-PCT 5 EtOAc extract was concentrated and the residue purified by SFC (Prep_SFC8) to afford (2R,3R)- N-((2S)-1-(((61R,65S,66S,4S)-12-(5-(4-cyclopropylpiperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin- 3-yl)-11-ethyl-25-hydroxy-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)- N-methyl-3-phenyltetrahydrofuran-2-carboxamide (7.8 mg).1H NMR (400 MHz, CDCl3) 8.47 (d, 10 J = 2.8 Hz, 1H), 8.01 (d, J = 1.7 Hz, 1H), 7.61 (dd, J = 8.6, 1.6 Hz, 1H), 7.46 (d, J = 7.3 Hz, 2H), 7.41 (d, J = 8.6 Hz, 1H), 7.34 (t, J = 7.5 Hz, 2H), 7.28 (s, 1H), 7.18 – 7.12 (m, 2H), 7.03 (d, J = 2.9 Hz, 1H), 6.73 (d, J = 13.5 Hz, 1H), 6.60 (s, 1H), 5.74 (d, J = 8.7 Hz, 1H), 5.51 – 5.42 (m, 1H), 5.26 (dd, J = 8.2, 6.3 Hz, 1H), 4.97 (t, J = 6.2 Hz, 1H), 4.56 – 4.42 (m, 2H), 4.31 (m, 4H), 4.17 (m, 1H), 3.87 (dd, J = 28.8, 11.4 Hz, 2H), 3.72 (d, J = 10.9 Hz, 1H), 3.65 – 3.55 (m, 3H), 3.38 (s, 3H), 15 3.34 (t, J = 5.2 Hz, 4H), 3.10 (dd, J = 13.4, 3.7 Hz, 2H), 2.81 – 2.71 (m, 5H), 2.65 (dd, J = 14.0, 9.9 Hz, 1H), 2.54 (d, J = 5.7 Hz, 3H), 2.41 (s, 3H), 1.86 (m, 1H), 1.77 – 1.66 (m, 4H), 1.45 (d, J = 6.1 Hz, 3H), 1.31 – 1.19 (m, 3H), 1.05 – 0.92 (m, 7H), 0.80 (d, J = 6.4 Hz, 3H), 0.43 (s, 4H), -0.10 (d, J = 6.5 Hz, 3H). HRMS m/z = 1081.6300 [M+H]+. 20 Compound 131 Note: axial chirality of compounds is as shown in below scheme. (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-12-(5-(4-cyclopropylpiperazin-1-yl)-2-((S)-1- methoxyethyl)pyridin-3-yl)-11-ethyl-25-hydroxy-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza- 1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3- 25 methyl-1-oxobutan-2-yl)-N-methyl-3-phenyltetrahydrofuran-2-carboxamide
PAT059646-WO-PCT 5 Procedure Step a: To a solution of Intermediate 33 (135 mg) and (1-ethoxycyclopropoxy)trimethylsilane (473 mg, 545 L) in MeOH (1.0 mL) at 0°C was added acetic acid (40.7 mg, 38.8 L) and sodium 10 cyanoborohydride (42.6 mg). The reaction was stirred at 60 °C for 1 hr, after which time sat. aq. NaHCO3 solution and DCM were added. The aq. layer was extracted with DCM (2x) and the organic extracts were passed through a phase separator and concentrated. The residue was purified by column chromatography (SiO2; 0 to 100% 3:1 EtOAc/EtOH in heptane) to afford tert- butyl ((61R,65S,66S,4S)-12-(5-(4-cyclopropylpiperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-15 11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola- 6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)carbamate (140 mg). LC/MS (Peptide Method) M/Z = 1032.6400 [M+H]+. Step b: To a solution of tert-butyl ((61R,65S,66S,4S)-12-(5-(4-cyclopropylpiperazin-1-yl)-2-((S)-1- methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8- PAT059646-WO-PCT 5 oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)carbamate (140 mg) in DCM (2 mL) was added TFA (1 mL). The reaction was stirred at RT for 20 min., after which time the reaction was concentrated. The remaining residue was dissolved in toluene and concentrated to afford (61R,65S,66S,4S)-4-amino-12-(5-(4-cyclopropylpiperazin-1-yl)- 2-((S)-1-methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-25-((triisopropylsilyl)oxy)-11H-8-oxa-10 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-5,7- dione trifluoroacetate salt, which was taken as-is to the next step. LC/MS (RXNMON-Acidic method): M/Z = 933.2 [M+H]+. Step c: To a solution of (61R,65S,66S,4S)-4-amino-12-(5-(4-cyclopropylpiperazin-1-yl)-2-((S)-1-15 methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64- diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-5,7-dione trifluoroacetate salt in DMF (1 mL) was added N-(tert-butoxycarbonyl)-N-methyl-L-valine (60.3 mg), DIPEA (134.7 mg, 182 L), and HATU (89.2 mg). The reaction was stirred at RT for 1 hr. The reaction was purified by column chromatography (SiO2; 0 to 15% MeOH in DCM) to afford20 tert-butyl ((2S)-1-(((61R,65S,66S,4S)-12-(5-(4-cyclopropylpiperazin-1-yl)-2-((S)-1- methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8- oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)amino)-3-methyl-1-oxobutan-2-yl)(methyl)carbamate, which contained some residual DMF as an impurity. This material was taken as-is to the next step. LC/MS (Peptide Method) M/Z = 25 1146.7200 [M+H]+. Step d: To a solution of tert-butyl ((2S)-1-(((61R,65S,66S,4S)-12-(5-(4-cyclopropylpiperazin-1-yl)- 2-((S)-1-methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)- 11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- 30 benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)(methyl)carbamate in DCM (2 mL) was added TFA (1 mL). The reaction was stirred at RT for 2.5 hr. The reaction was concentrated. The residue was purified by column chromatography (C18; 10 to 48% acetonitrile in water (+0.1% TFA)) to afford (2S)-N-((61R,65S,66S,4S)-12-(5-(4-cyclopropylpiperazin-1-yl)-2- ((S)-1-methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-35 11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)butanamide trifluoroacetate salt (24 mg). LC/MS (Peptide Method) M/Z = 1045.6600 [M+H]+. Step e: To a solution of (2S)-N-((61R,65S,66S,4S)-12-(5-(4-cyclopropylpiperazin-1-yl)-2-((S)-1-40 methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8- oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- PAT059646-WO-PCT 5 yl)-3-methyl-2-(methylamino)butanamide trifluoroacetate salt (24 mg) in DMF (207 L) was added (2R,3R)-3-phenyltetrahydrofuran-2-carboxylic acid (8 mg), DIPEA (21 mg, 29 L), and HATU (14.2 mg). The reaction was stirred for 2 hr at RT. The reaction was purified by column chromatography (SiO2; 0 to 15% MeOH in DCM) to afford (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)- 12-(5-(4-cyclopropylpiperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-10 5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)- N-methyl-3-phenyltetrahydrofuran-2-carboxamide (30 mg). LC/MS (Peptide Method) M/Z = 1220.7300 [M+H]+. 15 Step f: To a solution of (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-12-(5-(4-cyclopropylpiperazin-1-yl)- 2-((S)-1-methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)- 11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3- phenyltetrahydrofuran-2-carboxamide (30 mg) in DMF (300 L) was added TMAF (2.9 mg). The 20 reaction was stirred at RT for 1 hr. The reaction was purified by column chromatography (C18, 10 to 70% acetonitrile in water (+ 0.1% NH4OH)) to afford (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)- 12-(5-(4-cyclopropylpiperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-11-ethyl-25-hydroxy-10,10- dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3- 25 phenyltetrahydrofuran-2-carboxamide (8.5 mg).1H NMR (400 MHz, DMSO) 8.45 (t, J = 2.7 Hz, 1H), 7.86 (s, 1H), 7.76 – 7.32 (m, 2H), 7.30 – 7.02 (m, 6H), 6.94 (d, J = 2.1 Hz, 2H), 6.53 (s, 1H), 5.60 – 4.61 (m, 2H), 4.36 – 3.99 (m, 5H), 3.95 – 3.51 (m, 4H), 3.25 – 3.04 (m, 11H), 2.89 – 2.58 (m, 9H), 2.42 – 2.19 (m, 2H), 1.78 – 1.47 (m, 2H), 1.42 – 1.07 (m, 8H), 1.04 – 0.62 (m, 11H), 0.58 – 0.27 (m, 7H), 0.10 (d, J = 5.6 Hz, 1H), 0.01 – -0.12 (m, 3H). 30 LC/MS (Peptide Method) M/Z = 1063.5800 [M+H]+. Compound 132 Note: axial chirality of compounds is as shown in below scheme. (2R,3S)-N-((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(5-(4-(2-hydroxyethyl)piperazin-35 1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)- indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxopent-4-en-2-yl)-N-methyl-3-phenyloxetane-2-carboxamide PAT059646-WO-PCT 5 Procedure Step a: To a solution of Intermediate 38 (8 mg) in DCE (340 L) was added 2-((tert- butyldimethylsilyl)oxy)acetaldehyde (3.6 mg, 3.9 L). The mixture was stirred at RT for 10 min., after which time sodium triacetoxyhydroborate (5.8 mg) was added. The resulting mixture was 10 stirred at RT for 2 hr, after which time it was diluted with DCM, and quenched with 1:1 brine and sat. aq. NaHCO3. The aq. layer was extracted with DCM (3x), and the combined organic extracts were passed through a phase separator and concentrated. The residue was dissolved in DMF (380 μL) and treated with TMAF (5.1 mg). The mixture was stirred at RT for 2 hr, and was then directly purified by preparatory HPLC (C4; 10 to 80% acetonitrile in water (+ 0.1% NH4OH)) to15 afford (2R,3S)-N-((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(5-(4-(2- hydroxyethyl)piperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8- oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)amino)-3-methyl-1-oxopent-4-en-2-yl)-N-methyl-3-phenyloxetane-2-carboxamide (4.3 mg) as a white solid after lyophilization. 1H NMR (400 MHz, DMSO) 9.31 (s, 1H), 8.38 (d, J = 2.9 Hz, 20 1H), 7.80 (s, 1H), 7.65 (d, J = 8.4 Hz, 1H), 7.51 (d, J = 8.5 Hz, 1H), 7.45 – 7.36 (m, 3H), 7.25 (t, J = 7.5 Hz, 2H), 7.21 – 7.13 (m, 2H), 7.03 (s, 1H), 6.88 (s, 1H), 6.39 (s, 1H), 5.90 (d, J = 7.7 Hz, 1H), 5.44 (m, 1H), 5.27 (m, 1H), 4.94 (t, J = 6.2 Hz, 1H), 4.82 – 4.60 (m, 3H), 4.48 – 4.31 (m, 4H), 4.21 – 3.95 (m, 4H), 3.82 (d, J = 11.6 Hz, 1H), 3.65 (d, J = 10.9 Hz, 1H), 3.57 – 3.42 (m, 3H), 3.19 (m, 4H), 3.14 – 3.00 (m, 5H), 2.73 – 2.56 (m, 3H), 2.50 (m, 4H), 2.36 (t, J = 6.2 Hz, 2H), 2.31 (s, 25 2H), 2.09 (m, 1H), 1.29 (d, J = 6.2 Hz, 3H), 1.20 – 1.06 (m, 3H), 0.91 (t, J = 7.1 Hz, 3H), 0.73 (s, 4H), 0.40 (s, 3H), 0.06 (s, 1H), -0.16 (d, J = 6.9 Hz, 3H). LC/MS ( HRMS_2min) M/Z=1065.5814 [M+H]+. The following compounds of Table 11 were synthesized using the above procedure or 30 modifications of the above procedure employing the corresponding macrocycle and aldehyde/ketone. Compounds labeled with an * denote a modified reaction condition employing the corresponding enantiopure alkyl bromide, potassium carbonate, potassium iodide in acetonitrile at 80°C. Note: axial chirality of compounds is as shown in structures in Table 11. PAT059646-WO-PCT 5 Table 11: PanRAS inhibitor compounds PAT059646-WO-PCT 219 PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT 222 PAT059646-WO-PCT PAT059646-WO-PCT 5 Compound 141 Note: axial chirality of compounds is as shown in below scheme. (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(2-((S)-1-methoxyethyl)-5- (piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-10 bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)- N-methyl-3-phenyltetrahydrofuran-2-carboxamide PAT059646-WO-PCT 5 Procedure Step a: To a solution of Intermediate 29 (30 mg) in THF (1 mL) was added 1N TBAF in THF (27 mg, 103 μL). The reaction mixture was stirred at RT for 1 hr, after which time it was diluted with 10 EtOAc and washed with water and brine. The EtOAc extract was dried over Na2SO4, filtered, and concentrated. The residue was purified by column chromatography (C18; 10 to 100% acetonitrile in water (+ 0.1% formic acid)) to afford (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy- 12-(2-((S)-1-methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa- 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- 15 yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3-phenyltetrahydrofuran-2-carboxamide (9 mg) as a white solid after lyophilization.1H NMR (400 MHz, DMSO): 9.15 (s, 1H), 8.49 (d, J = 2.9 Hz, 1H), 7.87 (s, 1H), 7.72 (d, J = 8.3 Hz, 1H), 7.58 (dd, J = 8.8, 3.7 Hz, 1H), 7.48 (d, J = 8.6 Hz, 1H), 7.37 (dd, J = 8.9, 3.1 Hz, 1H), 7.34 – 7.18 (m, 4H), 7.14 (t, J = 7.2 Hz, 1H), 7.08 (s, 1H), 6.94 (s, 1H), 6.86 (dd, J = 27.4, 6.5 Hz, 1H), 6.53 (d, J = 5.1 Hz, 1H), 5.30 (q, J = 7.4 Hz, 1H), 5.08 (d, J 20 = 7.8 Hz, 1H), 4.80 (d, J = 11.7 Hz, 1H), 4.31 – 4.12 (m, 5H), 3.88 – 3.71 (m, 3H), 3.58 (d, J = 11.0 Hz, 1H), 3.45 (t, J = 5.3 Hz, 4H), 3.20 (d, J = 5.2 Hz, 4H), 3.10 (d, J = 17.1 Hz, 4H), 2.79 (d, J = 18.1 Hz, 2H), 2.66 (s, 4H), 2.41 – 2.19 (m, 3H), 1.76 – 1.62 (m, 1H), 1.37 (t, J = 6.8 Hz, 3H), 1.26 – 1.14 (m, 2H), 0.97 (p, J = 6.6 Hz, 4H), 0.80 (d, J = 9.8 Hz, 4H), 0.72 (t, J = 6.3 Hz, 1H), 0.66 (d, J = 6.5 Hz, 3H), 0.53 (s, 1H), 0.46 (s, 2H), 0.14 – 0.04 (m, 1H), -0.07 (d, J = 6.6 Hz, 3H). 25 LC/MS (Peptide method) M/Z = 1023.5700 [M+H]+. Compound 142 Note: axial chirality of compounds is as shown in below scheme. (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(5-(4-(2-hydroxyacetyl)piperazin-1-30 yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)- indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxobutan-2-yl)-N-methyl-3-phenyltetrahydrofuran-2-carboxamide PAT059646-WO-PCT 5 Procedure Step a: To a solution of (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1- methoxyethyl)-5-(piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)- 11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-10 benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3- phenyltetrahydrofuran-2-carboxamide (42 mg), 2-hydroxyacetic acid (13 mg) and TSTU (51 mg) in DMF (500 μL), was added DIPEA (44 mg, 59 μL). The reaction mixture was stirred at RT for 30 min., after which time it was concentrated. The remaining residue was purified by column chromatography (C18; 10 to 100% acetonitrile in water + (0.1% formic acid)) to afford (2R,3R)-N-15 ((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(5-(4-(2-hydroxyacetyl)piperazin-1-yl)-2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64- diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)- 3-methyl-1-oxobutan-2-yl)-N-methyl-3-phenyltetrahydrofuran-2-carboxamide (23 mg). LC/MS (Peptide Method): M/Z = 1237.7100 [M+H]+. 20 Step b: To a solution of (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-12-(5-(4-(2- hydroxyacetyl)piperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3- 25 phenyltetrahydrofuran-2-carboxamide (23 mg) in THF (1 mL) was added TBAF (11.9 mg). The reaction mixture was stirred at RT for 1 hr, after which time it was diluted with EtOAc and washed with water and brine. The EtOAc extract was dried over Na2SO4, filtered, and concentrated. The residue was purified by column chromatography (C18; 10 to 100% acetonitrile in water + 0.1% formic acid) to afford (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(5-(4-(2-30 hydroxyacetyl)piperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H- 8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane- 4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3-phenyltetrahydrofuran-2-carboxamide (11 mg) as a white solid.1H NMR (400 MHz, DMSO) 9.20 (s, 1H), 8.53 (d, J = 2.9 Hz, 1H), 7.93 (s, 1H), 7.78 (d, J = 8.3 Hz, 1H), 7.63 (dd, J = 8.7, 3.1 Hz, 1H), 7.53 (d, J = 8.5 Hz, 1H), 7.43 – 7.31 PAT059646-WO-PCT 5 (m, 3H), 7.24 (dt, J = 29.5, 7.3 Hz, 3H), 7.14 (s, 1H), 6.97 (d, J = 18.5 Hz, 1H), 6.89 (d, J = 7.5 Hz, 1H), 6.59 (s, 1H), 5.36 (m, 1H), 5.14 (d, J = 7.7 Hz, 1H), 4.84 (d, J = 11.6 Hz, 1H), 4.70 (t, J = 5.5 Hz, 1H), 4.37 – 4.09 (m, 7H), 4.03 – 3.74 (m, 4H), 3.71 – 3.54 (m, 5H), 3.26 – 3.19 (m, 1H), 3.17 (s, 2H), 3.13 (s, 1H), 2.89 (d, J = 16.4 Hz, 1H), 2.83 (s, 1H), 2.72 (s, 3H), 2.70 (s, 1H), 2.42 (m, 1H), 2.32 (m, 1H), 1.85 – 1.68 (m, 1H), 1.42 (d, J = 6.2 Hz, 3H), 1.22 (t, J = 7.1 Hz, 2H), 1.03 10 (m, 3H), 0.89 – 0.75 (m, 4H), 0.72 (d, J = 6.4 Hz, 2H), 0.59 (s, 1H), 0.51 (s, 2H), 0.20 – 0.10 (m, 1H), -0.01 (d, J = 6.6 Hz, 2H). HRMS m/z = 1081.5800 [M+H]+. The following compounds of Table 12 were synthesized using the above procedure or modifications of the above procedure using the corresponding macrocycle, amino acid, carboxylic 15 acid, or carboxylic acid equivalent. Note: axial chirality of compounds is as shown in structures in Table 12. Table 12: PanRAS inhibitor compounds 227 PAT059646-WO-PCT PAT059646-WO-PCT 229 PAT059646-WO-PCT PAT059646-WO-PCT 5 Compound 149 Note: axial chirality of compounds is as shown in below scheme. methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-25-hydroxy-4-((2S,3R)-2-((3S,4S)-1-(methoxycarbonyl)- N-methyl-4-phenylpyrrolidine-3-carboxamido)-3-methylpent-4-enamido)-10,10-dimethyl-5,7-10 dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate PAT059646-WO-PCT 5 Procedure Step a: To a solution of Intermediate 34 (22 mg), Intermediate 14 (6.5 mg), and DIPEA (13.3 mg, 18 L) in 1,4-dioxane (100 L) was added PyBroP (12 mg). The reaction stirred for 18 hr, after which time additional Intermediate 14 (5.0 mg), DIPEA (8.1mg, 11 L), and PyBroP (12 mg) were 10 added. The reaction was stirred at 30 °C for 2 hr. The reaction stirred for a total of 20 hr and was then concentrated, azeotropically dried with DCM and heptane (2x), and concentrated again to afford crude methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-4-((2S,3R)-2-((3S,4S)-1-(methoxycarbonyl)- N-methyl-4-phenylpyrrolidine-3-carboxamido)-3-methylpent-4-enamido)-10,10-dimethyl-5,7- dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-15 2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1- carboxylate. LC/MS (Peptide Method) M/Z = 1307.7200 [M+H]+. Step b: To a solution of crude methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-4-((2S,3R)-2-((3S,4S)-1- (methoxycarbonyl)-N-methyl-4-phenylpyrrolidine-3-carboxamido)-3-methylpent-4-enamido)-20 10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3- yl)piperazine-1-carboxylate in DMF (300 L) was added NH4OH (4.8 mg, 5.3 L, 30% wt.). The solution was stirred at RT for 30 min. after which time TMAF (8 mg) was added, and the mixture was briefly sonicated and stirred at RT for 45 min. The reaction was diluted in DMSO, filtered, 25 and purified by preparatory HPLC (C18; 45 to 70% acetonitrile in water + 0.1% NH4OH). Desired fractions were concentrated, and the remaining residue was dissolved in DCM and washed with water. The aq. layer was extracted with DCM (3x), and combined organic extracts were passed PAT059646-WO-PCT 5 through a phase separator and concentrated. The resulting residue was dissolved in 1:1 acetonitrile:water and lyophilized to afford methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-25-hydroxy-4- ((2S,3R)-2-((3S,4S)-1-(methoxycarbonyl)-N-methyl-4-phenylpyrrolidine-3-carboxamido)-3- methylpent-4-enamido)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3- 10 yl)piperazine-1-carboxylate (6.6 mg) as a white solid. LC/MS (Peptide Method) M/Z = 1150.5699 [M+H]+.1H NMR (400 MHz, DMSO) 9.40 – 9.15 (m, 1H), 8.50 – 8.45 (m, 1H), 7.93 – 7.79 (m, 1H), 7.75 – 7.67 (m, 1H), 7.63 – 7.54 (m, 1H), 7.52 – 7.43 (m, 1H), 7.33 – 7.16 (m, 5H), 7.11 (s, 1H), 7.01 – 6.95 (m, 1H), 6.78 – 6.43 (m, 1H), 6.39 (s, 1H), 5.92 – 5.51 (m, 1H), 5.44 – 5.26 (m, 1H), 5.14 – 4.87 (m, 1H), 4.86 – 4.82 (m, 1H), 4.83 – 4.78 (m, 1H), 4.59 – 4.30 (m, 1H), 4.30 – 15 4.17 (m, 1H), 4.18 – 4.11 (m, 2H), 4.11 – 4.03 (m, 1H), 3.99 – 3.87 (m, 2H), 3.86 – 3.77 (m, 2H), 3.77 – 3.69 (m, 2H), 3.67 – 3.64 (m, 3H), 3.62 (s, 4H), 3.56 – 3.48 (m, 4H), 3.31 – 3.24 (m, 4H), 3.24 – 3.17 (m, 1H), 3.16 – 3.04 (m, 3H), 2.87 – 2.71 (m, 2H), 2.71 – 2.58 (m, 5H), 2.44 – 2.34 (m, 1H), 1.41 – 1.33 (m, 3H), 1.28 – 1.22 (m, 2H), 1.22 – 1.14 (m, 2H), 1.04 – 0.92 (m, 3H), 0.87 – 0.75 (m, 5H), 0.57 – 0.41 (m, 3H), 0.29 – 0.12 (m, 3H). 20 The following compounds of Table 13 were synthesized using the above procedure or modifications of the above procedure using the corresponding macrocycle, amino acid, carboxylic acid, or carboxylic acid equivalent. Note: axial chirality of compounds is as shown in structures in Table 13. For Table 13, * indicates that HATU/NMP was used for acylation of the corresponding 25 N-methylated amino acid. PAT059646-WO-PCT 5 Compound 151 Note: axial chirality of compounds is as shown in below scheme. methyl 4-(5-((61R,65S,66S,4S)-4-((2S,3R)-2-((3S,4S)-1-(ethoxycarbonyl)-N-methyl-4- phenylpyrrolidine-3-carboxamido)-3-methylpent-4-enamido)-11-ethyl-25-hydroxy-10,10-dimethyl-10 5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate PAT059646-WO-PCT 5 Step a: To a solution of Intermediate 35 (20 mg) and DIPEA (21 mg, 28 L) in DCM (150 μL) was added ethyl carbonochloridate (8.8 mg, 7.7 L). The reaction stirred at RT for 30 min, after which time it was concentrated to afford crude methyl 4-(5-((61R,65S,66S,4S)-4-((2S,3R)-2-((3S,4S)-1- (ethoxycarbonyl)-N-methyl-4-phenylpyrrolidine-3-carboxamido)-3-methylpent-4-enamido)-11- ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-10 6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1- methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate, which was carried forward as-is to the next step. LC/MS (Peptide method) M/Z = 1321.7400 [M+H]+. Step b: Crude methyl 4-(5-((61R,65S,66S,4S)-4-((2S,3R)-2-((3S,4S)-1-(ethoxycarbonyl)-N-15 methyl-4-phenylpyrrolidine-3-carboxamido)-3-methylpent-4-enamido)-11-ethyl-10,10-dimethyl- 5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3- yl)piperazine-1-carboxylate was dissolved in DMF (200 μL) and aq. NH4OH (11 L, 30% wt.) was added. The mixture stirred at RT for 30 min. then TMAF (7.0 mg) was added. The reaction mixture 20 was sonicated several times and stirred at RT for 30 min. The reaction mixture was filtered, diluted with DMSO, and purified by preparatory HPLC (50 to 70% acetonitrile in water (+0.1% NH4OH)), and concentrated. The purified product was dissolved in DCM and diluted with water. The aqueous layer was extracted with DCM (3x), and the combined organic layers were filtered through a phase separator, concentrated and lyophilized to afford methyl 4-(5-((61R,65S,66S,4S)-25 4-((2S,3R)-2-((3S,4S)-1-(ethoxycarbonyl)-N-methyl-4-phenylpyrrolidine-3-carboxamido)-3- methylpent-4-enamido)-11-ethyl-25-hydroxy-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza- 1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1- methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (10.8 mg) as a white solid.1H NMR (400 MHz, DMSO) 9.39 – 9.16 (m, 1H), 8.52 – 8.43 (m, 1H), 7.93 – 7.80 (m, 1H), 7.74 – 7.64 (m, 1H), 7.58 30 (t, J = 9.6 Hz, 1H), 7.51 – 7.44 (m, 1H), 7.32 – 7.18 (m, 5H), 7.11 (s, 1H), 7.00 – 6.95 (m, 1H), 6.84 – 6.62 (m, 1H), 6.39 (s, 1H), 5.92 – 5.51 (m, 1H), 5.42 – 5.28 (m, 1H), 5.14 – 4.79 (m, 3H), 4.54 (d, J = 10.0 Hz, 1H), 4.30 – 4.18 (m, 1H), 4.18 – 4.02 (m, 5H), 3.92 (dd, J = 10.7, 4.9 Hz, 2H), 3.82 – 3.70 (m, 4H), 3.62 (s, 4H), 3.56 – 3.48 (m, 5H), 3.31 – 3.24 (m, 4H), 3.23 – 3.16 (m, 1H), 3.12 (s, 2H), 3.07 (s, 1H), 2.88 – 2.77 (m, 1H), 2.76 – 2.72 (m, 1H), 2.71 – 2.61 (m, 5H), 2.40 35 (m, 1H), 1.40 – 1.34 (m, 3H), 1.25 – 1.21 (m, 4H), 1.21 – 1.19 (m, 1H), 1.19 – 1.16 (m, 1H), 1.01 – 0.94 (m, 3H), 0.87 – 0.76 (m, 5H), 0.56 – 0.43 (m, 3H), 0.30 – 0.12 (m, 3H). LC/MS (Peptide method) M/Z = 1164.6100 [M+H]+. The following compounds of Table 14 were synthesized using the above procedure or 40 modifications of the above. Note: axial chirality of compounds is as shown in structures in Table 14. PAT059646-WO-PCT 5 Table 14: PanRAS inhibitor compounds Compound 153 Note: axial chirality of compounds is as shown in below scheme. 10 methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-25-hydroxy-4-((2S,3R)-2-((3S,4S)-1-(methoxycarbonyl)- N-methyl-4-(4-(4-methylpiperazin-1-yl)phenyl)pyrrolidine-3-carboxamido)-3-methylpent-4- enamido)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3- yl)piperazine-1-carboxylate PAT059646-WO-PCT 5 Procedure Step a: To a solution of Intermediate 37 (61 mg) in MeOH (500 μL) was added aq. formaldehyde (11.2 mg, 10.3 L, 37% wt). The reaction stirred for 30 min. before addition of sodium triacetoxyborohydrate (19.5 mg). The reaction stirred at RT for 30 min., after which time the 10 reaction mixture was diluted with DCM and quenched with sat. aq. NaHCO3. The aq. layer was extracted with DCM (3x), and the organic extracts were passed through a phase separator and concentrated to afford methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-4-((2S,3R)-2-((3S,4S)-1- (methoxycarbonyl)-N-methyl-4-(4-(4-methylpiperazin-1-yl)phenyl)pyrrolidine-3-carboxamido)-3- methylpent-4-enamido)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-15 diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)- 1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (56 mg) as a yellow solid. LC/MS (Peptide method) M/Z = 1405.8000 [M+H]+. Step b: To a solution of methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-4-((2S,3R)-2-((3S,4S)-1-20 (methoxycarbonyl)-N-methyl-4-(4-(4-methylpiperazin-1-yl)phenyl)pyrrolidine-3-carboxamido)-3- methylpent-4-enamido)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64- diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)- 1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (19 mg) in DMF (400 μL) was added TMAF (12.8 mg). The reaction mixture was sonicated then stirred at RT for 30 min. The reaction mixture 25 was filtered, diluted with DMSO, and purified by preparatory HPLC (50 to 70% acetonitrile in water (+0.1% NH4OH)), and concentrated. The material was dissolved in DCM and washed with water. PAT059646-WO-PCT 5 The aq. layer was extracted with DCM (3x), and the combined organic layers were filtered through a phase separator, and concentrated to afford methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-25-hydroxy- 4-((2S,3R)-2-((3S,4S)-1-(methoxycarbonyl)-N-methyl-4-(4-(4-methylpiperazin-1- yl)phenyl)pyrrolidine-3-carboxamido)-3-methylpent-4-enamido)-10,10-dimethyl-5,7-dioxo-11H-8- oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12- 10 yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (8.9 mg) as a white solid after lyophilization.1H NMR (400 MHz, DMSO) 9.41 – 9.18 (m, 1H), 8.52 – 8.43 (m, 1H), 7.94 – 7.82 (m, 1H), 7.71 – 7.62 (m, 1H), 7.62 – 7.52 (m, 1H), 7.52 – 7.44 (m, 1H), 7.36 – 7.25 (m, 1H), 7.14 – 7.04 (m, 2H), 6.98 (s, 1H), 6.86 – 6.70 (m, 2H), 6.64 – 6.44 (m, 1H), 6.37 (s, 1H), 5.93 – 5.51 (m, 1H), 5.41 – 5.25 (m, 1H), 4.95 – 4.78 (m, 2H), 4.56 (d, J = 9.9 Hz, 1H), 4.35 – 4.19 (m, 1H), 15 4.17 – 4.07 (m, 2H), 3.94 (d, J = 11.8 Hz, 1H), 3.90 – 3.83 (m, 1H), 3.76 – 3.67 (m, 4H), 3.65 – 3.61 (m, 6H), 3.55 – 3.50 (m, 4H), 3.29 – 3.25 (m, 4H), 3.12 (s, 2H), 3.09 – 3.02 (m, 4H), 2.91 – 2.76 (m, 2H), 2.71 – 2.65 (m, 1H), 2.65 – 2.60 (m, 4H), 2.46 – 2.38 (m, 4H), 2.28 (s, 1H), 2.20 (s, 3H), 2.17 – 2.11 (m, 1H), 1.41 – 1.34 (m, 3H), 1.23 (s, 3H), 1.22 – 1.15 (m, 3H), 1.04 (t, J = 7.0 Hz, 1H), 0.96 (t, J = 7.1 Hz, 3H), 0.88 – 0.79 (m, 5H), 0.74 (s, 1H), 0.56 – 0.43 (m, 3H), 0.32 – 20 0.14 (m, 3H). HRMS (Peptide_method) m/z [M+H]+: 1248.7000 [M+H]+. Compound 154 Note: axial chirality of compounds is as shown in below scheme. methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-25-hydroxy-4-((2S,3R)-2-((3S,4S)-4-(4-(4-(2-25 hydroxyacetyl)piperazin-1-yl)phenyl)-1-(methoxycarbonyl)-N-methylpyrrolidine-3-carboxamido)- 3-methylpent-4-enamido)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3- yl)piperazine-1-carboxylate 30 Procedure Step a: To a solution of Intermediate 34 (18 mg), Intermediate 18 (8.3 mg), and DIPEA (5.5 mg, 7.4 L) in 1,4-dioxane (200 L) was added PyBroP (11 mg). The reaction stirred for 20 hr at RT, after which time additional PyBroP (2.4 mg) and DIPEA (1.1 mg, 1.5 L) were added. The reaction 35 was then stirred at 40°C for 24 hr, representing a total reaction time of 44 hr. The mixture was concentrated and dried under reduced pressure to afford crude methyl 4-(5-((61R,65S,66S,4S)-11- PAT059646-WO-PCT 5 ethyl-4-((2S,3R)-2-((3S,4S)-4-(4-(4-(2-hydroxyacetyl)piperazin-1-yl)phenyl)-1- (methoxycarbonyl)-N-methylpyrrolidine-3-carboxamido)-3-methylpent-4-enamido)-10,10- dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3- yl)piperazine-1-carboxylate as a yellow foam, which was taken as-is to the next step. 10 Step b: To a solution of crude methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-4-((2S,3R)-2-((3S,4S)-4-(4- (4-(2-hydroxyacetyl)piperazin-1-yl)phenyl)-1-(methoxycarbonyl)-N-methylpyrrolidine-3- carboxamido)-3-methylpent-4-enamido)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H- 8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane- 15 12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate in DMF (200 L) was added aq. NH4OH (4 mg, 4.5 L, 30% wt.). The reaction stirred for 30 min., after which time TMAF (6 mg) was added. The reaction was sonicated then stirred at RT for 45 min. The reaction mixture was filtered, diluted with DMSO, and purified by preparatory HPLC (C18, 30 to 50% acetonitrile in water (+ 0.1% TFA)), and concentrated. The solid material was dissolved in DCM and washed 20 with sat. aq. NaHCO3. The aq. layer was extracted with DCM (3x), and the combined organic extracts were passed through a phase separator and concentrated to afford methyl 4-(5- ((61R,65S,66S,4S)-11-ethyl-25-hydroxy-4-((2S,3R)-2-((3S,4S)-4-(4-(4-(2-hydroxyacetyl)piperazin- 1-yl)phenyl)-1-(methoxycarbonyl)-N-methylpyrrolidine-3-carboxamido)-3-methylpent-4- enamido)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-25 bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3- yl)piperazine-1-carboxylate (5.3 mg) as a white solid after lyophilization. 1H NMR (400 MHz, DMSO) 9.38 (s, 1H), 8.51 – 8.38 (m, 1H), 7.93 – 7.81 (m, 1H), 7.70 – 7.62 (m, 1H), 7.62 – 7.51 (m, 1H), 7.47 (d, J = 8.5 Hz, 1H), 7.34 – 7.24 (m, 1H), 7.09 (d, J = 8.6 Hz, 2H), 6.98 (s, 1H), 6.85 (d, J = 8.5 Hz, 1H), 6.73 – 6.47 (m, 1H), 6.37 (s, 1H), 5.95 – 5.48 (m, 1H), 5.41 – 5.26 (m, 1H), 30 5.13 – 4.77 (m, 2H), 4.62 – 4.52 (m, 1H), 4.30 – 4.18 (m, 1H), 4.17 – 4.03 (m, 4H), 3.98 – 3.85 (m, 2H), 3.76 – 3.67 (m, 3H), 3.65 – 3.61 (m, 5H), 3.60 – 3.56 (m, 2H), 3.56 – 3.49 (m, 4H), 3.46 (s, 1H), 3.29 – 3.24 (m, 4H), 3.24 – 3.17 (m, 1H), 3.12 (s, 2H), 3.10 – 3.01 (m, 4H), 2.97 – 2.75 (m, 2H), 2.70 – 2.65 (m, 1H), 2.65 – 2.60 (m, 3H), 2.41 – 2.35 (m, 1H), 2.04 – 1.89 (m, 1H), 1.40 – 1.34 (m, 3H), 1.28 – 1.21 (m, 6H), 1.20 – 1.13 (m, 4H), 1.05 – 0.93 (m, 3H), 0.88 – 0.72 (m, 35 8H), 0.45 (s, 3H), 0.32 – 0.11 (m, 3H). LC/MS (Peptide method) M/Z = 1292.6801 [M+H]+. Compound 155 Note: axial chirality of compounds is as shown in below scheme. methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-25-hydroxy-10,10-dimethyl-4-((2S,3R)-3-methyl-2-40 ((2R,3R)-N-methyl-3-(4-(4-methylpiperazin-1-yl)phenyl)tetrahydrofuran-2-carboxamido)pent-4- PAT059646-WO-PCT 5 enamido)-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate Procedure 10 Step a: To a solution of Intermediate 34 (25 mg), Intermediate 19 (8.8 mg), and DIPEA (8.9 mg, 12 L) in 1,4-dioxane (100 μL) was added PyBroP (16.3 mg). The reaction stirred at RT for 17 hr and additional Intermediate 19 (1.4 mg), DIPEA (1.8 mg, 2.4 L), and PyBroP (3.3 mg) were added, and the reaction was continued. The reaction stirred for a total of 20 hr, after which time the mixture was concentrated to afford crude methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-10,10-15 dimethyl-4-((2S,3R)-3-methyl-2-((2R,3R)-N-methyl-3-(4-(4-methylpiperazin-1- yl)phenyl)tetrahydrofuran-2-carboxamido)pent-4-enamido)-5,7-dioxo-25-((triisopropylsilyl)oxy)- 11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate, which was taken as-is to the next step. LC/MS (Peptide method) M/Z = 1347.7700 [M+H]+. 20 Step b: Crude methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-10,10-dimethyl-4-((2S,3R)-3-methyl-2- ((2R,3R)-N-methyl-3-(4-(4-methylpiperazin-1-yl)phenyl)tetrahydrofuran-2-carboxamido)pent-4- enamido)-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3- 25 yl)piperazine-1-carboxylate was dissolved in DMF (300 μL) and aq. NH4OH (6.2 L, 30% wt) was added. The reaction stirred at RT for 30 min., after which time TMAF (10 mg) was added. The reaction mixture was briefly sonicated and then was left stirring at RT for 1 hr. The reaction was PAT059646-WO-PCT 5 diluted in DMSO, filtered, and purified by preparatory HPLC (C18; 45 to 70% acetonitrile in water (+ 0.1% NH4OH)). The product was dissolved in DCM and diluted with water. The aq. layer was extracted with DCM (3x), and the combined organic extracts were passed through a phase separator, and concentrated to afford methyl 4-(5-((61R,65S,66S,4S)-11-ethyl-25-hydroxy-10,10- dimethyl-4-((2S,3R)-3-methyl-2-((2R,3R)-N-methyl-3-(4-(4-methylpiperazin-1-10 yl)phenyl)tetrahydrofuran-2-carboxamido)pent-4-enamido)-5,7-dioxo-11H-8-oxa-63,64-diaza- 1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1- methoxyethyl)pyridin-3-yl)piperazine-1-carboxylate (8.8 mg) as a white solid after lyophilization. 1H NMR (400 MHz, DMSO) 9.29 – 9.20 (m, 1H), 8.50 – 8.45 (m, 1H), 7.90 – 7.83 (m, 1H), 7.61 – 7.54 (m, 1H), 7.52 – 7.44 (m, 1H), 7.32 – 7.26 (m, 1H), 7.13 (d, J = 8.6 Hz, 2H), 7.08 (s, 1H), 15 6.96 – 6.92 (m, 1H), 6.84 – 6.77 (m, 2H), 6.48 – 6.43 (m, 1H), 5.58 – 5.46 (m, 1H), 5.35 (q, J = 6.6 Hz, 1H), 5.08 (d, J = 8.0 Hz, 1H), 4.88 – 4.73 (m, 2H), 4.49 (d, J = 9.8 Hz, 1H), 4.31 – 4.18 (m, 2H), 4.18 – 4.02 (m, 3H), 3.92 – 3.80 (m, 2H), 3.77 – 3.67 (m, 2H), 3.65 – 3.61 (m, 3H), 3.56 – 3.49 (m, 4H), 3.29 – 3.24 (m, 4H), 3.11 (s, 2H), 3.09 – 3.00 (m, 4H), 2.89 – 2.72 (m, 2H), 2.69 – 2.61 (m, 3H), 2.59 (s, 2H), 2.45 (s, 3H), 2.32 – 2.18 (m, 6H), 1.37 (m, J = 6.4 Hz, 3H), 1.27 – 20 1.21 (m, 3H), 1.17 (s, 3H), 1.01 – 0.94 (m, 3H), 0.88 – 0.72 (m, 7H), 0.45 (s, 3H), 0.18 – 0.09 (m, 3H). LC/MS (Peptide method) M/Z = 1191.6200 [M+H]+. Compound 156 Note: axial chirality of compounds is as shown in below scheme. 25 (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-12-(5-((R)-3,4-dimethylpiperazin-1-yl)-2-((S)-1- methoxyethyl)pyridin-3-yl)-11-ethyl-25-hydroxy-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza- 1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3- methyl-1-oxobutan-2-yl)-N-methyl-3-phenyltetrahydrofuran-2-carboxamide
PAT059646-WO-PCT 5 Procedure Step a: To a solution of Intermediate 9a (300 mg) and Intermediate 12 (285 mg) in 1,4-dioxane (2.6 mL) and water (352 L) was added Cs2CO3 (281 mg) and Pd(dppf)Cl2- DCM adduct (16.9 10 mg). The reaction was stirred at 80°C under nitrogen for 16 hr. The reaction was then concentrated. The resulting residue was dissolved in EtOAc and washed with water. The EtOAc extract was dried with Na2SO4, filtered, and concentrated. The remaining residue was purified by column chromatography (SiO2; 0 to 37% 3:1 EtOAc/EtOH in heptane) to afford benzyl (2R)-4-(5- ((61R,65S,66S,4S)-4-((tert-butoxycarbonyl)amino)-10,10-dimethyl-5,7-dioxo-25-15 ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3-yl)-2-methylpiperazine-1- carboxylate (301.3 mg). LC/MS (RXNMON-Acidic method): M/Z = 1112.6 [M+H]+. Step b: To a solution of benzyl (2R)-4-(5-((61R,65S,66S,4S)-4-((tert-butoxycarbonyl)amino)-10,10-20 dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- 242 PAT059646-WO-PCT 5 bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1-methoxyethyl)pyridin-3- yl)-2-methylpiperazine-1-carboxylate (301 mg) in DMF (2.5 mL) was added DIPEA (140 mg, 189 L), Cs2CO3 (353 mg), and EtI (106 mg, 54.4 L). The reaction was stirred at RT for 4.5 hr after which time EtOAc and water were added. The aq. layer was extracted with EtOAc (2x). The combined EtOAc extracts were washed with brine, dried with Na2SO4, filtered, and 10 concentrated. The residue was purified by column chromatography (SiO2; 0 to 40% 3:1 EtOAc/EtOH in heptane) to afford benzyl (2R)-4-(5-((61R,65S,66S,4S)-4-((tert- butoxycarbonyl)amino)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa- 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)- 6-((S)-1-methoxyethyl)pyridin-3-yl)-2-methylpiperazine-1-carboxylate (217 mg). LC/MS 15 (RXNMON-Acidic method): M/Z = 1140.7 [M+H]+. Step c: To a solution of benzyl (2R)-4-(5-((61R,65S,66S,4S)-4-((tert-butoxycarbonyl)amino)-11- ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola- 6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-12-yl)-6-((S)-1- 20 methoxyethyl)pyridin-3-yl)-2-methylpiperazine-1-carboxylate (217 mg) in MeOH (3 mL) was added Pd(OH)2 on carbon (27 mg, 20% wt.). The reaction vessel was evacuated and backfilled with H2. The reaction was stirred under an atmosphere of H2 at RT for 16 hr. The reaction mixture was filtered and concentrated to afford tert-butyl ((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1- methoxyethyl)-5-((R)-3-methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-25 ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)carbamate (182 mg). LC/MS (Peptide Method) M/Z = 1006.6100 [M+H]+. Step d: To a solution of tert-butyl ((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-((R)-3-30 methylpiperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa- 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)carbamate (182 mg) in MeOH (2.7 mL) was added aq. formaldehyde (44 mg, 40.4 L, 37% wt.) and sodium triacetoxyborohydride (115 mg). The reaction was stirred at RT for 1 hr. The reaction was diluted with DCM and sat. NaHCO3 solution, passed through a phase separator, and 35 concentrated. The residue was purified by column chromatography (C18; 10 to 60% acetonitrile in water + 0.1% TFA) to afford tert-butyl ((61R,65S,66S,4S)-12-(5-((R)-3,4-dimethylpiperazin-1-yl)- 2-((S)-1-methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)- 11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)carbamate (35 mg) as the second eluting isomer. LC/MS 40 (Peptide Method) M/Z = 1020.6300 [M+H]+. PAT059646-WO-PCT 5 Step e: To a solution of tert-butyl ((61R,65S,66S,4S)-12-(5-((R)-3,4-dimethylpiperazin-1-yl)-2-((S)- 1-methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8- oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)carbamate (35 mg) in DCM (2 mL) was added TFA (1 mL). The reaction was stirred at RT for 30 min. The reaction was concentrated. The remaining residue was dissolved in toluene and10 concentrated to afford (61R,65S,66S,4S)-4-amino-12-(5-((R)-3,4-dimethylpiperazin-1-yl)-2-((S)-1- methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64- diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-5,7-dione trifluoroacetate salt. This material was carried forward directly into the next reaction. LC/MS (RXNMON-Acidic method): M/Z = 920.9 [M+H]+. 15 Step f: To a solution of (61R,65S,66S,4S)-4-amino-12-(5-((R)-3,4-dimethylpiperazin-1-yl)-2-((S)-1- methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64- diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-5,7-dione trifluoroacetate salt in DMF (343 L) was added N-(tert-butoxycarbonyl)-N-methyl-L-valine (14.3 20 mg), DIPEA (31.9 mg, 43.0 L), and HATU (21.1 mg). The reaction was stirred at RT for 45 min. The reaction was purified by column chromatography (SiO2; 0 to 15% MeOH in DCM) to afford tert-butyl ((2S)-1-(((61R,65S,66S,4S)-12-(5-((R)-3,4-dimethylpiperazin-1-yl)-2-((S)-1- methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8- oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- 25 yl)amino)-3-methyl-1-oxobutan-2-yl)(methyl)carbamate (38.8 mg). LC/MS (Peptide Method) M/Z = 1133.7100 [M+H]+. Step g: To a solution of tert-butyl ((2S)-1-(((61R,65S,66S,4S)-12-(5-((R)-3,4-dimethylpiperazin-1- yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-30 ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)(methyl)carbamate (38.8 mg) in DCM (2 mL) was added TFA (1 mL). The reaction was stirred at RT for 30 min. The reaction was concentrated. The remaining residue was dissolved in toluene and concentrated to afford (2S)-N-((61R,65S,66S,4S)-12-(5-((R)-3,4-dimethylpiperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-35 yl)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)- indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)-3-methyl-2- (methylamino)butanamide trifluoroacetate salt. This material was carried forward without further purification. LC/MS (RXNMON-Acidic method): M/Z = 1033.9 [M+H]+. 40 Step h: To a solution of (2S)-N-((61R,65S,66S,4S)-12-(5-((R)-3,4-dimethylpiperazin-1-yl)-2-((S)-1- methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8- PAT059646-WO-PCT 5 oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)-3-methyl-2-(methylamino)butanamide trifluoroacetate salt in DMF (376 L) was added (2R,3R)-3-phenyltetrahydrofuran-2-carboxylic acid (13.2 mg), DIPEA (35.4 mg, 47.7 L), and HATU (23.4 mg). The reaction was stirred at RT for 16 hr. The reaction was purified by column chromatography (SiO2; 0 to 15% MeOH in DCM) to afford (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-10 12-(5-((R)-3,4-dimethylpiperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-11-ethyl-10,10- dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)- N-methyl-3-phenyltetrahydrofuran-2-carboxamide (51.9 mg). LC/MS (Peptide Method) M/Z = 1028.7200. 15 Step i: To a solution of (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-12-(5-((R)-3,4-dimethylpiperazin-1- yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-11-ethyl-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3- 20 phenyltetrahydrofuran-2-carboxamide (51.9 mg) in THF (1 mL) was added TBAF (1M in THF, 51.3 L). The reaction was stirred at RT for 16 hr. Water (3 mL) was added to the reaction and the mixture was extracted with EtOAc (4x). The combined EtOAc extracts were concentrated under reduced pressure. The residue was purified by column chromatography (C18; 10 to 65% acetonitrile in water + 0.1% NH4OH) to afford (2R,3R)-N-((2S)-1-(((61R,65S,66S,4S)-12-(5-((R)-25 3,4-dimethylpiperazin-1-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-11-ethyl-25-hydroxy-10,10- dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxobutan-2-yl)-N-methyl-3- phenyltetrahydrofuran-2-carboxamide (6.3 mg). LC/MS (Peptide Method) M/Z = 1051.5900 [M+H]+.1H NMR (400 MHz, DMSO) 9.14 (s, 1H), 8.45 (d, J = 2.8 Hz, 1H), 7.87 (d, J 30 = 1.7 Hz, 1H), 7.78 – 7.41 (m, 3H), 7.31 – 7.02 (m, 6H), 6.97 – 6.47 (m, 3H), 5.49 – 5.04 (m, 2H), 4.79 (d, J = 11.7 Hz, 1H), 4.35 – 4.02 (m, 6H), 3.94 – 3.50 (m, 7H), 3.09 (d, J = 15.9 Hz, 4H), 2.93 – 2.58 (m, 9H), 2.38 – 2.08 (m, 7H), 1.37 (dd, J = 7.4, 6.2 Hz, 3H), 1.29 – 0.89 (m, 10H), 0.88 – 0.39 (m, 11H), 0.03 – -0.22 (m, 3H). 35 Compound 157 Note: axial chirality of compounds is as shown in below scheme. (2R,3S)-N-((3R)-1-(((61R,65S,66S,4S)-11-ethyl-12-(5-((S)-hexahydropyrazino[2,1-c][1,4]oxazin- 8(1H)-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-25-hydroxy-10,10-dimethyl-5,7-dioxo-11H-8-oxa- 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- 40 yl)amino)-3-methyl-1-oxopent-4-en-2-yl)-N-methyl-3-phenyloxetane-2-carboxamide PAT059646-WO-PCT 5 Step a: To a solution of Intermediate 9a (300 mg) and Intermediate 13 (271 mg) in 1,4-dioxane (3 mL) and water (405 L) was added Cs2CO3 (281 mg) and Pd(dppf)Cl2-DCM adduct (16.9 mg). The reaction was stirred at 80°C under nitrogen for 2 hr. The reaction was then diluted with EtOAc 10 and water. The aq. layer was extracted with EtOAc (2x). The EtOAc extracts were washed with brine, dried with Na2SO4, filtered, and concentrated. The residue was purified by column chromatography (SiO2; 0 to 100% 3:1 EtOAc/EtOH in heptane) to afford tert-butyl ((61R,65S,66S,4S)-12-(5-((S)-hexahydropyrazino[2,1-c][1,4]oxazin-8(1H)-yl)-2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-15 diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)carbamate (223 mg). LC/MS (Peptide Method) M/Z = 1020.6100 [M+H]+. 246 PAT059646-WO-PCT 5 Step b: To a solution of tert-butyl ((61R,65S,66S,4S)-12-(5-((S)-hexahydropyrazino[2,1- c][1,4]oxazin-8(1H)-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)carbamate (223 mg) in DMF (2.0 mL) was added DIPEA (113 10 mg, 152 L), Cs2CO3 (284 mg) and EtI (85 mg, 43.8 L). The reaction was stirred at RT for 5.5 hr, after which time EtOAc and water were added and the aq. layer was extracted with EtOAc (2x). The EtOAc extracts were washed with brine, dried with Na2SO4, filtered, and concentrated. The residue was purified by column chromatography (SiO2; 0 to 100% 3:1 EtOAc/EtOH in heptane) to afford tert-butyl ((61R,65S,66S,4S)-11-ethyl-12-(5-((S)-15 hexahydropyrazino[2,1-c][1,4]oxazin-8(1H)-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)carbamate (232 mg) as a mixture of atropisomers. LC/MS (RXNMON-Acidic method): M/Z = 1048.9 [M+H]+. 20 Step c: To a solution of tert-butyl ((61R,65S,66S,4S)-11-ethyl-12-(5-((S)-hexahydropyrazino[2,1- c][1,4]oxazin-8(1H)-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)carbamate (232 mg) in DCM (2 mL) was added TFA (1 mL). The reaction was stirred at RT for 20 min. and concentrated. The residue was purified by column25 chromatography (C18; 10 to 43% acetonitrile in water + 0.1% TFA) to afford (61R,65S,66S,4S)-4- amino-11-ethyl-12-(5-((S)-hexahydropyrazino[2,1-c][1,4]oxazin-8(1H)-yl)-2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza- 1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-5,7-dione trifluoroacetate (55.4 mg) as a mixture of atropisomers. LC/MS (Peptide Method) M/Z = 948.5500 30 [M+H]+. Step d: To a solution of (61R,65S,66S,4S)-4-amino-11-ethyl-12-(5-((S)-hexahydropyrazino[2,1- c][1,4]oxazin-8(1H)-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- 35 benzenacycloundecaphane-5,7-dione trifluoroacetate (55.4 mg) in DMF (474 L) was added Intermediate 23 (23.1 mg), DIPEA (49.1 mg, 66.1 L), and HATU (32.5 mg). The reaction was stirred at RT for 1 hr. The reaction was purified by column chromatography (SiO2; 0 to 15% MeOH in DCM) to afford tert-butyl ((2R,3R)-1-(((61R,65S,66S,4S)-11-ethyl-12-(5-((S)- hexahydropyrazino[2,1-c][1,4]oxazin-8(1H)-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-40 dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopent-4-en- PAT059646-WO-PCT 5 2-yl)(methyl)carbamate (52.6 mg) as a mixture of atropisomers. LC/MS (Peptide Method) M/Z = 1173.6700 [M+H]+. Step e: To a solution of tert-butyl ((2R,3R)-1-(((61R,65S,66S,4S)-11-ethyl-12-(5-((S)- hexahydropyrazino[2,1-c][1,4]oxazin-8(1H)-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-10 dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopent-4-en- 2-yl)(methyl)carbamate (52.6 mg) in DCM (2 mL) was added TFA (1.0 mL). The reaction was stirred at RT for 30 min. and concentrated. The residue was purified by column chromatography (C18; 10 to 48% acetonitrile in water + 0.1% TFA) to afford (2R,3R)-N-((61R,65S,66S,4S)-11-ethyl-15 12-(5-((S)-hexahydropyrazino[2,1-c][1,4]oxazin-8(1H)-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)- 10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)pent-4- enamide trifluoroacetate (19.2 mg) as a mixture of atropisomers. LC/MS (Peptide Method) M/Z = 1073.6400 [M+H]+. 20 Step f: To a solution of (2R,3R)-N-((61R,65S,66S,4S)-11-ethyl-12-(5-((S)-hexahydropyrazino[2,1- c][1,4]oxazin-8(1H)-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)-3-methyl-2-(methylamino)pent-4-enamide trifluoroacetate (19.2 25 mg) in DMF (200 L) was added Intermediate 10 (5.8 mg), DIPEA (16.7 mg, 22.5 L), and HATU (11.1 mg). The reaction was stirred at RT for 2 hr. The reaction was purified by column chromatography (SiO2; 0 to 15% MeOH in DCM) to afford (2R,3S)-N-((2R,3R)-1- (((61R,65S,66S,4S)-11-ethyl-12-(5-((S)-hexahydropyrazino[2,1-c][1,4]oxazin-8(1H)-yl)-2-((S)-1- methoxyethyl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-30 diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)- 3-methyl-1-oxopent-4-en-2-yl)-N-methyl-3-phenyloxetane-2-carboxamide (15.6 mg) as a mixture of atropisomers. LC/MS (Peptide Method) M/Z = 1234.6899 Step g: To a solution of (2R,3S)-N-((2R,3R)-1-(((61R,65S,66S,4S)-11-ethyl-12-(5-((S)-35 hexahydropyrazino[2,1-c][1,4]oxazin-8(1H)-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-10,10- dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)- bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopent-4-en- 2-yl)-N-methyl-3-phenyloxetane-2-carboxamide (15.6 mg) in DMF (200 L) was added tetramethylammonium fluoride (1.5 mg). The reaction was stirred at RT for 20 min. The reaction 40 was purified by preparatory HPLC (C18; 25 to 50% acetonitrile in water + 0.1% NH4OH) to afford (2R,3S)-N-((2R,3R)-1-(((61R,65S,66S,4S)-11-ethyl-12-(5-((S)-hexahydropyrazino[2,1- PAT059646-WO-PCT 5 c][1,4]oxazin-8(1H)-yl)-2-((S)-1-methoxyethyl)pyridin-3-yl)-25-hydroxy-10,10-dimethyl-5,7-dioxo- 11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopent-4-en-2-yl)-N-methyl-3- phenyloxetane-2-carboxamide (4 mg) as the second eluting isomer. LC/MS (Peptide Method) M/Z = 1077.5900 [M+H]+. 1H NMR (400 MHz, DMSO) 8.45 (d, J = 2.9 Hz, 1H), 7.90 – 7.80 (m, 1H), 10 7.77 – 7.41 (m, 5H), 7.39 – 7.19 (m, 4H), 7.15 – 6.92 (m, 2H), 6.53 – 6.35 (m, 1H), 5.97 (d, J = 7.7 Hz, 1H), 5.65 – 5.21 (m, 2H), 5.01 (dd, J = 7.0, 5.2 Hz, 1H), 4.91 – 4.61 (m, 3H), 4.56 – 4.37 (m, 3H), 4.30 – 4.02 (m, 4H), 3.96 – 3.44 (m, 8H), 3.24 – 3.01 (m, 6H), 2.96 – 2.57 (m, 7H), 2.43 – 2.09 (m, 8H), 1.36 (d, J = 6.0 Hz, 3H), 1.27 – 0.88 (m, 6H), 0.86 – 0.31 (m, 7H), -0.09 (d, J = 6.8 Hz, 3H). 15 The following compounds of Table 15 were synthesized using the above procedure or modifications of the above procedure using the corresponding aryl boronic ester, amino acid, and carboxylic acid. Note: axial chirality of compounds is as shown in structures in Table 15. 20 Table 15: PanRAS inhibitor compounds PAT059646-WO-PCT 5 Compound 159 Note: axial chirality of compounds is as shown in below scheme. (2R,3S)-N-((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-25-hydroxy-12-(2-((S)-1-methoxyethyl)-5-(4-10 (oxetan-3-yl)piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-11H-8-oxa-63,64-diaza-1(5,3)- indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4-yl)amino)-3-methyl-1- oxopent-4-en-2-yl)-N-methyl-3-phenyloxetane-2-carboxamide 15 Procedure Step a: To a solution of Intermediate 33 (20.0 mg) and oxetan-3-one (4.4 mg, 3.9 L) in THF (200 L) was added acetic acid (605 g, 0.577 L). The reaction stirred for 1 hr then sodium triacetoxyborohydride (12.8 mg) was added. The reaction stirred at RT for 1.5 hr. The reaction PAT059646-WO-PCT 5 mixture was diluted with sat. NaHCO3 and EtOAc. The aqueous layer was extracted with EtOAc (3x), and the combined organic layers were washed with brine, dried over Na2SO4, filtered, and concentrated to afford crude tert-butyl ((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- (oxetan-3-yl)piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H- 8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane- 10 4-yl)carbamate (21 mg) as an off-white foam. LC/MS (Peptide Method): M/Z = 1048.7000 [M+H]+. Step b: To a solution of tert-butyl ((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- (oxetan-3-yl)piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H- 8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane- 15 4-yl)carbamate (21.0 mg) in DCM (150 L) was added TFA (50 L). The reaction stirred at RT for 40 min. The reaction mixture was concentrated. The product was dissolved in DCM and concentrated (3x) then dried under reduced pressure to afford (61R,65S,66S,4S)-4-amino-11-ethyl- 12-(2-((S)-1-methoxyethyl)-5-(4-(oxetan-3-yl)piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- 20 benzenacycloundecaphane-5,7-dione (30 mg) as a yellow foam. LC/MS (Peptide Method): M/Z = 948.5800 [M+H]+. Step c: To a solution of (61R,65S,66S,4S)-4-amino-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- (oxetan-3-yl)piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-25-((triisopropylsilyl)oxy)-11H-8-oxa-25 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-5,7- dione (30 mg), Intermediate 23 (7.6 mg), and DIPEA (25.9 mg, 34.9 L) in acetonitrile (150 L) and 2-MeTHF (50 L) was added COMU (13.7 mg). The reaction stirred at RT for 30 min. The reaction mixture was poured into a separatory funnel and diluted with EtOAc and sat. NaHCO3. The aqueous layer was extracted with EtOAc (3x), and the combined organic layers were washed 30 with brine, dried over Na2SO4, filtered, concentrated, and dried under reduced pressure to afford crude tert-butyl ((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4-(oxetan- 3-yl)piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H-8-oxa- 63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane-4- yl)amino)-3-methyl-1-oxopent-4-en-2-yl)(methyl)carbamate (31 mg) as a dull yellow foam. LC/MS 35 (Peptide Method): M/Z = 1173.7200 [M+H]+. Step d: To a solution of tert-butyl ((2S,3R)-1-(((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1- methoxyethyl)-5-(4-(oxetan-3-yl)piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25- ((triisopropylsilyl)oxy)-11H-8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)- 40 benzenacycloundecaphane-4-yl)amino)-3-methyl-1-oxopent-4-en-2-yl)(methyl)carbamate (31 mg) in DCM (150 L) was added TFA (50.0 L). The reaction stirred at RT for 40 min. The reaction PAT059646-WO-PCT 5 mixture was concentrated. It was dissolved in DCM and concentrated (3x). It was dried under reduced pressure to afford (2S,3R)-N-((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- (oxetan-3-yl)piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H- 8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane- 4-yl)-3-methyl-2-(methylamino)pent-4-enamide (41 mg) as a yellow foam. LC/MS (Peptide 10 Method): M/Z = 1073.6700 [M+H]+. Step e: To a solution of (2S,3R)-N-((61R,65S,66S,4S)-11-ethyl-12-(2-((S)-1-methoxyethyl)-5-(4- (oxetan-3-yl)piperazin-1-yl)pyridin-3-yl)-10,10-dimethyl-5,7-dioxo-25-((triisopropylsilyl)oxy)-11H- 8-oxa-63,64-diaza-1(5,3)-indola-6(3,5)-bicyclo[4.1.0]heptana-2(1,3)-benzenacycloundecaphane- 15 4-yl)-3-methyl-2-(methylamino)pent-4-enamide (41 mg), Intermediate 10 (5.4 mg) and DIPEA (25.9 mg, 34.9 L) in DMF (0.20 mL) was added HATU (11.4 mg). The reaction stirred for 16 hr at RT. Step f: To the reaction mixture from Step e was added ammonium hydroxide (10.6 L, 30% wt.). 20 The reaction stirred for 15 min. then TMAF (6 mg) was added. The mixture was sonicated then stirred for 30 min. at RT. The reaction mixture was diluted with DMSO, filtered, and purified by HPLC (50-70% CH3CN in H2O + 12 mM NH4OH). The fractions containing the desired product were partially concentrated and the aqueous layer was extracted with DCM (3x). The combined organic layers were passed through a phase separator, concentrated, and lyophilized to afford 25 Compound 159 (9.2 mg) as an off-white solid.1H NMR (400 MHz, DMSO) 9.42 (s, 1H), 8.46 (d, J = 2.9 Hz, 1H), 7.87 (s, 1H), 7.72 (d, J = 8.5 Hz, 1H), 7.58 (d, J = 8.7 Hz, 1H), 7.50 – 7.45 (m, 3H), 7.32 (t, J = 7.5 Hz, 2H), 7.29 – 7.22 (m, 2H), 7.09 (s, 1H), 6.95 (s, 1H), 6.46 (s, 1H), 5.97 (d, J = 7.6 Hz, 1H), 5.57 – 5.44 (m, 1H), 5.38 – 5.30 (m, 1H), 5.05 – 4.98 (m, 1H), 4.89 – 4.81 (m, 1H), 4.79 – 4.68 (m, 2H), 4.56 (t, J = 6.4 Hz, 2H), 4.51 (d, J = 9.8 Hz, 1H), 4.49 – 4.38 (m, 4H), 30 4.29 – 4.19 (m, 1H), 4.17 – 4.03 (m, 3H), 3.89 (d, J = 11.6 Hz, 1H), 3.72 (d, J = 11.0 Hz, 1H), 3.59 (d, J = 10.8 Hz, 1H), 3.48 – 3.41 (m, 1H), 3.30 (m, 4H), 3.23 – 3.15 (m, 1H), 3.12 – 3.08 (m, 3H), 2.85 – 2.69 (m, 2H), 2.67 (s, 1H), 2.65 – 2.61 (m, 1H), 2.44 – 2.39 (m, 4H), 2.37 (s, 2H), 2.20 – 2.12 (m, 1H), 1.36 (d, J = 6.1 Hz, 3H), 1.23 (s, 3H), 1.20 – 1.15 (m, 2H), 0.97 (t, J = 7.1 Hz, 2H), 0.80 (s, 4H), 0.46 (s, 2H), 0.15 – 0.10 (m, 1H), -0.05 – -0.13 (m, 3H). LC/MS (Peptide Method): 35 M/Z = 1077.5900 [M+H]+. The following compounds of Table 16 were synthesized using the above procedure or modifications of the above procedure using the corresponding macrocycle, amino acid, and terminal carboxylic acid. Note: axial chirality of compounds is as shown in structures in Table 16. 40 Table 16: PanRAS inhibitor compounds PAT059646-WO-PCT 5 Example 2: Determination of Axial isomers Some of the compounds of the disclosure show axial chirality due to hindered rotation about the bond that connects a substituted pyridine with a substituted indole. Due to the high energy 10 barrier of this rotation, there are two possible separable axial isomers (called atropisomers) due to the rotation position possible for these cases, and if stated that the structures have “axial chirality as shown,” and a single atropisomer is drawn, then the compound as numbered is the single atropisomer which is depicted. This includes compounds 101-103 and 106-158. The determination of the axial stereochemistry of the compounds by X-ray diffraction was performed 15 as follows. PAT059646-WO-PCT 5 Compound 101 was co-crystallized with NRAS_Q61R and CypA according to the following procedure. NRAS_Q61R, compound 101 and CYPA were mixed at 1.25mM : 1mM : 1mM molar ratio in a buffer of 20mM HEPES pH 7.5, 100mM NaCl. The protein complex was crystallized under 0.1 M Bis-Tris propane/HCl, pH 7 and 2 M ammonium citrate dibasic using sitting drop vapor diffusion method. Single crystal was harvested and diffracted under synchrotron radiation. 10 The dataset was processed to 1.94 angstrom resolution. Structure was solved by molecular replacement and refined to Rwork/Rfree: 0.182/0.230. NRAS_Q61R is a peptide with the following amino acid sequence: SEQ ID No.1: GPMTEYKLVVVGAGGVGKSALTIQLIQNHFVDEYDPTIEDSYRKQVVIDGETCLLDILDTAG 15 REEYSAMRDQYMRTGEGFLCVFAINNSKSFADINLYREQIKRVKDSDDVPMVLVGNKCDL PTRTVDTKQAHELAKSYGIPFIETSAKTRQGVEDAFYTLVREIRQYRMK. CypA is a peptide with the following sequence: SEQ ID No.2: GMVNPTVFFDIAVDGEPLGRVSFELFADKVPKTAENFRALSTGEKGFGYKGSCFHRIIPGFMCQ 20 GGDFTRHNGTGGKSIYGEKFEDENFILKHTGPGILSMANAGPNTNGSQFFICTAKTEWLDGKH VVFGKVKEGMNIVEAMERFGSRNGKTSKKITIADCGQLE. The structure including axial chirality was determined to be according to the below structure: 25 The axial chirality of 101 derives from the axial chirality of Intermediate 2. As Intermediate 2 was used in the synthesis of compounds 101-103 and 106-158, it is therefore determined that the axial stereochemistry of compounds 102-103 and 106-158 is the same as that of compound 101. 30 The 3-D structure of compound 101 as revealed by X-ray diffraction is shown in Fig.1. The PDB coordinates for the X-ray diffraction are shown in Fig.2. PAT059646-WO-PCT 5 Example 3: Assays of biological activity Cell Lines Compounds were tested against the following cancer cell lines: NCI-H358: ATCC No. CRL-5807 cultured in RPMI-1640 + 10% FBS 10 NCI-H441: ATCC No. HTB-174 cultured in RPMI-1640 + 10% FBS Panc0203: ATCC No. CRL-2553 cultured in RPMI-1640 + 15% FBS + 4 ug/mL insulin NCI-H2122: ATCC No. CRL-5985 cultured in RPMI-1640 + 10 %FBS Inhibition of cell proliferation and survival 15 The ability of the compounds to inhibit cell proliferation and survival was assessed using the Promega CellTiter-Glo® proliferation assay. Cell lines were cultured in media that is optimal for their growth at 5% CO2, 37°C in a tissue culture incubator. Prior to seeding for the proliferation assay, the cells were split at least 2 days before the assay to ensure optimal growth density. On the day of seeding, cell viability and 20 cell density were determined using a cell counter (Vi-Cell XR Cell Viability Analyzer, Beckman Coulter). Cells with higher than 85% viability were seeded in white clear bottom 384-well TC treated plates (Corning cat. # 3765). Cells were seeded at a density of 1,000 cells per well in 45 L of standard growth media. Plates were incubated at 5% CO2, 37°C overnight in a tissue culture incubator. The next day, indicated compounds were prepared at 10X in standard growth media. 25 The prepared compounds were then added to the cells resulting in final concentrations of 0.005 – 100 nM and a final volume of 50 uL per well. Each compound concentration was tested in quadruplets. Plates were incubated at 5% CO2, 37°C for 5 days in a tissue culture incubator, after which cell viability was assessed through the addition of 25 L of CellTiter Glo® (Promega, cat# G7573), a reagent which lyses cells and measures total adenosine triphosphate (ATP) content. 30 Plates were incubated at room temperature for 10 min. to stabilize luminescent signals prior to reading using a luminescence reader (EnVision Multilabel Plate Reader, PerkinElmer). To evaluate the effect of the drug treatments, luminescent counts from wells containing untreated cells (100% viability) were used to normalize treated samples. A variable slope model was applied to fit a nonlinear regression curve to the data in GraphPad PRISM version 7.02 software. IC50 35 and Amax values were extrapolated from the resultant curves. The concentrations of treatment required to inhibit 50% of cell growth or survival (GI50) were calculated with representative GI50 values of the cell lines tested summarized in Table 17 along with associated structures in Table 20. 40 Cell activities PAT059646-WO-PCT 5 Table 17: Biological assay results for panRAS inhibitors PAT059646-WO-PCT 5 Binary SPR assay Materials and instruments: 1. Avi-tagged Cyclophilin A (human) 10 2. Streptavidin(SA) sensor chip: Cytiva series S Cat# BR100531 3. SPR assay buffer: 20 mM HEPES pH 7.5, 150 mM NaCl, 1 mM TECP, 0.025% P20, 2% DMSO 4.8K Biacore: Cytiva 5. Biomek liquid handler: Beckman 15 6. ECHO 550: Labcyte 7. Dragonfly Discovery: SPT Labtech Method: The binary SPR assay was used to measure the direct binding between a Pan-RAS inhibitor 20 and Cyclophilin A (CypA). The assay was run using the multi-cycle kinetics binding assay format with 8K Biacore instrument employing 10-points and 1 to 3 serial dilution compound plate (prepared with ECHO instrument).0% DMSO assay buffer was added to the plate to DMSO final concentration at 2%. Compound final concentration was from 10 uM to 0.51 nM and the last point was a blank control. Avi-CypA was immobilized to SA-chip surface to 300 RU level. 25 The assay was run at 25°C . Flow rate was set to 50 μl/min, contact time 60 sec., dissociation time 600 sec. and 15 start-up cycles. DMSO solvent correction was run before and after the sample running. All analysis of data were performed using Cytiva’s Biacore Insight Evaluation Software. The predefined evaluation method “LMW multi-cycle kinetics” was selected for data analysis.1:1 binding kinetics fit model was applied, and steady state affinity fit mode was 30 applied to weak compounds which couldn’t be analyzed with kinetics fit mode. PAT059646-WO-PCT 5 Ternary SPR assay Materials and instruments: 1. Cyclophilin A (human) 2. Avi-tagged KRAS G12C GMPPNP 10 3. Streptavidin(SA) sensor chip: Cytiva series S Cat# BR100531 4. SPR assay buffer: 50 mM HEPES pH 7.5, 150 mM NaCl, 1 mM MgCl2, 0.01 mM GMPPNP, 1 mM TECP, 0.025% P20, 2% DMSO 5.8K Biacore: Cytiva 6. Biomek liguid handler: Beckman 15 7. ECHO 550: Labcyte 8. Dragonfly Discovery: SPT Labtech Method: The ternary SPR assay was used to measure the binding between KRAS G12C and (Pan-RAS 20 inhibitor + Cyclophilin A). The assay was run using the multi-cycle kinetics binding assay format with 8K Biacore instrument employing 10-points and 1 to 3 serial dilution compound plate (prepared with ECHO instrument).2.5 uM Cyclophilin A solution, which was made with 0% DMSO assay buffer, was added to the plate to DMSO final concentration at 2%. Compound final concentration was from 10 uM to 0.51 nM and the last point was blank control. Avi-KRAS G12C 25 GMPPNP was immobilized to SA-chip surface to 100 RU level. The assay was run at 22°C . The flow rate was set to 30 μl/min, contact time 60 sec., dissociation time 600 sec. and 15 start- up cycles.7-points DMSO solvent correction was run before and after the sample running. All analysis of data were performed using Cytiva’s Biacore Insight Evaluation Software. The predefined evaluation method “LMW multi-cycle kinetics” was selected for data analysis.1:1 30 binding kinetics fit model was applied, and steady state affinity fit mode was applied to weak compounds which couldn’t be analyzed with kinetics fit mode. SPR activities Table 18: Biological assay results for panRAS inhibitors PAT059646-WO-PCT 259 PAT059646-WO-PCT 5 Example 4: Single dose pharmacokinetic study in male mice. The purpose of this study was to determine the pharmacokinetics of selected compounds following single intravenous bolus in male C57BL/6 mice. Mice (N = 3/group) were treated with a 10 single dose of compound intravenously at 1 mg/kg. Blood samples were collected at 0.83 hr, 0.25 hr, 0.5 hr, 1 hr, 3 hr, 7 hr, and 24 hr post-dose. The concentration of compound in blood samples was determined using high performance LC-MS/MS. Table 19: Pharmacokinetic data for select compounds 15 Table 20: Structures of panRAS inhibitors Note: axial chirality of compounds is as shown in the structures in Table 20. 260 PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT
PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT PAT059646-WO-PCT

Claims

PAT059646-WO-PCT 5 CLAIMS 1. A compound of formula (I) or formula (XX), 10 wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; 15 ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 20 N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, NRxCOR6, and COR6 R2 is optionally substituted 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, 25 and S; PAT059646-WO-PCT 5 R3; is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6, or R3 is absent; each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 10 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; 15 L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl, aryl, or 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; or R7 is C3-6 cycloalkyl; wherein R7 is optionally further substituted; 20 R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; each Rx is independently selected from H, C1-6 alkyl, C3-6 cycloalkyl, and C1-6 haloalkyl; W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S; and R9 is optionally substituted; 25 m is 0 or 1; and n is 0, 1, 2, or 3, wherein at least one of the following is true: a) R9 is 2-oxetanyl or 2-azetidinyl, each optionally substituted; or b) R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; 30 wherein R7 is optionally further substituted. 2. A compound of formula (I), PAT059646-WO-PCT 5 wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- 10 membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and 15 ring B is edge-fused to ring C to form a BC ring system; each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6 ; R2 is substituted pyridine or substituted phenyl; R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 20 haloalkyl, NH2, and COR6, or R3 is absent; each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or 25 two R5 groups are taken together with the carbon atom to which they are connected to form a C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; 30 R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl, aryl, or 5-6 membered heteroaryl with 1-3 ring atoms selected from N, O, and S; or R7 is C3-6 cycloalkyl; wherein R7 is optionally further substituted; R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms 35 independently selected from the group consisting of N, O and S, and R9 is optionally substituted; m is 0 or 1; and n is 0, 1, 2, or 3, wherein at least one of the following is true: 40 a) R9 is 2-oxetanyl or 2-azetidinyl, each optionally substituted; or PAT059646-WO-PCT 5 b) R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; wherein R7 is optionally further substituted. 3. A compound of formula (I) 10 wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; 15 ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 20 N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6 ; R2 is substituted pyridine or substituted phenyl; 25 R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6, or R3 is absent; each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 30 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; PAT059646-WO-PCT 5 L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W, or L is absent; R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl; or R7 is C3-6 cycloalkyl; wherein R7 is optionally further substituted; R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; W is -COR9, wherein R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms 10 independently selected from the group consisting of N, O and S; and R9 is optionally substituted; m is 0 or 1; and n is 0, 1, 2, or 3; wherein at least one of the following is true: 15 a) R9 is 2-oxetanyl or 2-azetidinyl, each optionally substituted; or b) R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; wherein R7 is optionally further substituted. 4. A compound of formula (Iw) or formula (XXw) 20 wherein ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; PAT059646-WO-PCT 5 ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 10 N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6 ; R2 is substituted pyridine or substituted phenyl; 15 R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6, or R3 is absent; each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 20 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; 25 R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S; and R9 is optionally substituted; m is 0 or 1; and n is 0, 1, 2, or 3. 30 5. A compound of formula (Iw) PAT059646-WO-PCT 5 ring A is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring A is optionally substituted; ring B is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of 10 N, O, and S, and ring B is optionally substituted; ring C is 6-membered aryl, 6-membered heteroaryl with 1-3 ring atoms which are N, or 5- membered heteroaryl with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and ring C is optionally substituted; and ring B is edge-fused to ring C to form a BC ring system; 15 each R1 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6 ; R2 is substituted pyridine or substituted phenyl; R3 is selected from the group consisting of H, halo, OH, CN, C1-10 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6, or R3 is absent; 20 each R4 is independently selected from the group consisting of H, halo, C1-6 alkyl, and C1-6 haloalkyl; each R5 is independently selected from the group consisting of H, halo, OH, CN, C1-3 alkyl, C1-3 heteroalkyl, C1-3 haloalkyl, and C3-6 cycloalkyl; or two R5 groups are taken together with the carbon atom to which they are connected to form a 25 C3-6 cycloalkyl; each R6 is independently selected from the group consisting of H, OH, C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, and NH2; R9 is C3-7 cycloalkyl or 3-7 membered heterocyclyl having 1-3 ring atoms independently selected from the group consisting of N, O and S; and R9 is optionally substituted; 30 m is 0 and n is 0; or m is 0 and n is 1. 6. The compound of any one of claims 1-5, wherein ring A is phenyl, pyridine, thiazole, triazole, oxazole, or oxadiazole, and ring A is substituted with 0-3 R10; 35 wherein R10 is selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6. 7. The compound of claim 6, wherein ring A is PAT059646-WO-PCT 5 8. The compound of claim 7, wherein ring A is 10 9. The compound of any one of claims 1-8, wherein the BC ring system is indole, benzothiophene, benzoxazole, or indolizine, each of which optionally has 1-3 additional N ring atoms in its 6-membered ring, wherein each BC ring system is optionally substituted, for example wherein the BC ring system is optionally substituted with 1-3 R11, wherein each R11 is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 15 heteroalkyl, C1-6 haloalkyl, NH2, and COR6. 10. The compound of any one of claims 1-9, wherein the BC ring system is 20 for example wherein either 0 or 1 of X2 are N, and the remaining X2 are CR11, for example wherein the BC ring system is , wherein each R11 is independently selected from the group consisting of H, halo, OH, CN, C1-6 25 alkyl, C1-6 heteroalkyl, C1-6 haloalkyl, NH2, and COR6. 11. The compound of any one of claims 1-10, wherein PAT059646-WO-PCT 5 each R1 is independently selected from the group consisting of H, halo, C1-6 alkyl, or C1-6 haloalkyl; for example wherein each R1 is H. 12. The compound of any one of claims 1-11, wherein R2 is substituted pyridine. 10 13. The compound of any one of claims 1-12, wherein R2 is , wherein R2a is C1-10 alkyl, C3-6 cycloalkyl, or C1-10 heteroalkyl, and R2a is optionally substituted by 1-3 15 substituents independently selected from the group consisting of halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, -(CH2)0-2 phenyl, -(CH2)0-2-C3-6 cycloalkyl, C1-6 haloalkyl, NH2, NRxCOR6, and COR6; for example, wherein R2a is C1-10 optionally substituted heteroalkyl; R2b is 4-10-membered heterocycloalkyl, with 1-4 ring atoms selected from the group consisting of N, O, P(O)xx, and S(O)xx, wherein xx is 0, 1, or 2, and R2b is optionally substituted, for 20 example with 1-3 R2ba, wherein each R2ba is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, C1-6 heteroalkyl, -(CH2)0-2 phenyl, -(CH2)0-2-C3-6 cycloalkyl, , C1-6 haloalkyl, NH2, COR6, and -(CH2)0-2- heterocycloalkyl wherein the heterocycloalkyl is 4-7 membered with 1-3 ring atoms selected from the group consisting of N, O, P(O)xx, and S(O)xx, wherein xx is 0, 1, or 2, and wherein each 25 phenyl, heterocycloalkyl, and cycloalkyl is optionally substituted, for example with 1-3 substituents selected from the group consisting of halo, OH, CN, C1-6 alkyl, C1-6 haloalkyl, and C1-6 heteroalkyl; each R2c is independently selected from the group consisting of H, halo, OH, CN, C1-6 alkyl, and C1-6 heteroalkyl; for example, wherein each R2c is H; and 30 each Rx is independently selected from H, C1-6 alkyl, C3-6 cycloalkyl, and C1-6 haloalkyl. 14. The compound of claim 13, wherein R2b is 35 wherein X3 is C, CRx, N, or P(O); is a single or double bond; and PAT059646-WO-PCT 5 each Rx is independently selected from H, C1-6 alkyl, C3-6 cycloalkyl, and C1-6 haloalkyl; for example, wherein R2ba is H, C1-6 alkyl, -(CH2)0-2 phenyl, C1-6 heteroalkyl, C1-6 haloalkyl, C3-6 cycloalkyl, COR6, or heterocycloalkyl wherein the heterocycloalkyl is 4-7 membered with 1-3 ring atoms selected from the group consisting of N, O, and S. 10 15. The compound of claim 13 or 14 wherein R2a is , wherein X4 is O or N, and X5 is CH; or X5 is O or N, and X4 is CH; and 15 R2aa is C1-3 alkyl or C3-6 cycloalkyl, each optionally substituted with 1-3 halo, or R2aa is absent or H; R2ab is C1-3 alkyl or C3-6 cycloalkyl, each optionally substituted with 1-3 halo, or R2ab is absent or H; and R2ac is C1-3 alkyl or C3-6 cycloalkyl, each optionally substituted with 1-3 halo or one OH, or R2ac is 20 absent or H. 16. The compound of any one of claims 1-12, wherein 25 is a single or double bond R2aa is C1-3 alkyl or C3-6 cycloalkyl, each optionally substituted with 1-3 halo, or R2aa is absent or 30 H; R2ab is C1-3 alkyl or C3-6 cycloalkyl, each optionally substituted with 1-3 halo, or R2ab is absent or H; R2ac is C1-3 alkyl or C3-6 cycloalkyl, each optionally substituted with 1-3 halo or one OH; and PAT059646-WO-PCT 5 R2ba is H, C1-6 alkyl, -(CH2)0-2 phenyl, C1-6 heteroalkyl, C1-6 haloalkyl, C3-6 cycloalkyl, COR6, or heterocycloalkyl wherein the heterocycloalkyl is 4-7 membered with 1-3 ring atoms selected from the group consisting of N, O, and S, and each phenyl, cycloalkyl, or heterocycloalkyl is optionally substituted. 10 17. The compound of any one of claims 1-16, wherein R3 is selected from the group consisting of H, C1-6 alkyl, C1-6 heteroalkyl, and C1-6 haloalkyl, or R3 is absent; for example, R3 is C1-6 alkyl; for example, R3 is ethyl. 18. The compound of any one of claims 1-17, wherein 15 each R4 is independently selected from the group consisting of H, C1-6 alkyl, and C1-6 haloalkyl; for example, wherein each R4 is H. 19. The compound of any one of claims 1-18, wherein each R5 is independently selected from the group consisting of H and C1-6 alkyl; or 20 two R5 groups are taken together with the carbon atom to which they are connected to form a C3-6 cycloalkyl; for example wherein both R5 are CH3. 20. The compound of any one of claims 1-19, wherein 25 L is absent, or L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W; R7 is C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted by C3-6 cycloalkyl; or R7 is C3-6 cycloalkyl; wherein R7 is optionally further substituted by 1-3 substituents selected from the group consisting of halo, OH, C1-3 haloalkyl, C1-3 heteroalkyl, and C1-3 alkyl; and R8 is H or C1-3 alkyl. 30 21. The compound of any one of claims 1-20, wherein R7 is C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, or C1-6 alkyl substituted by C3-6 cycloalkyl, wherein R7 is optionally further substituted by 1-3 substituents selected from the group consisting of halo and C1-3 alkyl. 35 22. The compound of any one of claims 1-21, wherein R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; wherein R7 is optionally further substituted by 1-3 substituents selected from the group consisting of halo and C1-3 alkyl. 40 23. The compound of any one of claims 1-22, wherein PAT059646-WO-PCT 5 R7 is 10 24. The compound of any one of claims 1-23, wherein R9 is C3-7 cycloalkyl or 4-6 membered heterocycloalkyl having 1-3 ring atoms independently selected from the group consisting of N, O and S, wherein each R9 is optionally substituted by 1-3 R9a, and optionally substituted with 1 R9b on a ring nitrogen atom, if present; each R9a is independently selected from the group consisting of halo, OH, CN, C1-6 alkyl, C1-6 15 alkenyl, C1-6 alkynyl, C1-6 heteroalkyl, -(CH2)0-2 phenyl, -(CH2)0-2-C3-6 cycloalkyl, C1-6 haloalkyl, NH2, COR6, NRxCOR6, and -(CH2)0-2 heterocyclyl of 4-7 ring atoms with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and wherein each phenyl, heterocyclyl and cycloalkyl is optionally substituted, for example with 1-3 substituents selected from the group consisting of halo, OH, CN, C1-6 alkyl, C1-6 haloalkyl, and C1-6 heteroalkyl. 20 25. The compound of any one of claims 1-24, wherein R9 is 2-oxetanyl optionally substituted with 1-3 R9a. 26. The compound of any one of claims 1-25, wherein W is 25 PAT059646-WO-PCT 5 ,wherein R9b is H, C1-6 alkyl, C1-6 heteroalkyl, -(CH2)0-2 phenyl, -(CH2)0-2-C3-6 cycloalkyl, C1-6 haloalkyl, NH2, COR6, NRxCOR6, and -(CH2)0-2 heterocyclyl of 4-7 ring atoms with 1-3 ring atoms independently selected from the group consisting of N, O, and S, and wherein each phenyl, heterocyclyl and cycloalkyl is optionally substituted, for example with 1-3 substituents selected from the group consisting of halo, OH, CN, C1-6 alkyl, C1-6 haloalkyl, and 10 C1-6 heteroalkyl, for example wherein R9b is H, C1-3 alkyl, or NRxCO-C1-3 alkyl. 27. The compound of claim 26, wherein W is . 15 28. The compound of any one of claims 1-27 wherein the compound is a compound of formula (III), formula (XXIII), or formula (XXXIII) 20 PAT059646-WO-PCT 5 for example, wherein all R1 are H, for example wherein n is 0 or 1;for example a compound of formula (IIIa) 10 29. The compound of any one of claims 1-27, wherein the compound is a compound of formula (IV), formula (XXIV), or formula (XXXIV) 15 PAT059646-WO-PCT 5 wherein n is 0 or 1; for example, wherein all R1 are H; for example, wherein there are no R11 10 substituents; for example, wherein the compound is a compound of formula (IVa) for example, wherein all R1 are H; for example, wherein there are no R11 substituents. 15 30. The compound of any one of claims 1-27, wherein the compound is a compound of formula (V), formula (VI), formula (XXV), formula (XXVI), formula (XXXV), formula (XXXVI) PAT059646-WO-PCT 5 10 wherein n is 0 or 1; for example, wherein all R1 are H; for example, wherein all R5 are CH3; for example, wherein there are no R11 substituents; for example, a compound of formula (V), formula (XXV), or formula (XXXV) wherein there is one R10 selected from the group consisting of OH, NH2, and halo, and the remaining R10 are H; 15 for example, a compound of formula (VI), formula (XXVI), or formula (XXXVI) wherein R10 is H. 31. The compound of any one of claims 1-27, wherein the compound is a compound of formula (Vw) PAT059646-WO-PCT 5 for example, wherein m is 0, and n is 0 or 1; for example, wherein all R1 are H; for example, wherein all R5 are CH3; for example, wherein there are no R11 substituents; for example, a compound of formula (Vw) wherein there is one R10 selected from the group 10 consisting of OH, NH2, and halo, and the remaining R10 are H. 32. The compound of any one of claims 1-27, wherein the compound is a compound of formula (Va) or formula (VIa) 15 for example, wherein all R1 are H; for example, wherein all R5 are CH3; for example, wherein there are no R11 substituents; for example, a compound of formula (IVa) wherein there is one R10 selected from the group consisting of OH, NH2, and halo, and the remaining R10 are H; 20 for example, a compound of formula (Va) wherein R10 is H. 33. The compound of any one of claims 1-27, wherein the compound is a compound of formula (VII), formula (XXVII), or formula (XXXVII) PAT059646-WO-PCT 5 10 for example, wherein all R1 are H. 34. The compound of any one of claims 1-27 wherein the compound is a compound of formula (Iy’) or formula (XXy) PAT059646-WO-PCT 5 for example, wherein all R1 are H, for example wherein m is 1 and n is 0 or 1; 10 35. The compound of any one of claims 1-27 wherein the compound is a compound of formula (IIIy’), formula (XXIIIy’), or formula (XXXIIIy’) PAT059646-WO-PCT 5 10 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H, for example wherein n is 0 or 1; for example a compound of formula (IIIya’) 15 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H. 36. The compound of any one of claims 1-27, wherein the compound is a compound of 20 formula (Ivy’), formula (XXIVy’), or formula (XXXIVy’) PAT059646-WO-PCT 5 10 wherein n is 0 or 1; wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H; for example, wherein there are no R11 substituents; for example, wherein the compound is a compound of formula (IVya’) PAT059646-WO-PCT 5 wherein the variables are defined as in various embodiments of the disclosure; for example, wherein all R1 are H; for example, wherein there are no R11 substituents. 10 37. The compound of any one of claims 1-27, wherein the compound is a compound of formula (Vy’) or formula (VIy’), formula (XXVy’), formula (XXVIy’), formula (XXXVIy’) or formula (XXXVIy’)
wherein n is 0 or 1 ; wherein the variables are defined as in various embodiments of the disclosure; PAT059646-WO-PCT 5 for example, wherein all R1 are H; for example, wherein all R5 are CH3; for example, wherein there are no R11 substituents; for example, a compound of formula (Vy’), formula (XXVy’), or formula (XXXVy’) wherein there is one R10 selected from the group consisting of OH, NH2, and halo, and the remaining R10 are H; 10 for example, a compound of formula (VIy’), formula (XXVIy’), or formula (XXXVIy’) wherein R10 is H. 38. The compound of any one of claims 1-27, wherein the compound is a compound of formula (Vya’) or formula (VIya’) 15 wherein the variables are defined as in various embodiments of the disclosure; 20 for example, wherein all R1 are H; for example, wherein all R5 are CH3; for example, wherein there are no R11 substituents; for example, a compound of formula (Vya’) wherein there is one R10 selected from the group consisting of OH, NH2, and halo, and the remaining R10 are H; for example, a compound of formula (VIya’) wherein R10 is H. 25 39. The compound of any one of claims 1-27, wherein the compound is a compound of formula (VIIy), formula (VIIIy), formula (XXVIIy), formula (XXVIIIy), formula (XXXVIIy), or formula (XXXVIIIy) PAT059646-WO-PCT 5 10 PAT059646-WO-PCT 5 for example, wherein all R1 are H. 10 40. A compound of any one of claims 1-39, wherein L is -COCHR7N(R8)-*, wherein * indicates the point of attachment to W; R7 is C2-6 alkenyl, C2-6 alkynyl, C4-6 cycloalkenyl, or C1-6 alkyl substituted by C4-6 cycloalkenyl; wherein R7 is optionally further substituted; and R8 is H, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl; 15 for example wherein R7 is . 41. A compound selected from the compounds disclosed in the specification. 20 42. A pharmaceutical composition comprising the compound of any one of claims 1-41 and a pharmaceutically acceptable carrier. 43. A method of treating cancer in a subject in need thereof, the method comprising 25 administering to the subject a therapeutically effective amount of the compound of any one of claims 1-41, or the pharmaceutical composition of claim 42. PAT059646-WO-PCT 5 44. The method of claim 43, wherein the cancer is a tumor or a hematological cancer, optionally, the cancer is a breast cancer including ER positive breast cancer, multiple myeloma, plasma cell myeloma, leukemia, lymphoma, sarcoma, gastric or stomach cancer, acute myeloid leukemia, bladder cancer, brain cancer, bone marrow cancer, cervical cancer, chronic 10 lymphocytic leukemia, colorectal cancer, pancreatic cancer, esophageal cancer, hepatocellular cancer, lymphoblastic leukemia including acute lymphoblastic leukemia, follicular lymphoma, lymphoid malignancies of T-cell or B-cell origin, metastatic castration resistant prostate cancer, bladder urothelial carcinoma, melanoma, myelogenous leukemia, myeloma, oral cancer, ovarian cancer, non-small cell lung cancer, prostate cancer, small cell lung cancer, spleen cancer, or 15 head and neck cancer. 45. The method of claim 43, wherein the cancer is non-small cell lung cancer, pancreatic cancer, colorectal cancer, melanoma, head and neck cancer, acute myeloid leukemia, or bladder cancer. 20
PCT/IB2025/053708 2024-04-10 2025-04-08 Macrocyclic panras inhibitors for the treatment of cancer Pending WO2025215536A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202463632181P 2024-04-10 2024-04-10
US63/632,181 2024-04-10
US202463698688P 2024-09-25 2024-09-25
US63/698,688 2024-09-25

Publications (1)

Publication Number Publication Date
WO2025215536A1 true WO2025215536A1 (en) 2025-10-16

Family

ID=95477365

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2025/053708 Pending WO2025215536A1 (en) 2024-04-10 2025-04-08 Macrocyclic panras inhibitors for the treatment of cancer

Country Status (1)

Country Link
WO (1) WO2025215536A1 (en)

Citations (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2779780A (en) 1955-03-01 1957-01-29 Du Pont 1, 4-diamino-2, 3-dicyano-1, 4-bis (substituted mercapto) butadienes and their preparation
WO2000035436A2 (en) 1998-12-16 2000-06-22 Warner-Lambert Company Treatment of arthritis with mek inhibitors
WO2001051056A1 (en) 2000-01-10 2001-07-19 Astrazeneca Ab Fulvestrant formulation
WO2002006213A2 (en) 2000-07-19 2002-01-24 Warner-Lambert Company Oxygenated esters of 4-iodo phenylamino benzhydroxamic acids
WO2003076424A1 (en) 2002-03-08 2003-09-18 Eisai Co. Ltd. Macrocyclic compounds useful as pharmaceuticals
WO2003077914A1 (en) 2002-03-13 2003-09-25 Array Biopharma, Inc N3 alkylated benzimidazole derivatives as mek inhibitors
WO2005121142A1 (en) 2004-06-11 2005-12-22 Japan Tobacco Inc. 5-amino-2,4,7-trioxo-3,4,7,8-tetrahydro-2h-pyrido’2,3-d! pyrimidine derivatives and related compounds for the treatment of cancer
WO2007004415A1 (en) 2005-07-01 2007-01-11 Murata Manufacturing Co., Ltd. Multilayer ceramic substrate, process for producing the same and composite green sheet for production of multilayer ceramic substrate
WO2008016893A1 (en) 2006-08-02 2008-02-07 Novartis Ag Smac peptidomimetics useful as iap inhibitors
WO2008024725A1 (en) 2006-08-21 2008-02-28 Genentech, Inc. Aza-benzofuranyl compounds and methods of use
WO2009085983A1 (en) 2007-12-19 2009-07-09 Genentech, Inc. 5-anilinoimidazopyridines and methods of use
US7612114B2 (en) 2002-12-26 2009-11-03 Eisai R&D Management Co., Ltd. Selective estrogen receptor modulator
WO2010020675A1 (en) 2008-08-22 2010-02-25 Novartis Ag Pyrrolopyrimidine compounds as cdk inhibitors
WO2011076786A1 (en) 2009-12-22 2011-06-30 Novartis Ag Substituted isoquinolinones and quinazolinones
WO2011156518A2 (en) 2010-06-10 2011-12-15 Aragon Pharmaceuticals, Inc. Estrogen receptor modulators and uses thereof
WO2011159769A2 (en) 2010-06-17 2011-12-22 Aragon Pharmaceuticals, Inc. Indane estrogen receptor modulators and uses thereof
US20120071535A1 (en) 2010-09-16 2012-03-22 Aragon Pharmaceuticals, Inc. Estrogen receptor modulators and uses thereof
WO2013019906A1 (en) 2011-08-01 2013-02-07 Genentech, Inc. Methods of treating cancer using pd-1 axis binding antagonists and mek inhibitors
WO2013110890A1 (en) 2012-01-24 2013-08-01 Les Laboratoires Servier New indolizine derivatives, method for preparing same and pharmaceutical compositions containing same
WO2014130310A1 (en) 2013-02-19 2014-08-28 Novartis Ag Benzothiophene derivatives and compositions thereof as selective estrogen receptor degraders
WO2015011400A1 (en) 2013-07-23 2015-01-29 Les Laboratoires Servier Novel pyrrole derivatives, method for the production thereof and pharmaceutical compositions containing same
WO2015066188A1 (en) 2013-11-01 2015-05-07 Novartis Ag Aminoheteroaryl benzamides as kinase inhibitors
WO2015136017A1 (en) 2014-03-13 2015-09-17 F. Hoffmann-La Roche Ag Methods and compositions for modulating estrogen receptor mutants
WO2021091956A1 (en) 2019-11-04 2021-05-14 Revolution Medicines, Inc. Ras inhibitors
WO2022060836A1 (en) 2020-09-15 2022-03-24 Revolution Medicines, Inc. Indole derivatives as ras inhibitors in the treatment of cancer
WO2024067857A1 (en) * 2022-09-29 2024-04-04 南京明德新药研发有限公司 Macrocyclic derivative and use thereof

Patent Citations (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2779780A (en) 1955-03-01 1957-01-29 Du Pont 1, 4-diamino-2, 3-dicyano-1, 4-bis (substituted mercapto) butadienes and their preparation
WO2000035436A2 (en) 1998-12-16 2000-06-22 Warner-Lambert Company Treatment of arthritis with mek inhibitors
WO2001051056A1 (en) 2000-01-10 2001-07-19 Astrazeneca Ab Fulvestrant formulation
WO2002006213A2 (en) 2000-07-19 2002-01-24 Warner-Lambert Company Oxygenated esters of 4-iodo phenylamino benzhydroxamic acids
WO2003076424A1 (en) 2002-03-08 2003-09-18 Eisai Co. Ltd. Macrocyclic compounds useful as pharmaceuticals
WO2003077914A1 (en) 2002-03-13 2003-09-25 Array Biopharma, Inc N3 alkylated benzimidazole derivatives as mek inhibitors
US7612114B2 (en) 2002-12-26 2009-11-03 Eisai R&D Management Co., Ltd. Selective estrogen receptor modulator
WO2005121142A1 (en) 2004-06-11 2005-12-22 Japan Tobacco Inc. 5-amino-2,4,7-trioxo-3,4,7,8-tetrahydro-2h-pyrido’2,3-d! pyrimidine derivatives and related compounds for the treatment of cancer
WO2007004415A1 (en) 2005-07-01 2007-01-11 Murata Manufacturing Co., Ltd. Multilayer ceramic substrate, process for producing the same and composite green sheet for production of multilayer ceramic substrate
WO2008016893A1 (en) 2006-08-02 2008-02-07 Novartis Ag Smac peptidomimetics useful as iap inhibitors
WO2008024725A1 (en) 2006-08-21 2008-02-28 Genentech, Inc. Aza-benzofuranyl compounds and methods of use
WO2009085983A1 (en) 2007-12-19 2009-07-09 Genentech, Inc. 5-anilinoimidazopyridines and methods of use
WO2010020675A1 (en) 2008-08-22 2010-02-25 Novartis Ag Pyrrolopyrimidine compounds as cdk inhibitors
US8415355B2 (en) 2008-08-22 2013-04-09 Novartis Ag Pyrrolopyrimidine compounds and their uses
US8685980B2 (en) 2008-08-22 2014-04-01 Novartis Ag Pyrrolopyrimidine compounds and their uses
WO2011076786A1 (en) 2009-12-22 2011-06-30 Novartis Ag Substituted isoquinolinones and quinazolinones
WO2011156518A2 (en) 2010-06-10 2011-12-15 Aragon Pharmaceuticals, Inc. Estrogen receptor modulators and uses thereof
WO2011159769A2 (en) 2010-06-17 2011-12-22 Aragon Pharmaceuticals, Inc. Indane estrogen receptor modulators and uses thereof
WO2012037411A2 (en) 2010-09-16 2012-03-22 Aragon Pharmaceuticals, Inc. Estrogen receptor modulators and uses thereof
WO2012037410A2 (en) 2010-09-16 2012-03-22 Aragon Pharmaceuticals, Inc. Estrogen receptor modulators and uses thereof
US20120071535A1 (en) 2010-09-16 2012-03-22 Aragon Pharmaceuticals, Inc. Estrogen receptor modulators and uses thereof
WO2013019906A1 (en) 2011-08-01 2013-02-07 Genentech, Inc. Methods of treating cancer using pd-1 axis binding antagonists and mek inhibitors
WO2013110890A1 (en) 2012-01-24 2013-08-01 Les Laboratoires Servier New indolizine derivatives, method for preparing same and pharmaceutical compositions containing same
WO2014130310A1 (en) 2013-02-19 2014-08-28 Novartis Ag Benzothiophene derivatives and compositions thereof as selective estrogen receptor degraders
WO2015011400A1 (en) 2013-07-23 2015-01-29 Les Laboratoires Servier Novel pyrrole derivatives, method for the production thereof and pharmaceutical compositions containing same
WO2015066188A1 (en) 2013-11-01 2015-05-07 Novartis Ag Aminoheteroaryl benzamides as kinase inhibitors
WO2015136017A1 (en) 2014-03-13 2015-09-17 F. Hoffmann-La Roche Ag Methods and compositions for modulating estrogen receptor mutants
WO2021091956A1 (en) 2019-11-04 2021-05-14 Revolution Medicines, Inc. Ras inhibitors
WO2022060836A1 (en) 2020-09-15 2022-03-24 Revolution Medicines, Inc. Indole derivatives as ras inhibitors in the treatment of cancer
WO2024067857A1 (en) * 2022-09-29 2024-04-04 南京明德新药研发有限公司 Macrocyclic derivative and use thereof

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1990, MACK PRINTING COMPANY, pages: 1289 - 1329
"Roche Molecular Biochemicals", BIOCHEMICA, vol. 2, 1999, pages 34 - 7
BERGE ET AL.: "Pharmaceutical Salts", J. PHARMACEUTICAL SCIENCES, vol. 66, no. 1, 1977, XP002675560, DOI: 10.1002/jps.2600660104
BOER K. ET AL., THERAPEUTIC ADVANCES IN MEDICAL ONCOLOGY, vol. 9, no. 7, 2017, pages 465 - 479
DUKECOHEN ET AL.: "Current Protocols in Immunology", 1992, pages: 1 - 16
FINN ET AL., BREAST CANCER RESEARCH, vol. 11, no. 571190-30-2, 2009, pages 77
GARNER F ET AL., ANTICANCER DRUGS, vol. 26, no. 722533-56-4, 2015, pages 948 - 56
GREENP.G.M. WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY
HAYNES ET AL.: "Commentary: Occurrence of Pharmaceutically Acceptable Anions and Cations in the Cambridge Structural Database", J. PHARMACEUTICAL SCIENCES, vol. 94, no. 10, 2005, XP002593272
KLEIN ET AL., NATURE MED., vol. 3, 1997, pages 402 - 8
MCDONELL ET AL., JOURNAL OF MEDICINAL CHEMISTRY, vol. 58, no. 1365888-06-7, 2015, pages 4883 - 4887
PAGE ET AL., INTL J ONCOLOGY, vol. 3, 1993, pages 473 - 6
TORRES-GUZMAN R ET AL., ONCOTARGET, no. 1231929-97-7, 2017

Similar Documents

Publication Publication Date Title
US12157735B2 (en) Compounds for targeted degradation of BRD9
CN114025844B (en) Protein tyrosine phosphatase inhibitors and methods of use thereof
JP7728367B2 (en) Urea Derivatives That Can Be Used to Treat Cancer
WO2021031952A1 (en) Oxygen-substituted six-membered ring pyrimidine compound, preparation method and medical use thereof
ES2928169T3 (en) 4-(3H-indol-5-yl)-N-(pyridin-2-yl)pyrimidin-2-amine derivatives as protein kinase inhibitors, and method of preparation and medical use thereof
CN119585286A (en) Pyrido[4,3-D]pyrimidine compounds
WO2022228543A1 (en) Bridged ring compound, preparation method therefor, and application thereof in medicine
JP2019178135A (en) Benzoxazepin oxazolidinone compounds and methods of use
JP2013541566A (en) Derivatives of carbazole and carboline and their preparation and therapeutic application
CA3188313A1 (en) Compounds for targeted degradation of ret
EP4288434A1 (en) Selective bcl-xl protac compounds and methods of use
UA119537C2 (en) Use of substituted 2,3-dihydroimidazo[1,2-c]quinazolines for treating lymphomas
JP2018519304A (en) Benzoxazepine oxazolidinone compounds and methods of use
ES2987356T3 (en) Antiproliferation compounds and their uses
JP6912460B2 (en) Quinoxaline and pyridopyrazine derivatives as PI3Kβ inhibitors
TW202222311A (en) Hsp90-binding conjugates and formulations thereof
JP2020504139A (en) Substituted fused heteroaryl compounds as kinase inhibitors and uses thereof
CA3115068A1 (en) Method for preparing and delivering bisantrene formulations
CN111406050B (en) Indoleamine 2,3-dioxygenase inhibitors and uses thereof
TW202110848A (en) A substituted fused bicyclic derivative, a preparation method thereof, and medical applications thereof
WO2025215536A1 (en) Macrocyclic panras inhibitors for the treatment of cancer
CN106573927B (en) Oxepan-2-yl-pyrazol-4-yl-heterocyclyl-carboxamide compounds and methods of use
TW202539634A (en) Panras inhibitors and methods of use thereof
CN103582645B (en) Indane indole analogs
KR20250100539A (en) Pyrido[4,3-d]pyrimidine compounds

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: 25720509

Country of ref document: EP

Kind code of ref document: A1