KRAS G12C inhibitors and methods of using the same

Information

  • Patent Grant
  • 11053226
  • Patent Number
    11,053,226
  • Date Filed
    Monday, November 18, 2019
    4 years ago
  • Date Issued
    Tuesday, July 6, 2021
    3 years ago
Abstract
Provided herein are KRAS G12C inhibitors, composition of the same, and methods of using the same. These inhibitors are useful for treating a number of disorders, including pancreatic, colorectal, and lung cancers.
Description
FIELD OF THE INVENTION

The present invention relates to compounds that inhibit the KRAS G12C protein; methods of treating diseases or conditions, such as cancer, using the compounds; and pharmaceutical compositions containing the compounds.


BACKGROUND

KRAS gene mutations are common in pancreatic cancer, lung adenocarcinoma, colorectal cancer, gall bladder cancer, thyroid cancer, and bile duct cancer. KRAS mutations are also observed in about 25% of patients with NSCLC, and some studies have indicated that KRAS mutations are a negative prognostic factor in patients with NSCLC. Recently, V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations have been found to confer resistance to epidermal growth factor receptor (EGFR) targeted therapies in colorectal cancer; accordingly, the mutational status of KRAS can provide important information prior to the prescription of TKI therapy. Taken together, there is a need for new medical treatments for patients with pancreatic cancer, lung adenocarcinoma, or colorectal cancer, especially those who have been diagnosed to have such cancers characterized by a KRAS mutation, and including those who have progressed after chemotherapy.


The compounds disclosed herein can be in the form of a pharmaceutically acceptable salt. The compounds provided can be formulated into a pharmaceutical formulation comprising a compound disclosed herein and a pharmaceutically acceptable excipient.


Also provided is a method of inhibiting KRAS G12C in a cell, comprising contacting the cell with a compound or composition disclosed herein. Further provided is a method of treating cancer in a subject comprising administering to the subject a therapeutically effective amount of a compound or composition disclosed herein. In some embodiments, the cancer is lung cancer, pancreatic cancer, or colorectal cancer.


SUMMARY

In one aspect of the present invention, the invention provides a compound having a structure of formula (I)




embedded image



wherein


R1 and R1A are independently H, —C1-6alkyl, —C1-6alkylene-OH, —C1-6alkylene-O—C1-6alkyl, or C1-6alkylene-NR10R11;


R2 is independently H, or —C1-6alkyl;


R3 is independently H, —C1-6alkyl, or —C1-6alkyl-OH;


R4 is independently H, or —C1-6alkyl;


R4A is independently H, or —C1-6alkyl;


R4 and R4A, together with the atoms to which they are attached, form a 6- to 12-membered aryl or heteroaryl, a 3- to 8-membered monocyclic or bicyclic cycloalkyl, a 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl, or a 3- to 8-membered monocyclic or bicyclic heterocycloalkyl group, wherein the heteroaryl and heterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl group may include a S═O or SO2;


R4B is independently H, or —C1-6alkyl;


R5 is independently H, halo, —C1-6alkyl, or —C1-6alkyl-CN;


R6 is independently H, halo or —C1-6alkyl, a 6- to 12-membered aryl, a 5- to 12-membered heteroaryl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, a 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl group, wherein the heteroaryl, spiroheterocycloalkyl and heterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl group may include a S═O or SO2;


R7 is independently H, halo or —C1-6alkyl, —O—C1-6alkyl, —S—C1-6alkyl, —NO2, —CN, —CF4, a 6- to 12-membered aryl, 5- to 12-membered heteroaryl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, a 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl group, wherein the heteroaryl, and heterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2;


R8 is independently H, halo or —C1-6alkyl.


R9 is independently selected from H, OH, —C1-6alkyl, —OC1-6alkyl, —C1-6alkyl-O—C1-6alkyl, halo, —O-haloC1-6alkyl, —CN, —C1-6alkyl-C(═O)—NRaRb, —NRaRb, —(NRaRbRc)n, —OSO2Ra, —SO2Ra, —(CH2CH2O)nCH3, —(═O), —C(═O), —C(═O)Ra, —OC(═O)Ra, —C(═O)ORa, —C(═O)C(═O)ORa, —C(═O)NRaRb, —O—SiRaRbRc, —SiRaRbRc, —O-(3- to 12-membered cycloakyl), —O-(3- to 12-membered heterocycloakyl), —C(═O)-6- to 12-membered aryl, —C(═O)-5- to 12-membered heteroaryl, —C(═O)-3- to 12-membered cycloalkenyl, —C(═O)-3- to 12-membered monocyclic or bicyclic cycloalkyl, —C(═O)-3- to 12-membered monocyclic or bicyclic heterocycloalkyl, —C(═O)-5- to 12-membered spirocycloalkyl, —C(═O)-5- to 12-membered spiroheterocycloalkyl, —C1-6alkyl-C(═O)-6- to 12-membered aryl, —C1-6alkyl-C(═O)-5- to 12-membered heteroaryl, a —C1-6alkyl-C(═O)-3- to 12-membered monocyclic or bicyclic cycloalkyl, a —C1-6alkyl-C(═O)-3- to 12-membered monocyclic or bicyclic heterocycloalkyl, —C1-6alkyl-C(═O)-5- to 12-membered monocyclic or bicyclic spirocycloalkyl, —C1-6alkyl-C(═O)-5- to 12-membered spiroheterocycloalkyl group, 6- to 12-membered aryl, 5- to 12-membered heteroaryl, a 3- to 12-membered cycloalkenyl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl group, a 5- to 12-membered monocyclic or bicyclic spirocycloalkyl, or a 5- to 12-membered monocyclic or bicyclic spiroheterocycloalkyl group, wherein the heteroaryl, heterocycloalkyl and spiroheterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2; and still further wherein two adjacent carbon atoms or an adjacent carbon atom and adjacent N atom on a R9 group that includes a cycloalkyl, heterocycloalkyl, aryl, heteroaryl, spirocycloalkyl, or a spiroheterocycloalkyl group may join together to form a 5-6 membered cycloalkyl, heterocycloalkyl, aryl, heteroaryl spirocycloalkyl, or a spiroheterocycloalkyl ring that is unsubstituted or is substituted with 1, 2, or 3 R11 groups;


R10 is independently H, OH, halo, —NH—C1-8alkyl, —N(C1-8alkyl)2, —NH—C0-3alkylene-C6-14 aryl, —NH—C0-3alkylene-C2-14heteroaryl, —NH—C0-3alkylene-C3-14cycloalkyl, —NH—C0-3 alkylene-C2-14heterocycloalkyl, cyano, or C1-6alkylene-amine, —C1-6alkyl, —C(═O)NRaRb, —C(═O)ORa, —C(═O), —C0-3alkylene-C3-14cycloalkyl, —C0-3alkylene-C6-14aryl, —C0-3alkylene-C3-14heteroaryl, —C0-3 alkylene-C3-14cycloalkyl, —C0-3alkylene-C2-14heterocycloalkyl, —C1-6alkoxy, —O—C0-3 alkylene-C6-14aryl, —O—C0-3alkylene-C3-14heteroaryl, —O—C0-3 alkylene-C3-14cycloalkyl, —O—C0-3 alkylene-C2-14heterocycloalkyl, —C(═O)-(3- to 10-membered heterocycloakyl), —C(═O)-6- to 12-membered aryl, —C(═O)-5- to 12-membered heteroaryl, —C(═O)-3- to 12-membered cycloalkenyl, —C(═O)-3- to 12-membered monocyclic or bicyclic cycloalkyl, —C(═O)-3- to 12-membered monocyclic or bicyclic heterocycloalkyl, a —C(═O)-5- to 12-membered bicyclic spirocycloalkyl, a —C(═O)-5- to 12-membered spiroheterocycloalkyl, a 6- to 12-membered aryl, 5- to 12-membered heteroaryl, a 3- to 12-membered cycloalkenyl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl, a 5- to 12-membered monocyclic or bicyclic spirocycloalkyl, or a 5- to 12-membered monocyclic or bicyclic spiroheterocycloalkyl group, wherein the heteroaryl, heterocycloalkyl and spiroheterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2;


or R9 and R10, together with the atoms to which they are attached, form a ring selected from a 6- to 12-membered aryl, a 5- to 12-membered heteroaryl, a 3-8 membered cycloalkyl, or a 3- to 8-membered heterocycloalkyl group, wherein the heteroaryl and heterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein when R9 and R10 form the ring, the ring is unsubstituted or substituted with 1, 2, or 3 R11 groups; further wherein when the ring is a cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl, or heterocycloalkyl group, the ring may further include a C═O group;


R11 is independently selected from H, —OH, halo, cyano, —C1-6alkyl, —C1-6haloalkyl, —C1-6alkyl-OH, —C2-C6 alkenyl, —C2-C6 alkynyl, —(CH2CH2O)nRa, —CSRa, —CS(═O)Ra, —SRa, —SORa, —OSO2Ra, —SO2Ra, —(CH2CH2O)nCH3, —(═O), —C(═O), —O—Ra, —C(═O)Ra, —(CH2)n—NRaRb, —NRaRb; —C(═O)NRaRb, —C(═O)ORa, —CH(═O), —C0-3alkylene-C3-14cycloalkyl, —C0-3alkylene-C2-14heterocycloalkyl, —C0-3alkylene-C6-14aryl, —C0-3alkylene-C3-14heteroaryl, —C1-6alkylene-amine, —C1-6alkoxy, —O—C0-3 alkylene-C6-14aryl, —O—C0-3alkylene-C3-14heteroaryl, —O—C0-3 alkylene-C3-14cycloalkyl, —O—C0-3 alkylene-C2-14heterocycloalkyl, —NH—C1-8alkyl, —N(C1-8alkyl)2, —NH—C0-3alkylene-C6-14aryl, —NH—C0-3alkylene-C2-14heteroaryl, —NH—C0-3alkylene-C3-14cycloalkyl, —NH—C0-3 alkylene-C2-14heterocycloalkyl, —C1-6alkylene-amine-C(═O)-(3- to 10-membered heterocycloakyl), —C(═O)-6- to 12-membered aryl, —C(═O)-5- to 12-membered heteroaryl, —C(═O)-3- to 12-membered cycloalkenyl, —C(═O)-3- to 12-membered monocyclic or bicyclic cycloalkyl, —C(═O)-3- to 12-membered monocyclic or bicyclic heterocycloalkyl, a —C(═O)-5- to 12-membered bicyclic spirocycloalkyl, a —C(═O)-5- to 12-membered spiroheterocycloalkyl, a 6- to 12-membered aryl, 5- to 12-membered heteroaryl, a 3- to 12-membered cycloalkenyl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl group, a 5- to 12-membered monocyclic or bicyclic spirocycloalkyl, or a 5- to 12-membered monocyclic or bicyclic spiroheterocycloalkyl group, wherein the heteroaryl, heterocycloalkyl and spiroheterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2;


wherein the heteroaryl, spiroheterocycloalkyl and heterocycloalkyl groups of any of the R4, R4A, R6, R7, R9, R10, and R11 substituents have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2;


wherein the —C1-6alkyl, —C2-6alkenyl, —C2-6alkynyl and the —OC1-6alkyl of any of the R1, R2, R3, R4, R4a, R4B, R5, R6, R7, R8, R9, R10 and R11 substituents is unsubstituted or substituted by 1, 2 or 3 R12 substituents independently selected from OH, —OC1-6alkyl, —C1-6alkyl-O—C1-6alkyl, halo, —O-haloC1-6alkyl, —CN, —NRaRb, —(NRaRbRc)n, —OSO2Ra, —SO2Ra, (CH2CH2O)nCH3, —(═O), —C(═O), —C(═O)Ra, —OC(═O)Ra, —C(═O)ORa, —C(═O)NRaRb, —O—SiRaRbRc, —SiRaRbRc, —O-(3- to 10-membered heterocycloalkyl), 6- to 12-membered aryl or heteroaryl, 3- to 12-membered cycloalkenyl, 3- to 12-membered monocyclic or bicyclic cycloalkyl, 3- to 12-membered monocyclic or bicyclic heterocycloalkyl or 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl group, wherein the heteroaryl, spiroheterocycloalkyl and heterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2;


wherein the aryl, heteroaryl, cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups of any of the R4, R4A, R4B, R6, R7, R9, R10, R11, and R12 substituents can be unsubstituted or substituted with 1, 2, 3 or 4 R13 substituents independently selected from OH, halo, —NRcRd, —ORa, —C1-6alkyl, —OC1-6alkyl, —C1-6alkyl-OH, —C1-6alkyl-O—C1-6alkyl, C1-6haloalkyl, —O-haloC1-6alkyl, —SO2Rc, —CN, —C1-6alkyl-NRcRd, —C(═O)NRcRd, —C(═O)Rc, —OC(═O)Ra, —C(═O)ORc, —C1-6alkyl-6- to 12-membered aryl, —C1-6alkyl-5- to 12-membered heteroaryl, —C1-6alkyl-3- to 12-membered cycloalkenyl, —C1-6alkyl-3- to 12-membered monocyclic or bicyclic cycloalkyl, —C1-6alkyl-3- to 12-membered monocyclic or bicyclic heterocycloalkyl, 6- to 12-membered aryl, 5- to 12-membered heteroaryl, 3- to 12-membered cycloalkenyl, 3- to 12-membered monocyclic or bicyclic cycloalkyl, 3- to 12-membered monocyclic or bicyclic heterocycloalkyl, 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl, —C(═O)-5- to 12-membered bicyclic spirocycloalkyl, —C(═O)-5- to 12-membered spiroheterocycloalkyl group, wherein the heteroaryl, spiroheterocycloalkyl and heterocycloalkyl groups of R13 have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl or heterocycloalkyl group of R13 may include a C═O group, and further wherein the heterocycloalkyl and spiroheterocycloalkyl groups may include a S═O or SO2; wherein the alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, spirocycloalkyl, or spiroheterocycloalkyl groups of R13 may be unsubstituted or substituted by 1, 2 or 3 Ra substituents;


wherein each Ra, Rb, Rc and Rd is independently hydrogen, OH, O, cyano, —C1-6alkyl, —C1-6alkyl(halo)n, —(CH2CH2O)nCH3, —O—C1-6alkyl, —NO2, —NR14R14, —C1-6alkyl-NR14R14, phenyl, —C1-6alkyl-C(═O)OH, —C1-6alkyl-C(═O)—O—C1-6alkyl, —C1-6alkyl-C(═O)—NH—C1-6alkyl-6- to 12-membered aryl, —C1-6alkyl-C(═O)—NH—6- to 12-membered aryl, —C1-6alkyl-3- to 12-membered cycloalkyl, —C1-6alkyl-3- to 12-membered heterocycloalkyl, —C1-6alkyl-6- to 12-membered aryl, —C1-6alkyl-5- to 12-membered heteroaryl, —C1-6alkyl-O-3- to 12-membered cycloalkyl, —C1-6alkyl-O-3- to 12-membered heterocycloalkyl, —C1-6alkyl-O-6- to 12-membered aryl, —C1-6alkyl-O-5- to 12-membered heteroaryl, 6- to 12-membered aryl, 5- to 12-membered heteroaryl, 3- to 12-membered monocyclic or bicyclic cycloalkyl, 3- to 12-membered monocyclic or bicyclic heterocycloalkyl, or 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl group, wherein the heteroaryl group, heterocycloalkyl or spiroheterocycloalkyl group of Ra, Rb, Rc, and Rd or the heterocycloalkyl group of the —C1-6alkyl-heterocycloalkyl group of Ra, Rb, Rc, and Rd has from 1, 2, 3, or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups of Ra, Rb, Rc, and Rd and the heterocycloalkyl group of the —C1-6alkyl-heterocycloalkyl groups of Ra, Rb, Rc, and Rd may include a double bond, and further wherein the cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups of Ra, Rb, Rc, and Rd and the heterocycloalkyl group of the —C1-6alkyl-heterocycloalkyl groups of Ra, Rb, Rc, and Rd may contain a C═O group; and


the alkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, spirocycloalkyl and spiroheterocycloalkyl groups of Ra, Rb, Rc, and Rd or the heterocycloalkyl groups of the —C1-6alkyl-heterocycloalkyl groups of Ra, Rb, Rc, and Rd can be unsubstituted or substituted with from 1, 2, 3, or 4 R14 substituents, wherein each R14 is independently selected from H, OH, halo, —C1-6alkyl, —CN, —C1-6alkyl(halo)n, —N(CH3)2, —C(O)NH2, —C(O)N(CH3)2, —C1-6haloalkyl, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, —O—C1-6alkyl or —C1-6alkyl-O—C1-6alkyl, —C1-6alkyl-N(CH3)2, —C(═O)-3- to 12-membered cycloalkyl, 5- to 12-membered heteroaryl, —C1-6alkyl-6- to 12-membered aryl or -6- to 12-membered aryl;


wherein n is 0, 1, 2, or 3; or


a stereoisomer thereof, an atropisomer thereof, a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable salt of the stereoisomer thereof, or a pharmaceutically acceptable salt of the atropisomer thereof.


In another aspect of the present invention, the present invention comprises a compound having a structure of formula (Ia)




embedded image



or


a stereoisomer thereof, an atropisomer thereof, a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable salt of the stereoisomer thereof, or a pharmaceutically acceptable salt of the atropisomer thereof.


One aspect of the present invention provides various compounds, stereoisomers, atropisomers, pharmaceutically acceptable salts, pharmaceutically acceptable salts of the stereoisomers, and pharmaceutically acceptable salts of the atropisomers as described in the embodiments set forth below.


Another aspect of the present invention provides a pharmaceutical composition that includes the compound of any of the embodiments or the pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or diluent.


Another aspect of the present invention provides a method of treating cancer. Such methods include: administering to a patient in need thereof a therapeutically effective amount of the compound of any of the embodiments or a pharmaceutically acceptable salt thereof. In some such methods, the cancer is a hematologic malignancy. In some such methods, the cancer is selected from the group consisting of breast cancer, colorectal cancer, skin cancer, melanoma, ovarian cancer, kidney cancer, lung cancer, non-small cell lung cancer, lymphoma, non-Hodgkin's lymphoma, myeloma, multiple myeloma, leukemia, and acute myelogenous leukemia. In some other such methods, the cancer is multiple myeloma. In some other such methods, the cancer is acute myelogenous leukemia. In some other such methods, the cancer is non-Hodgkin's lymphoma. In another aspect, the method further includes administering to a patient in need thereof a therapeutically effective amount of an additional pharmaceutically active compound. For example, in some such methods the additional pharmaceutically active compound is carfilzomib. In others, the additional pharmaceutically active compound is venetoclax. In still other such methods, the additional pharmaceutically active compound is cytarabine. In still other such methods, the additional pharmaceutically active compound is daratumumab. In still other such methods, the additional pharmaceutically active compound is an MCl-1 inhibitor. In still other such methods, the MCl-1 inhibitor is AMG-176. In still other such methods, the additional pharmaceutically active compound is an IMiD.


Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Methods and materials are described herein for use in the present disclosure; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.


Other features and advantages of the disclosure will be apparent from the following detailed description and figures, and from the Claims.







DETAILED DESCRIPTION
Definitions
Abbreviations: The Following Abbreviations May be Used Herein














AcOH
acetic acid


aq or aq.
aqueous


BOC or Boc
tert-butyloxycarbonyl


DCM
dichloromethane


DIPEA or Hunig's Base
N,N-diisopropylethylamine


DMAP
4-dimethylaminopyridine


DME
1,2-dimethoxyethane


DMF
N,N-dimethylformamide


DMSO
dimethyl sulfoxide


Dppf, DPPF or dppf
1,1′-bis(diphenylphosphino)ferrocene


eq or eq. or equiv.
equivalent


ESI or ES
electrospray ionization


Et
ethyl


Et2O
diethyl ether


EtOAc
ethyl acetate


g
gram(s)


h or hr
hour(s)


HPLC
high pressure liquid chromatography


iPr
isopropyl


iPr2NEt or DIPEA
N-ethyl diisopropylamine (Hünig's base)


KHMDS
potassium hexamethyldisilazide


KOAc
potassium acetate


LC MS, LCMS,
liquid chromatography mass spectroscopy


LC-MS or LC/MS



LHMDS or LiHMDS
lithium hexamethyldisilazide


m/z
mass divided by charge


Me
methyl


MeCN
acetonitrile


MeOH
methanol


mg
milligrams


min
minutes


mL
milliliters


MS
mass spectra


NaHMDS
sodium hexamethyldisilazide


NBS
N-bromosuccinimide


n-BuLi
n-butyllithium


NCS
N-chlorosuccinimide


NMR
nuclear magnetic resonance


Pd2(dba)3
tris(dibenzylideneacetone)dipalladium (0)


Pd(dppf)Cl2•DCM,
[1,1′-


Pd(dppf)Cl2
bis(diphenylphosphino)ferrocene]



dichloropalladium (II), complex with



dichloromethane


Pd(PPh3)4
tetrakis(triphenylphosphine)palladium (0)


Ph
phenyl


PR or PG or Prot. group
protecting group


rbf
round-bottom flask


RP-HPLC
reverse phase high pressure liquid



chromatography


RT or rt or r.t.
room temperature


sat. or sat'd
saturated


SFC
supercritical fluid chromatography


TBAF
tetra-n-butylammonium fluoride


TEA or Et3N
triethylamine


TFA
trifluoroacetic acid


THF
tetrahydrofuran


UV
ultraviolet









The use of the terms “a,” “an,” “the,” and similar referents in the context of describing the invention (especially in the context of the claims) are to be construed to cover both the singular and the plural, unless otherwise indicated. Recitation of ranges of values herein merely are intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended to better illustrate the invention and is not a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.


As used herein, the term “alkyl” refers to straight chained and branched C1-C8 hydrocarbon groups, including but not limited to, methyl, ethyl, n-propyl, i-propyl, n-butyl, sec-butyl, t-butyl, n-pentyl, 2-methylbutyl, 3-methylbutyl, 2,2-dimethylpropyl, n-hexyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,2-dimethylbutyl, 2,3-dimethylbutyl, 3,3-dimethylbutyl, and 2-ethylbutyl. The term Cm-n means the alkyl group has “m” to “n” carbon atoms. The term “alkylene” refers to an alkyl group having a substituent. An alkyl (e.g., methyl), or alkylene (e.g., —CH2—), group can be substituted with one or more, and typically one to three, of independently selected, for example, halo, trifluoromethyl, trifluoromethoxy, hydroxy, alkoxy, nitro, cyano, alkylamino, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, —NC, amino, —CO2H, —CO2C1-C6alkyl, —OCOC1-C6alkyl, C3-C10 cycloalkyl, C3-C10 heterocycloalkyl, C5-C10aryl, and C5-C10 heteroaryl. The term “haloalkyl” specifically refers to an alkyl group wherein at least one, e.g., one to six, or all of the hydrogens of the alkyl group are substituted with halo atoms.


The terms “alkenyl” and “alkynyl” indicate an alkyl group that further includes a double bond or a triple bond, respectively.


As used herein, the term “halo” refers to fluoro, chloro, bromo, and iodo. The term “alkoxy” is defined as —OR, wherein R is alkyl.


As used herein, the term “amino” or “amine” interchangeably refers to a —NR2 group, wherein each R is, e.g., H or a substituent. In some embodiments, the amino group is further substituted to form an ammonium ion, e.g., NR3+. Ammonium moieties are specifically included in the definition of “amino” or “amine.” Substituents can be, for example, an alkyl, alkoxy, cycloalkyl, heterocycloalkyl, amide, or carboxylate. An R group may be further substituted, for example, with one or more, e.g., one to four, groups selected from halo, cyano, alkenyl, alkynyl, alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, urea, carbonyl, carboxylate, amine, and amide. An “amide” or “amido” group interchangeably refers to a group similar to an amine or amino group but further including a C(O), e.g., —C(O)NR2. Some contemplated amino or amido groups (some with optional alkylene groups, e.g., alkylene-amino, or alkylene-amido) include CH2NH2, CH(CH3)NH2, CH(CH3)2NH2, CH2CH2NH2, CH2CH2N(CH3)2, CH2NHCH3, C(O)NHCH3, C(O)N(CH3)2, CH2C(O)NHphenyl, CH2NHC(O)CH3, CH2NHCH2CH2OH, CH2NHCH2CO2H, CH2NH(CH3)CH2CO2CH3, CH2NHCH2CH2OCH3, CH2NH(CH3)CH2CH2OCH3, CH2NH(CH3)CH2C(O)N(CH3)2, CH2NH(CH3)CH2C(O)NHCH3, CH2CH2CCH, CH2NMe2, CH2NH(CH3)CH2CH2OH, CH2NH(CH3)CH2CH2F, CH2N+(CH3)3, CH2NHCH2CHF2, CH2NHCH2CH3,




embedded image


embedded image


As used herein, the term “aryl” refers to a C6-14 monocyclic or polycyclic aromatic group, preferably a C6-10 monocyclic or bicyclic aromatic group, or C10-14 polycyclic aromatic group. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, fluorenyl, azulenyl, anthryl, phenanthryl, pyrenyl, biphenyl, and terphenyl. Aryl also refers to C10-14 bicyclic and tricyclic carbon rings, where one ring is aromatic and the others are saturated, partially unsaturated, or aromatic, and one of the rings can be a heterocycloalkyl ring. Examples include, but are not limited to dihydronaphthyl, indenyl, indanyl, 1,2,3,4-tetrahydroisoquinoline, or tetrahydronaphthyl (tetralinyl). Unless otherwise indicated, an aryl group can be unsubstituted or substituted with one or more, and in particular one to four, groups independently selected from, for example, halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, —CF3, —OCF3, —NO2, —CN, —NC, —OH, alkoxy, amino, —CO2H, —CO2C1-C6alkyl, —OCOC1-C6alkyl, —C3-C10 cycloalkyl, —C3-C10 heterocycloalkyl, —C5-C10aryl, and —C5-C10 heteroaryl.


As used herein, the term “cycloalkyl” refers to a monocyclic or polycyclic non-aromatic carbocyclic ring, where the polycyclic ring can be fused, bridged, or spiro. The carbocyclic ring can have 3 to 10 carbon ring atoms. Contemplated carbocyclic rings include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and cyclononyl.


As used herein, the term “heterocycloalkyl” means a monocyclic or polycyclic (e.g., bicyclic), saturated or partially unsaturated, ring system containing 3 or more (e.g., 3 to 12, 4 to 10, 4 to 8, or 5 to 7) total atoms, of which one to five (e.g., 1, 2, 3, 4, or 5) of the atoms are independently selected from nitrogen, oxygen, and sulfur. Polycyclic heterocycloalkyl groups are those in which the heterocycloalkyl ring is bonded to the rest of the molecule. Nonlimiting examples of heterocycloalkyl groups include azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, dihydropyrrolyl, morpholinyl, thiomorpholinyl, dihydropyridinyl, oxacycloheptyl, dioxacycloheptyl, thiacycloheptyl, and diazacycloheptyl. Heterocycloalkyl groups also include groups where a heterocycloalkyl ring is fused to another ring that may be a cycloalkyl ring, a heterocycloalkyl ring, an aryl ring, or a heteroaryl ring. Examples include, but are not limited to, 1,2,3,4-tetrahydroisoquinoline, 5,6,7,8-tetrahydro-1,7-naphthyridine, decahydroisoquinoline, and octahydro-1H-2λ2-2,6-naphthyridine.


Unless otherwise indicated, a cycloalkyl or heterocycloalkyl group can be unsubstituted or substituted with one or more, and in particular one to four, groups. Some contemplated substituents include halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, —OCF3, —NO2, —CN, —NC, —OH, —O—C1-6 alkyl, alkoxy, amino, —CO2H, —CO2C1-C6alkyl, —OCOC1-C8alkyl, C3-C10 cycloalkyl, C3-C10 heterocycloalkyl, C5-C10aryl, and C5-C10 heteroaryl.


As used herein, the term “heteroaryl” refers to a monocyclic or polycyclic ring system (for example, bicyclic) containing one to three aromatic rings and wherein at least one of the aromatic rings contains one to four (e.g., 1, 2, 3, or 4) heteroatoms selected from nitrogen, oxygen, and sulfur. In certain embodiments, the heteroaryl group has from 5 to 20, from 5 to 15, from 5 to 10, or from 5 to 7 atoms. Heteroaryl also refers to C9-14 bicyclic and tricyclic rings, where one ring is aromatic and the others are saturated, partially unsaturated, or aromatic. Examples of heteroaryl groups include, but are not limited to, furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl, tetrazolyl, triazinyl, triazolyl, benzofuranyl, benzimidazolyl, benzoisoxazolyl, benzopyranyl, benzothiadiazolyl, benzothiazolyl, benzothienyl, benzothiophenyl, benzotriazolyl, benzoxazolyl, furopyridyl, imidazopyridinyl, imidazothiazolyl, indolizinyl, indolyl, indazolyl, isobenzofuranyl, isobenzothienyl, isoindolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxazolopyridinyl, phthalazinyl, pteridinyl, purinyl, pyridopyridyl, pyrrolopyridyl, quinolinyl, quinoxalinyl, quiazolinyl, thiadiazolopyrimidyl, and thienopyridyl. Unless otherwise indicated, a heteroaryl group can be unsubstituted or substituted with one or more, and in particular one to four or one or two, substituents. Contemplated substituents include halo, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, —OCF3, —NO2, —CN, —NC, —OH, alkoxy, amino, —CO2H, —CO2C1-C6alkyl, —OCOC1-C6alkyl, C3-C10 cycloalkyl, C3-C10 heterocycloalkyl, C5-C10aryl, and C5-C10 heteroaryl.


As used herein, the term Boc refers to the structure




embedded image


As used herein, the term Cbz refers to the structure




embedded image


As used herein, the term Bn refers to the structure




embedded image


As used herein, the term trifluoroacetamide refers to the structure




embedded image


As used herein, the term trityl refers to the structure




embedded image


As used herein, the term tosyl refers to the structure




embedded image


As used herein, the term Troc refers to the structure




embedded image


As used herein, the term Teoc refers to the structure




embedded image


As used herein, the term Alloc refers to the structure




embedded image


As used herein, the term Fmoc refers to the structure




embedded image


Compounds of the Disclosure

The compounds disclosed herein include all pharmaceutically acceptable isotopically-labeled compounds wherein one or more atoms of the compounds disclosed herein are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2H, 3H, 11C, 13C, 14C, 13N, 15N, 15O, 17O, 18O, 31P, 32P, 35S, 18F, 36Cl, 123I, and 125I, respectively. These radiolabelled compounds could be useful to help determine or measure the effectiveness of the compounds, by characterizing, for example, the site or mode of action, or binding affinity to pharmacologically important site of action. Certain isotopically-labeled compounds of the disclosure, for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.


Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence are preferred in some circumstances.


Substitution with positron emitting isotopes, such as 11C, 18F, 15O and 13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds of structure (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Preparations and Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.


Isotopically-labeled compounds as disclosed herein can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying examples and schemes using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.


Certain of the compounds as disclosed herein may exist as stereoisomers (i.e., isomers that differ only in the spatial arrangement of atoms) including optical isomers and conformational isomers (or conformers). The compounds disclosed herein include all stereoisomers, both as pure individual stereoisomer preparations and enriched preparations of each, and both the racemic mixtures of such stereoisomers as well as the individual diastereomers and enantiomers that may be separated according to methods that are known to those skilled in the art. Additionally, the compounds disclosed herein include all tautomeric forms of the compounds.


Certain of the compounds disclosed herein may exist as atropisomers, which are conformational stereoisomers that occur when rotation about a single bond in the molecule is prevented, or greatly slowed, as a result of steric interactions with other parts of the molecule. The compounds disclosed herein include all atropisomers, both as pure individual atropisomer preparations, enriched preparations of each, or a non-specific mixture of each. Where the rotational barrier about the single bond is high enough, and interconversion between conformations is slow enough, separation and isolation of the isomeric species may be permitted.


EMBODIMENTS
Embodiment 1

In one embodiment of the present invention, the present invention comprises a compound having formula (I)




embedded image



wherein


R1 and R1A are independently H, —C1-6alkyl, —C1-6alkylene-OH, —C1-6alkylene-O—C1-6alkyl, or C1-6alkylene-NR10R11;


R2 is independently H, or —C1-6alkyl;


R3 is independently H, —C1-6alkyl, or —C1-6alkyl-OH;


R4 is independently H, or —C1-6alkyl;


R4A is independently H, or —C1-6alkyl;


R4 and R4A, together with the atoms to which they are attached, form a 6- to 12-membered aryl or heteroaryl, a 3- to 8-membered monocyclic or bicyclic cycloalkyl, a 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl, or a 3- to 8-membered monocyclic or bicyclic heterocycloalkyl group, wherein the heteroaryl and heterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl group may include a S═O or SO2;


R4B is independently H, or —C1-6alkyl;


R5 is independently H, halo, —C1-6alkyl, or —C1-6alkyl-CN;


R6 is independently H, halo or —C1-6alkyl, a 6- to 12-membered aryl, a 5- to 12-membered heteroaryl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, a 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl group, wherein the heteroaryl, spiroheterocycloalkyl and heterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl group may include a S═O or SO2;


R7 is independently H, halo or —C1-6alkyl, —O—C1-6alkyl, —S—C1-6alkyl, —NO2, —CN, —CF3, a 6- to 12-membered aryl, 5- to 12-membered heteroaryl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, a 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl group, wherein the heteroaryl, and heterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2;


R8 is independently H, halo or —C1-6alkyl;


R9 is independently selected from H, OH, —C1-6alkyl, —OC1-6alkyl, —C1-6alkyl-O—C1-6alkyl, halo, —O-haloC1-6alkyl, —CN, —C1-6alkyl-C(═O)—NRaRb, —NRaRb, —(NRaRb)n, —OSO2Ra, —SO2Ra, —(CH2CH2O)nCH3, —(═O), —C(═O), —C(═O)Ra, —OC(═O)Ra, —C(═O)ORa, —C(═O)C(═O)ORa, —C(═O)NRaRb, —O—SiRaRbRc, —SiRaRbRc, —O-(3- to 12-membered cycloakyl), —O-(3- to 12-membered heterocycloakyl), —C(═O)-6- to 12-membered aryl, —C(═O)-5- to 12-membered heteroaryl, —C(═O)-3- to 12-membered cycloalkenyl, —C(═O)-3- to 12-membered monocyclic or bicyclic cycloalkyl, —C(═O)-3- to 12-membered monocyclic or bicyclic heterocycloalkyl, —C(═O)-5- to 12-membered spirocycloalkyl, —C(═O)-5- to 12-membered spiroheterocycloalkyl, —C1-6alkyl-C(═O)-6- to 12-membered aryl, —C1-6alkyl-C(═O)-5- to 12-membered heteroaryl, a —C1-6alkyl-C(═O)-3- to 12-membered monocyclic or bicyclic cycloalkyl, a —C1-6alkyl-C(═O)-3- to 12-membered monocyclic or bicyclic heterocycloalkyl, —C1-6alkyl-C(═O)-5- to 12-membered monocyclic or bicyclic spirocycloalkyl, —C1-6alkyl-C(═O)-5- to 12-membered spiroheterocycloalkyl group, 6- to 12-membered aryl, 5- to 12-membered heteroaryl, a 3- to 12-membered cycloalkenyl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl group, a 5- to 12-membered monocyclic or bicyclic spirocycloalkyl, or a 5- to 12-membered monocyclic or bicyclic spiroheterocycloalkyl group, wherein the heteroaryl, heterocycloalkyl and spiroheterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2; and still further wherein two adjacent carbon atoms or an adjacent carbon atom and adjacent N atom on a R9 group that includes a cycloalkyl, heterocycloalkyl, aryl, heteroaryl, spirocycloalkyl, or a spiroheterocycloalkyl group may join together to form a 5-6 membered cycloalkyl, heterocycloalkyl, aryl, heteroaryl spirocycloalkyl, or a spiroheterocycloalkyl ring that is unsubstituted or is substituted with 1, 2, or 3 R11 groups;


R10 is independently H, halo, —NH—C1-8alkyl, —N(C1-8alkyl)2, —NH—C0-3alkylene-C6-14aryl, —NH—C0-3alkylene-C2-14heteroaryl, —NH—C0-3alkylene-C3-14cycloalkyl, —NH—C0-3 alkylene-C2-14heterocycloalkyl, cyano, or C1-6alkylene-amine, —C1-6alkyl, —C(═O)NRaRb, —C(═O)ORa, —C(═O), —C0-3alkylene-C3-14cycloalkyl, —C0-3alkylene-C6-14aryl, —C0-3alkylene-C3-14heteroaryl, —C0-3 alkylene-C3-14cycloalkyl, —C0-3alkylene-C2-14heterocycloalkyl, —C1-6alkoxy, —O—C0-3 alkylene-C6-14aryl, —O—C0-3alkylene-C3-14heteroaryl, —O—C0-3 alkylene-C3-14cycloalkyl, —O—C0-3 alkylene-C2-14heterocycloalkyl, —C(═O)-(3- to 10-membered heterocycloakyl), —C(═O)-6- to 12-membered aryl, —C(═O)-5- to 12-membered heteroaryl, —C(═O)-3- to 12-membered cycloalkenyl, —C(═O)-3- to 12-membered monocyclic or bicyclic cycloalkyl, —C(═O)-3- to 12-membered monocyclic or bicyclic heterocycloalkyl, a —C(═O)-5- to 12-membered bicyclic spirocycloalkyl, a —C(═O)-5- to 12-membered spiroheterocycloalkyl, a 6- to 12-membered aryl, 5- to 12-membered heteroaryl, a 3- to 12-membered cycloalkenyl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl, a 5- to 12-membered monocyclic or bicyclic spirocycloalkyl, or a 5- to 12-membered monocyclic or bicyclic spiroheterocycloalkyl group, wherein the heteroaryl, heterocycloalkyl and spiroheterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2;


or R9 and R10, together with the atoms to which they are attached, form a ring selected from a 6- to 12-membered aryl, a 5- to 12-membered heteroaryl, a 3-8 membered cycloalkyl, or a 3- to 8-membered heterocycloalkyl group, wherein the heteroaryl and heterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein when R9 and R10 form the ring, the ring is unsubstituted or substituted with 1, 2, or 3 R11 groups; further wherein when the ring is a cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl, or heterocycloalkyl group, the ring may further include a C═O group;


R11 is independently selected from H, —OH, halo, cyano, —C1-6alkyl, —C1-6haloalkyl, —C1-6alkyl-OH, —C2-C6 alkenyl, —C2-C6 alkynyl, —(CH2CH2O)nRa, —CSRa, —CS(═O)Ra, —SRa, —SORa, —OSO2Ra, —SO2Ra, —(CH2CH2O)nCH3, —(═O), —C(═O), —O—Ra, —C(═O)Ra, —(CH2)n—NRaRb, NRaRb; —C(═O)NRaRb, —C(═O)ORa, —CH(═O), —C0-3alkylene-C3-14cycloalkyl, —C0-3alkylene-C2-14heterocycloalkyl, —C0-3alkylene-C6-14aryl, —C0-3alkylene-C3-14heteroaryl, —C1-6alkylene-amine, —C1-6alkoxy, —O—C0-3 alkylene-C6-14aryl, —O—C0-3alkylene-C3-14heteroaryl, —O—C0-3 alkylene-C3-14cycloalkyl, —O—C0-3 alkylene-C2-14heterocycloalkyl, —NH—C1-8alkyl, —N(C1-8alkyl)2, —NH—C0-3alkylene-C6-14aryl, —NH—C0-3alkylene-C2-14heteroaryl, —NH—C0-3alkylene-C3-14cycloalkyl, —NH—C0-3 alkylene-C2-14heterocycloalkyl, —C1-6alkylene-amine-C(═O)-(3- to 10-membered heterocycloakyl), —C(═O)-6- to 12-membered aryl, —C(═O)-5- to 12-membered heteroaryl, —C(═O)-3- to 12-membered cycloalkenyl, —C(═O)-3- to 12-membered monocyclic or bicyclic cycloalkyl, —C(═O)-3- to 12-membered monocyclic or bicyclic heterocycloalkyl, a —C(═O)-5- to 12-membered bicyclic spirocycloalkyl, a —C(═O)-5- to 12-membered spiroheterocycloalkyl, a 6- to 12-membered aryl, 5- to 12-membered heteroaryl, a 3- to 12-membered cycloalkenyl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl group, a 5- to 12-membered monocyclic or bicyclic spirocycloalkyl, or a 5- to 12-membered monocyclic or bicyclic spiroheterocycloalkyl group, wherein the heteroaryl, heterocycloalkyl and spiroheterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2;


wherein the heteroaryl, spiroheterocycloalkyl and heterocycloalkyl groups of any of the R4, R4A, R6, R7, R9, R10, and R11 substituents have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2;


wherein the —C1-6alkyl, —C2-6alkenyl, —C2-6alkynyl and the —OC1-6alkyl of any of the R1, R2, R3, R4, R4a, R4B, R5, R6, R7, R8, R9, R10 and R11 substituents is unsubstituted or substituted by 1, 2 or 3 R12 substituents independently selected from OH, —OC1-6alkyl, —C1-6alkyl-O—C1-6alkyl, halo, —O-haloC1-6alkyl, —CN, —NRaRb, —(NRaRbRc)n, —OSO2Ra, —SO2Ra, (CH2CH2O)nCH3, —(═O), —C(═O), —C(═O)Ra, —OC(═O)Ra, —C(═O)ORa, —C(═O)NRaRb, —O—SiRaRbRc, —SiRaRbRc, —O-(3- to 10-membered heterocycloalkyl), 6- to 12-membered aryl or heteroaryl, 3- to 12-membered cycloalkenyl, 3- to 12-membered monocyclic or bicyclic cycloalkyl, 3- to 12-membered monocyclic or bicyclic heterocycloalkyl or 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl group, wherein the heteroaryl, spiroheterocycloalkyl and heterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2;


wherein the aryl, heteroaryl, cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups of any of the R4, R4A, R4B, R6, R7, R9, R10, R11, and R12 substituents can be unsubstituted or substituted with 1, 2, 3 or 4 R13 substituents independently selected from OH, halo, —NRcRd, —ORa, —C1-6alkyl, —OC1-6alkyl, —C1-6alkyl-OH, —C1-6alkyl-O—C1-6alkyl, C1-6haloalkyl, —O-haloC1-6alkyl, —SO2Rc, —CN, —C1-6alkyl-NRcRd, —C(═O)NRcRd, —C(═O)Rc, —OC(═O)Ra, —C(═O)ORc, —C1-6alkyl-6- to 12-membered aryl, —C1-6alkyl-5- to 12-membered heteroaryl, —C1-6alkyl-3- to 12-membered cycloalkenyl, —C1-6alkyl-3- to 12-membered monocyclic or bicyclic cycloalkyl, —C1-6alkyl-3- to 12-membered monocyclic or bicyclic heterocycloalkyl, 6- to 12-membered aryl, 5- to 12-membered heteroaryl, 3- to 12-membered cycloalkenyl, 3- to 12-membered monocyclic or bicyclic cycloalkyl, 3- to 12-membered monocyclic or bicyclic heterocycloalkyl, 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl, —C(═O)-5- to 12-membered bicyclic spirocycloalkyl, —C(═O)-5- to 12-membered spiroheterocycloalkyl group, wherein the heteroaryl, spiroheterocycloalkyl and heterocycloalkyl groups of R13 have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl or heterocycloalkyl group of R13 may include a C═O group, and further wherein the heterocycloalkyl and spiroheterocycloalkyl groups may include a S═O or SO2; wherein the alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, spirocycloalkyl, or spiroheterocycloalkyl groups of R13 may be unsubstituted or substituted by 1, 2 or 3 Ra substituents;


wherein each Ra, Rb, Rc and Rd is independently hydrogen, OH, O, cyano, —C1-6alkyl, —C1-6alkyl(halo)n, —(CH2CH2O)nCH3, —O—C1-6alkyl, —NO2, —NR14R14, —C1-6alkyl-NR14R14, phenyl, —C1-6alkyl-C(═O)OH, —C1-6alkyl-C(═O)—O—C1-6alkyl, —C1-6alkyl-C(═O)—NH—C1-6alkyl-6- to 12-membered aryl, —C1-6alkyl-C(═O)—NH—6- to 12-membered aryl, —C1-6alkyl-3- to 12-membered cycloalkyl, —C1-6alkyl-3- to 12-membered heterocycloalkyl, —C1-6alkyl-6- to 12-membered aryl, —C1-6alkyl-5- to 12-membered heteroaryl, —C1-6alkyl-O-3- to 12-membered cycloalkyl, —C1-6alkyl-O-3- to 12-membered heterocycloalkyl, —C1-6alkyl-O-6- to 12-membered aryl, —C1-6alkyl-O-5- to 12-membered heteroaryl, 6- to 12-membered aryl, 5- to 12-membered heteroaryl, 3- to 12-membered monocyclic or bicyclic cycloalkyl, 3- to 12-membered monocyclic or bicyclic heterocycloalkyl, or 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl group, wherein the heteroaryl group, heterocycloalkyl or spiroheterocycloalkyl group of Ra, Rb, Rc, and Rd or the heterocycloalkyl group of the —C1-6alkyl-heterocycloalkyl group of Ra, Rb, Rc, and Rd has from 1, 2, 3, or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups of Ra, Rb, Rc, and Rd and the heterocycloalkyl group of the —C1-6alkyl-heterocycloalkyl groups of Ra, Rb, Rc, and Rd may include a double bond, and further wherein the cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups of Ra, Rb, Rc, and Rd and the heterocycloalkyl group of the —C1-6alkyl-heterocycloalkyl groups of Ra, Rb, Rc, and Rd may contain a C═O group; and


the alkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, spirocycloalkyl and spiroheterocycloalkyl groups of Ra, Rb, Rc, and Rd or the heterocycloalkyl groups of the —C1-6alkyl-heterocycloalkyl groups of Ra, Rb, Rc, and Rd can be unsubstituted or substituted with from 1, 2, 3, or 4 R14 substituents, wherein each R14 is independently selected from H, OH, halo, —C1-6alkyl, —CN, —C1-6alkyl(halo)n, —N(CH3)2, —C(O)NH2, —C(O)N(CH3)2, —C1-6haloalkyl, —C(═O)CH3, —C(═O)OH, —C(═O)OCH3, —O—C1-6alkyl or —C1-6alkyl-O—C1-6alkyl, —C1-6alkyl-N(CH3)2, —C(═O)-3- to 12-membered cycloalkyl, 5- to 12-membered heteroaryl, —C1-6alkyl-6- to 12-membered aryl or -6- to 12-membered aryl;


wherein n is 0, 1, 2, or 3; or


a stereoisomer thereof, an atropisomer thereof, a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable salt of the stereoisomer thereof, or a pharmaceutically acceptable salt of the atropisomer thereof.


Embodiment 2

In another embodiment of the present invention, the present invention comprises a compound having a structure of formula (Ia)




embedded image



or a stereoisomer thereof, an atropisomer thereof, a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable salt of the stereoisomer thereof, or a pharmaceutically acceptable salt of the atropisomer thereof.


Embodiment 3

In another embodiment of the present invention, R1 and R1A is independently H, —C1-6alkyl, —C1-6alkylene-OH, —C1-6alkylene-O—C1-6alkyl, or C1-6alkylene-NR10R11.


Embodiment 4

In another embodiment of the present invention, R1 is H.


Embodiment 5

In another embodiment of the present invention, R1A is H.


Embodiment 6

In another embodiment of the present invention, R2 is independently H or —C1-6alkyl.


Embodiment 7

In another embodiment of the present invention, R2 is H.


Embodiment 8

In another embodiment of the present invention, R3 is —C1-6alkyl.


Embodiment 9

In another embodiment of the present invention, R3 is H.


Embodiment 10

In another embodiment of the present invention, R4 is H or —C1-6alkyl.


Embodiment 11

In another embodiment of the present invention, R4 is H.


Embodiment 12

In another embodiment of the present invention, R4 is —C1-6alkyl.


Embodiment 13

In another embodiment of the present invention, R4 is —CH3.


Embodiment 14

In another embodiment of the present invention, R4A is independently H, or —C1-6alkyl.


Embodiment 15

In another embodiment of the present invention, R4A is H.


Embodiment 16

In another embodiment of the present invention, wherein R4 and R4A, together with the atoms to which they are attached, form a 6- to 12-membered aryl or heteroaryl, a 3- to 8-membered monocyclic or bicyclic cycloalkyl, a 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl, or a 3- to 8-membered monocyclic or bicyclic heterocycloalkyl group, wherein the heteroaryl and heterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl group may include a S═O or SO2.


Embodiment 17

In another embodiment of the present invention, R4 and R4A, together with the atoms to which they are attached, form a heterocycloalkyl ring.


Embodiment 18

In another embodiment of the present invention, R4 and R4A, together with the atoms to which they are attached, form




embedded image


Embodiment 19

In another embodiment of the present invention, R4B is —C1-6alkyl.


Embodiment 20

In another embodiment of the present invention, R4B is H.


Embodiment 21

In another embodiment of the present invention, R5 is H.


Embodiment 22

In another embodiment of the present invention, R5 is —CH3.


Embodiment 23

In another embodiment of the present invention, R6 is independently H, halo or —C1-6alkyl, a 6- to 12-membered aryl, a 5- to 12-membered heteroaryl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, a 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl group, wherein the heteroaryl, spiroheterocycloalkyl and heterocycloalkyl groups have 1, 2, or 3 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl group may include a S═O or SO2.


Embodiment 24

In another embodiment of the present invention, R6 is H.


Embodiment 25

In another embodiment of the present invention, R7 is independently H, halo or —C1-6alkyl, —O—C1-6alkyl, —S—C1-6alkyl, —NO2, —CN, —CF3, a 6- to 12-membered aryl, 5- to 12-membered heteroaryl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, a 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl group, wherein the heteroaryl, and heterocycloalkyl groups have 1, 2, or 3 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2.


Embodiment 26

In another embodiment of the present invention, R7 is halo.


Embodiment 27

In another embodiment of the present invention, R7 is Cl.


Embodiment 28

In another embodiment of the present invention, R7 is Br.


Embodiment 29

In another embodiment of the present invention, R8 is —C1-6alkyl.


Embodiment 30

In another embodiment of the present invention, R8 is H.


Embodiment 31

In another embodiment of the present invention, R9 is independently selected from H, OH, —C1-6alkyl, —OC1-6alkyl, —C1-6alkyl-O—C1-6alkyl, halo, —O-haloC1-6alkyl, —CN, —C1-6alkyl-C(═O)—NRaRb, —NRaRb, —(NRaRbRc)n, —OSO2Ra, —SO2Ra, —(CH2CH2O)nCH3, —(═O), —C(═O), —C(═O)Ra, —OC(═O)Ra, —C(═O)ORa, —C(═O)C(═O)ORa, —C(═O)NRaRb, —O—SiRaRbRc, —SiRaRbRc, —O-(3- to 12-membered cycloakyl), —O-(3- to 12-membered heterocycloakyl), —C(═O)-6- to 12-membered aryl, —C(═O)-5- to 12-membered heteroaryl, —C(═O)-3- to 12-membered cycloalkenyl, —C(═O)-3- to 12-membered monocyclic or bicyclic cycloalkyl, —C(═O)-3- to 12-membered monocyclic or bicyclic heterocycloalkyl, —C(═O)-5- to 12-membered spirocycloalkyl, —C(═O)-5- to 12-membered spiroheterocycloalkyl, —C1-6alkyl-C(═O)-6- to 12-membered aryl, —C1-6alkyl-C(═O)-5- to 12-membered heteroaryl, a —C1-6alkyl-C(═O)-3- to 12-membered monocyclic or bicyclic cycloalkyl, a —C1-6alkyl-C(═O)-3- to 12-membered monocyclic or bicyclic heterocycloalkyl, —C1-6alkyl-C(═O)-5- to 12-membered monocyclic or bicyclic spirocycloalkyl, —C1-6alkyl-C(═O)-5- to 12-membered spiroheterocycloalkyl group, 6- to 12-membered aryl, 5- to 12-membered heteroaryl, a 3- to 12-membered cycloalkenyl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl group, a 5- to 12-membered monocyclic or bicyclic spirocycloalkyl, or a 5- to 12-membered monocyclic or bicyclic spiroheterocycloalkyl group, wherein the heteroaryl, heterocycloalkyl and spiroheterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2; and still further wherein two adjacent carbon atoms or an adjacent carbon atom and adjacent N atom on a R9 group that includes a cycloalkyl, heterocycloalkyl, aryl, heteroaryl, spirocycloalkyl, or a spiroheterocycloalkyl group may join together to form a 5-6 membered cycloalkyl, heterocycloalkyl, aryl, heteroaryl spirocycloalkyl, or a spiroheterocycloalkyl ring that is unsubstituted or is substituted with 1, 2, or 3 R11 groups.


Embodiment 32

In another embodiment of the present invention, R9 is




embedded image


Embodiment 33

In another embodiment of the present invention, R9 is




embedded image


Embodiment 34

In another embodiment of the present invention, R10 is independently H, halo, OH, —NH—C1-8alkyl, —N(C1-8alkyl)2, —NH—C0-3alkylene-C6-14aryl, —NH—C0-3alkylene-C2-14heteroaryl, —NH—C0-3alkylene-C3-14cycloalkyl, —NH—C0-3 alkylene-C2-14heterocycloalkyl, cyano, or C1-6alkylene-amine, —C1-6alkyl, —C(═O)NRaRb, —C(═O)ORa, —C(═O), —C0-3alkylene-C3-14cycloalkyl, —C0-3alkylene-C6-14aryl, —C0-3alkylene-C3-14heteroaryl, —C0-3 alkylene-C3-14cycloalkyl, —C0-3alkylene-C2-14heterocycloalkyl, —C1-6alkoxy, —O—C0-3 alkylene-C6-14aryl, —O—C0-3alkylene-C3-14heteroaryl, —O—C0-3 alkylene-C3-14cycloalkyl, —O—C0-3 alkylene-C2-14heterocycloalkyl, —C(═O)-(3- to 10-membered heterocycloakyl), —C(═O)-6- to 12-membered aryl, —C(═O)-5- to 12-membered heteroaryl, —C(═O)-3- to 12-membered cycloalkenyl, —C(═O)-3- to 12-membered monocyclic or bicyclic cycloalkyl, —C(═O)-3- to 12-membered monocyclic or bicyclic heterocycloalkyl, a —C(═O)-5- to 12-membered bicyclic spirocycloalkyl, a —C(═O)-5- to 12-membered spiroheterocycloalkyl, a 6- to 12-membered aryl, 5- to 12-membered heteroaryl, a 3- to 12-membered cycloalkenyl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl, a 5- to 12-membered monocyclic or bicyclic spirocycloalkyl, or a 5- to 12-membered monocyclic or bicyclic spiroheterocycloalkyl group, wherein the heteroaryl, heterocycloalkyl and spiroheterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2.


Embodiment 35

In another embodiment of the present invention, R10 is —CH3.


Embodiment 36

In another embodiment of the present invention, R10 is —CH2CH3.


Embodiment 37

In another embodiment of the present invention, R10 is a C0-3alkylene-cycloalkyl.


Embodiment 38

In another embodiment of the present invention, R10 is —CH2—C0-3alkylene-cycloalkyl.


Embodiment 39

In another embodiment of the present invention, the compound has a structure selected from the formula in the following table
















embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image












or a stereoisomer thereof, an atropisomer thereof, a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable salt of the stereoisomer thereof, or a pharmaceutically acceptable salt of the atropisomer thereof.


Embodiment 40

In another embodiment of the present invention, the compound has a structure selected from the formula below




embedded image


embedded image


embedded image



or a stereoisomer thereof, an atropisomer thereof, a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable salt of the stereoisomer thereof, or a pharmaceutically acceptable salt of the atropisomer thereof.


Embodiment 41

In another embodiment of the present invention, the compound is any one of the embodiments 1-40 in the form of a pharmaceutically acceptable salt.


Embodiment 42

In another embodiment of the present invention, the embodiment is a pharmaceutical formulation comprising the compound of any one of embodiments 1-41 and a pharmaceutically acceptable excipient.


Embodiment 43

In another embodiment of the present invention, the embodiment is a method of inhibiting KRAS G12C in a cell, comprising contacting the cell with the compound of any one of embodiments 1-42.


Embodiment 44

In another embodiment of the present invention, the embodiment is a method of treating cancer in a subject comprising administering to the subject a therapeutically effective amount of the compound of any one of embodiments 1-42.


Embodiment 45

In another embodiment of the present invention, the embodiment is the method of embodiment 44, wherein the cancer is lung cancer, pancreatic cancer, or colorectal cancer.


Embodiment 46

In another embodiment of the present invention, the embodiment is the method of embodiment 45, wherein the cancer is lung cancer.


Embodiment 47

In another embodiment of the present invention, the embodiment is the method of embodiment 45, wherein the cancer is pancreatic cancer.


Embodiment 48

In another embodiment of the present invention, the embodiment is the method of embodiment 45, wherein the cancer is colorectal cancer.


Embodiment 49

In another embodiment of the present invention, the embodiment is the method of embodiment 44, further comprising administering to the patient in need thereof a therapeutically effective amount of an additional pharmaceutically active compound.


Embodiment 50

In another embodiment of the present invention, the embodiment is the method of embodiment 49, wherein the additional pharmaceutically active compound is carfilzomib.


Embodiment 51

In another embodiment of the present invention, the embodiment is the method of claim 49, wherein the additional pharmaceutically active compound is venetoclax.


Embodiment 52

In another embodiment of the present invention, the embodiment is the method of embodiment 49, wherein the additional pharmaceutically active compound is cytarabine.


Embodiment 53

In another embodiment of the present invention, the embodiment is the method of embodiment 49, wherein the additional pharmaceutically active compound is an MCl-1 inhibitor.


Embodiment 54

In another embodiment of the present invention, the embodiment is the method of embodiment 49, wherein the additional pharmaceutically active compound is AMG-176.


Embodiment 55

In another embodiment of the present invention, the embodiment is the method of embodiment 49, wherein the additional pharmaceutically active compound is daratumumab.


Embodiment 56

In another embodiment of the present invention, the embodiment is the method of embodiment 49, wherein the additional pharmaceutically active compound is an IMiD.


Embodiment 57

In another embodiment of the present invention, the embodiment is the use of a compound according to any one of embodiments 1-42 for treating cancer in a subject.


Embodiment 58

In another embodiment of the present invention, the embodiment is a compound according to any one of embodiments 1-42 in the preparation of a medicament for treating cancer.


Embodiment 59

In another embodiment of the present invention, the embodiment is the compound according to embodiment 58, wherein the cancer is a hematologic malignancy.


Embodiment 60

In another embodiment of the present invention, the embodiment is the method of embodiment 44, wherein the cancer is endometrial cancer, appendix cancer, small intestine cancer or colorectal cancer.


Embodiment 61

In another embodiment of the present invention, the embodiment is the method of embodiment 46, wherein the lung cancer is non-small cell lung cancer.


Embodiment 62

In another embodiment of the present invention, the embodiment is the method of embodiment 44, wherein the cancer is endometrial cancer.


Embodiment 63

The method of claim 44, wherein the cancer is appendix cancer.


Embodiment 64

The method of claim 44, wherein the cancer is small intestine cancer.


Embodiment 65

The method of claim 44, wherein the cancer is a KRAS G12C mutated cancer.


Embodiment 66

In another embodiment of the present invention, the embodiment is the method of embodiment 44, further comprising administering to the patient in need thereof a therapeutically effective amount of an additional pharmaceutically active compound.


Embodiment 67

In another embodiment of the present invention, the embodiment is the method of embodiment 66, wherein the additional pharmaceutically active compound is an anti-PD-1 agent.


Embodiment 68

In another embodiment of the present invention, the embodiment is the method of embodiment 67, wherein the anti-PD-1 agent is BMS-936559.


Embodiment 69

In another embodiment of the present invention, the embodiment is the method of embodiment 67, wherein the anti-PD-1 antagonist is MK-3475.


Embodiment 70

In another embodiment of the present invention, the embodiment is the method of embodiment 66, wherein the additional pharmaceutically active compound is a MEK inhibitor.


Embodiment 71

In another embodiment of the present invention, the embodiment is the Use of a compound according to claim 1 for treating cancer in a subject.


Embodiment 72

In another embodiment of the present invention, the embodiment is the compound according to claim 1 in the preparation of a medicament for treating cancer.


Embodiment 73

In another embodiment of the present invention, the embodiment is a kit for treating cancer, the kit comprising: a compound of claim 1, or the stereoisomer thereof, the atropisomer thereof, the pharmaceutically acceptable salt thereof, the pharmaceutically acceptable salt of the stereoisomer thereof, or the pharmaceutically acceptable salt of the atropisomer thereof; and an additional pharmaceutical active compound.


Embodiment 74

In another embodiment of the present invention, the embodiment is the kit of embodiment 73, wherein the cancer is non-small cell lung cancer.


Embodiment 75

In another embodiment of the present invention, the embodiment is the kit of embodiment 73, wherein the cancer is colorectal cancer.


Embodiment 76

In another embodiment of the present invention, the embodiment is the kit of embodiment 73, wherein the cancer is pancreatic cancer.


Embodiment 77

In another embodiment of the present invention, the embodiment is the kit of embodiment 73, wherein the cancer is endometrial cancer.


Embodiment 78

In another embodiment of the present invention, the embodiment is the kit of embodiment 73, wherein the cancer is appendix cancer.


Embodiment 79

In another embodiment of the present invention, the embodiment is the kit of embodiment 73, wherein the cancer is small intestine cancer.


Embodiment 80

In another embodiment of the present invention, the embodiment is the kit of embodiment 73, wherein the cancer is a KRAS G12C mutated cancer.


Pharmaceutical Compositions, Dosing, and Routes of Administration

Also provided herein are pharmaceutical compositions that includes a compound as disclosed herein, together with a pharmaceutically acceptable excipient, such as, for example, a diluent or carrier. Compounds and pharmaceutical compositions suitable for use in the present invention include those wherein the compound can be administered in an effective amount to achieve its intended purpose. Administration of the compound described in more detail below.


Suitable pharmaceutical formulations can be determined by the skilled artisan depending on the route of administration and the desired dosage. See, e.g., Remington's Pharmaceutical Sciences, 1435-712 (18th ed., Mack Publishing Co, Easton, Pa., 1990). Formulations may influence the physical state, stability, rate of in vivo release and rate of in vivo clearance of the administered agents. Depending on the route of administration, a suitable dose may be calculated according to body weight, body surface areas or organ size. Further refinement of the calculations necessary to determine the appropriate treatment dose is routinely made by those of ordinary skill in the art without undue experimentation, especially in light of the dosage information and assays disclosed herein as well as the pharmacokinetic data obtainable through animal or human clinical trials.


The phrases “pharmaceutically acceptable” or “pharmacologically acceptable” refer to molecular entities and compositions that do not produce adverse, allergic, or other untoward reactions when administered to an animal or a human. As used herein, “pharmaceutically acceptable” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such excipients for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the therapeutic compositions, its use in therapeutic compositions is contemplated. Supplementary active ingredients also can be incorporated into the compositions. In exemplary embodiments, the formulation may comprise corn syrup solids, high-oleic safflower oil, coconut oil, soy oil, L-leucine, calcium phosphate tribasic, L-tyrosine, L-proline, L-lysine acetate, DATEM (an emulsifier), L-glutamine, L-valine, potassium phosphate dibasic, L-isoleucine, L-arginine, L-alanine, glycine, L-asparagine monohydrate, L-serine, potassium citrate, L-threonine, sodium citrate, magnesium chloride, L-histidine, L-methionine, ascorbic acid, calcium carbonate, L-glutamic acid, L-cystine dihydrochloride, L-tryptophan, L-aspartic acid, choline chloride, taurine, m-inositol, ferrous sulfate, ascorbyl palmitate, zinc sulfate, L-carnitine, alpha-tocopheryl acetate, sodium chloride, niacinamide, mixed tocopherols, calcium pantothenate, cupric sulfate, thiamine chloride hydrochloride, vitamin A palmitate, manganese sulfate, riboflavin, pyridoxine hydrochloride, folic acid, beta-carotene, potassium iodide, phylloquinone, biotin, sodium selenate, chromium chloride, sodium molybdate, vitamin D3 and cyanocobalamin.


The compound can be present in a pharmaceutical composition as a pharmaceutically acceptable salt. As used herein, “pharmaceutically acceptable salts” include, for example base addition salts and acid addition salts.


Pharmaceutically acceptable base addition salts may be formed with metals or amines, such as alkali and alkaline earth metals or organic amines. Pharmaceutically acceptable salts of compounds may also be prepared with a pharmaceutically acceptable cation. Suitable pharmaceutically acceptable cations are well known to those skilled in the art and include alkaline, alkaline earth, ammonium and quaternary ammonium cations. Carbonates or hydrogen carbonates are also possible. Examples of metals used as cations are sodium, potassium, magnesium, ammonium, calcium, or ferric, and the like. Examples of suitable amines include isopropylamine, trimethylamine, histidine, N,N′-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, dicyclohexylamine, ethylenediamine, N-methylglucamine, and procaine.


Pharmaceutically acceptable acid addition salts include inorganic or organic acid salts. Examples of suitable acid salts include the hydrochlorides, formates, acetates, citrates, salicylates, nitrates, phosphates. Other suitable pharmaceutically acceptable salts are well known to those skilled in the art and include, for example, formic, acetic, citric, oxalic, tartaric, or mandelic acids, hydrochloric acid, hydrobromic acid, sulfuric acid or phosphoric acid; with organic carboxylic, sulfonic, sulfo or phospho acids or N-substituted sulfamic acids, for example acetic acid, trifluoroacetic acid (TFA), propionic acid, glycolic acid, succinic acid, maleic acid, hydroxymaleic acid, methylmaleic acid, fumaric acid, malic acid, tartaric acid, lactic acid, oxalic acid, gluconic acid, glucaric acid, glucuronic acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, salicylic acid, 4-aminosalicylic acid, 2-phenoxybenzoic acid, 2-acetoxybenzoic acid, embonic acid, nicotinic acid or isonicotinic acid; and with amino acids, such as the 20 alpha amino acids involved in the synthesis of proteins in nature, for example glutamic acid or aspartic acid, and also with phenylacetic acid, methanesulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, ethane 1,2-disulfonic acid, benzenesulfonic acid, 4-methylbenzenesulfonic acid, naphthalene 2-sulfonic acid, naphthalene 1,5-disulfonic acid, 2- or 3-phosphoglycerate, glucose 6-phosphate, N-cyclohexylsulfamic acid (with the formation of cyclamates), or with other acid organic compounds, such as ascorbic acid.


Pharmaceutical compositions containing the compounds disclosed herein can be manufactured in a conventional manner, e.g., by conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or lyophilizing processes. Proper formulation is dependent upon the route of administration chosen.


For oral administration, suitable compositions can be formulated readily by combining a compound disclosed herein with pharmaceutically acceptable excipients such as carriers well known in the art. Such excipients and carriers enable the present compounds to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Pharmaceutical preparations for oral use can be obtained by adding a compound as disclosed herein with a solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients include, for example, fillers and cellulose preparations. If desired, disintegrating agents can be added. Pharmaceutically acceptable ingredients are well known for the various types of formulation and may be for example binders (e.g., natural or synthetic polymers), lubricants, surfactants, sweetening and flavoring agents, coating materials, preservatives, dyes, thickeners, adjuvants, antimicrobial agents, antioxidants and carriers for the various formulation types.


When a therapeutically effective amount of a compound disclosed herein is administered orally, the composition typically is in the form of a solid (e.g., tablet, capsule, pill, powder, or troche) or a liquid formulation (e.g., aqueous suspension, solution, elixir, or syrup).


When administered in tablet form, the composition can additionally contain a functional solid and/or solid carrier, such as a gelatin or an adjuvant. The tablet, capsule, and powder can contain about 1 to about 95% compound, and preferably from about 15 to about 90% compound.


When administered in liquid or suspension form, a functional liquid and/or a liquid carrier such as water, petroleum, or oils of animal or plant origin can be added. The liquid form of the composition can further contain physiological saline solution, sugar alcohol solutions, dextrose or other saccharide solutions, or glycols. When administered in liquid or suspension form, the composition can contain about 0.5 to about 90% by weight of a compound disclosed herein, and preferably about 1 to about 50% of a compound disclosed herein. In one embodiment contemplated, the liquid carrier is non-aqueous or substantially non-aqueous. For administration in liquid form, the composition may be supplied as a rapidly-dissolving solid formulation for dissolution or suspension immediately prior to administration.


When a therapeutically effective amount of a compound disclosed herein is administered by intravenous, cutaneous, or subcutaneous injection, the composition is in the form of a pyrogen-free, parenterally acceptable aqueous solution. The preparation of such parenterally acceptable solutions, having due regard to pH, isotonicity, stability, and the like, is within the skill in the art. A preferred composition for intravenous, cutaneous, or subcutaneous injection typically contains, in addition to a compound disclosed herein, an isotonic vehicle. Such compositions may be prepared for administration as solutions of free base or pharmacologically acceptable salts in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions also can be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations can optionally contain a preservative to prevent the growth of microorganisms.


Injectable compositions can include sterile aqueous solutions, suspensions, or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions, suspensions, or dispersions. In all embodiments the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must resist the contaminating action of microorganisms, such as bacteria and fungi, by optional inclusion of a preservative. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. In one embodiment contemplated, the carrier is non-aqueous or substantially non-aqueous. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size of the compound in the embodiment of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many embodiments, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.


Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the embodiment of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.


Slow release or sustained release formulations may also be prepared in order to achieve a controlled release of the active compound in contact with the body fluids in the GI tract, and to provide a substantially constant and effective level of the active compound in the blood plasma. For example, release can be controlled by one or more of dissolution, diffusion, and ion-exchange. In addition, the slow release approach may enhance absorption via saturable or limiting pathways within the GI tract. For example, the compound may be embedded for this purpose in a polymer matrix of a biological degradable polymer, a water-soluble polymer or a mixture of both, and optionally suitable surfactants. Embedding can mean in this context the incorporation of micro-particles in a matrix of polymers. Controlled release formulations are also obtained through encapsulation of dispersed micro-particles or emulsified micro-droplets via known dispersion or emulsion coating technologies.


For administration by inhalation, compounds of the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant. In the embodiment of a pressurized aerosol, the dosage unit can be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin, for use in an inhaler or insufflator can be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.


The compounds disclosed herein can be formulated for parenteral administration by injection (e.g., by bolus injection or continuous infusion). Formulations for injection can be presented in unit dosage form (e.g., in ampules or in multidose containers), with an added preservative. The compositions can take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing, and/or dispersing agents.


Pharmaceutical formulations for parenteral administration include aqueous solutions of the compounds in water-soluble form. Additionally, suspensions of the compounds can be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils or synthetic fatty acid esters. Aqueous injection suspensions can contain substances which increase the viscosity of the suspension. Optionally, the suspension also can contain suitable stabilizers or agents that increase the solubility of the compounds and allow for the preparation of highly concentrated solutions. Alternatively, a present composition can be in powder form for constitution with a suitable vehicle (e.g., sterile pyrogen-free water) before use.


Compounds disclosed herein also can be formulated in rectal compositions, such as suppositories or retention enemas (e.g., containing conventional suppository bases). In addition to the formulations described previously, the compounds also can be formulated as a depot preparation. Such long-acting formulations can be administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds can be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.


In particular, a compound disclosed herein can be administered orally, buccally, or sublingually in the form of tablets containing excipients, such as starch or lactose, or in capsules or ovules, either alone or in admixture with excipients, or in the form of elixirs or suspensions containing flavoring or coloring agents. Such liquid preparations can be prepared with pharmaceutically acceptable additives, such as suspending agents. A compound also can be injected parenterally, for example, intravenously, intramuscularly, subcutaneously, or intracoronarily. For parenteral administration, the compound is best used in the form of a sterile aqueous solution which can contain other substances, for example, salts, or sugar alcohols, such as mannitol, or glucose, to make the solution isotonic with blood.


For veterinary use, a compound disclosed herein is administered as a suitably acceptable formulation in accordance with normal veterinary practice. The veterinarian can readily determine the dosing regimen and route of administration that is most appropriate for a particular animal.


In some embodiments, all the necessary components for the treatment of KRAS-related disorder using a compound as disclosed herein either alone or in combination with another agent or intervention traditionally used for the treatment of such disease may be packaged into a kit. Specifically, the present invention provides a kit for use in the therapeutic intervention of the disease comprising a packaged set of medicaments that include the compound disclosed herein as well as buffers and other components for preparing deliverable forms of said medicaments, and/or devices for delivering such medicaments, and/or any agents that are used in combination therapy with the compound disclosed herein, and/or instructions for the treatment of the disease packaged with the medicaments. The instructions may be fixed in any tangible medium, such as printed paper, or a computer readable magnetic or optical medium, or instructions to reference a remote computer data source such as a world wide web page accessible via the internet.


A “therapeutically effective amount” means an amount effective to treat or to prevent development of, or to alleviate the existing symptoms of, the subject being treated. Determination of the effective amounts is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein. Generally, a “therapeutically effective dose” refers to that amount of the compound that results in achieving the desired effect. For example, in one preferred embodiment, a therapeutically effective amount of a compound disclosed herein decreases KRAS activity by at least 5%, compared to control, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, or at least 90%.


The amount of compound administered can be dependent on the subject being treated, on the subject's age, health, sex, and weight, the kind of concurrent treatment (if any), severity of the affliction, the nature of the effect desired, the manner and frequency of treatment, and the judgment of the prescribing physician. The frequency of dosing also can be dependent on pharmacodynamic effects on arterial oxygen pressures. However, the most preferred dosage can be tailored to the individual subject, as is understood and determinable by one of skill in the art, without undue experimentation. This typically involves adjustment of a standard dose (e.g., reduction of the dose if the patient has a low body weight).


While individual needs vary, determination of optimal ranges of effective amounts of the compound is within the skill of the art. For administration to a human in the curative or prophylactic treatment of the conditions and disorders identified herein, for example, typical dosages of the compounds of the present invention can be about 0.05 mg/kg/day to about 50 mg/kg/day, for example at least 0.05 mg/kg, at least 0.08 mg/kg, at least 0.1 mg/kg, at least 0.2 mg/kg, at least 0.3 mg/kg, at least 0.4 mg/kg, or at least 0.5 mg/kg, and preferably 50 mg/kg or less, 40 mg/kg or less, 30 mg/kg or less, 20 mg/kg or less, or 10 mg/kg or less, which can be about 2.5 mg/day (0.5 mg/kg×5 kg) to about 5000 mg/day (50 mg/kg×100 kg), for example. For example, dosages of the compounds can be about 0.1 mg/kg/day to about 50 mg/kg/day, about 0.05 mg/kg/day to about 10 mg/kg/day, about 0.05 mg/kg/day to about 5 mg/kg/day, about 0.05 mg/kg/day to about 3 mg/kg/day, about 0.07 mg/kg/day to about 3 mg/kg/day, about 0.09 mg/kg/day to about 3 mg/kg/day, about 0.05 mg/kg/day to about 0.1 mg/kg/day, about 0.1 mg/kg/day to about 1 mg/kg/day, about 1 mg/kg/day to about 10 mg/kg/day, about 1 mg/kg/day to about 5 mg/kg/day, about 1 mg/kg/day to about 3 mg/kg/day, about 3 mg/day to about 500 mg/day, about 5 mg/day to about 250 mg/day, about 10 mg/day to about 100 mg/day, about 3 mg/day to about 10 mg/day, or about 100 mg/day to about 250 mg/day. Such doses may be administered in a single dose or it may be divided into multiple doses.


Methods of Using KRAS G12C Inhibitors

The present disclosure provides a method of inhibiting RAS-mediated cell signaling comprising contacting a cell with an effective amount of one or more compounds disclosed herein. Inhibition of RAS-mediated signal transduction can be assessed and demonstrated by a wide variety of ways known in the art. Non-limiting examples include a showing of (a) a decrease in GTPase activity of RAS; (b) a decrease in GTP binding affinity or an increase in GDP binding affinity; (c) an increase in K off of GTP or a decrease in K off of GDP; (d) a decrease in the levels of signaling transduction molecules downstream in the RAS pathway, such as a decrease in pMEK, pERK, or pAKT levels; and/or (e) a decrease in binding of RAS complex to downstream signaling molecules including but not limited to Raf Kits and commercially available assays can be utilized for determining one or more of the above.


The disclosure also provides methods of using the compounds or pharmaceutical compositions of the present disclosure to treat disease conditions, including but not limited to conditions implicated by G12C KRAS, HRAS or NRAS mutation (e.g., cancer).


In some embodiments, a method for treatment of cancer is provided, the method comprising administering an effective amount of any of the foregoing pharmaceutical compositions comprising a compound as disclosed herein to a subject in need thereof. In some embodiments, the cancer is mediated by a KRAS, HRAS or NRAS G12C mutation. In various embodiments, the cancer is pancreatic cancer, colorectal cancer or lung cancer. In some embodiments, the cancer is gall bladder cancer, thyroid cancer, and bile duct cancer.


In some embodiments the disclosure provides method of treating a disorder in a subject in need thereof, wherein the said method comprises determining if the subject has a KRAS, HRAS or NRAS G12C mutation and if the subject is determined to have the KRAS, HRAS or NRAS G12C mutation, then administering to the subject a therapeutically effective dose of at least one compound as disclosed herein or a pharmaceutically acceptable salt thereof.


The disclosed compounds inhibit anchorage-independent cell growth and therefore have the potential to inhibit tumor metastasis. Accordingly, another embodiment the disclosure provides a method for inhibiting tumor metastasis, the method comprising administering an effective amount a compound disclosed herein.


KRAS, HRAS or NRAS G12C mutations have also been identified in hematological malignancies (e.g., cancers that affect blood, bone marrow and/or lymph nodes). Accordingly, certain embodiments are directed to administration of a disclosed compounds (e.g., in the form of a pharmaceutical composition) to a patient in need of treatment of a hematological malignancy. Such malignancies include, but are not limited to leukemias and lymphomas. For example, the presently disclosed compounds can be used for treatment of diseases such as Acute lymphoblastic leukemia (ALL), Acute myelogenous leukemia (AML), Chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), Chronic myelogenous leukemia (CML), Acute monocytic leukemia (AMoL) and/or other leukemias. In other embodiments, the compounds are useful for treatment of lymphomas such as all subtypes of Hodgkins lymphoma or non-Hodgkins lymphoma. In various embodiments, the compounds are useful for treatment of plasma cell malignancies such as multiple myeloma, mantle cell lymphoma, and Waldenstrom's macroglubunemia.


Determining whether a tumor or cancer comprises a G12C KRAS, HRAS or NRAS mutation can be undertaken by assessing the nucleotide sequence encoding the KRAS, HRAS or NRAS protein, by assessing the amino acid sequence of the KRAS, HRAS or NRAS protein, or by assessing the characteristics of a putative KRAS, HRAS or NRAS mutant protein. The sequence of wild-type human KRAS, HRAS or NRAS is known in the art, (e.g. Accession No. NP203524).


Methods for detecting a mutation in a KRAS, HRAS or NRAS nucleotide sequence are known by those of skill in the art. These methods include, but are not limited to, polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) assays, polymerase chain reaction-single strand conformation polymorphism (PCR-SSCP) assays, real-time PCR assays, PCR sequencing, mutant allele-specific PCR amplification (MASA) assays, direct sequencing, primer extension reactions, electrophoresis, oligonucleotide ligation assays, hybridization assays, TaqMan assays, SNP genotyping assays, high resolution melting assays and microarray analyses. In some embodiments, samples are evaluated for G12C KRAS, HRAS or NRAS mutations by real-time PCR. In real-time PCR, fluorescent probes specific for the KRAS, HRAS or NRAS G12C mutation are used. When a mutation is present, the probe binds and fluorescence is detected. In some embodiments, the KRAS, HRAS or NRAS G12C mutation is identified using a direct sequencing method of specific regions (e.g., exon 2 and/or exon 3) in the KRAS, HRAS or NRAS gene. This technique will identify all possible mutations in the region sequenced.


Methods for detecting a mutation in a KRAS, HRAS or NRAS protein are known by those of skill in the art. These methods include, but are not limited to, detection of a KRAS, HRAS or NRAS mutant using a binding agent (e.g., an antibody) specific for the mutant protein, protein electrophoresis and Western blotting, and direct peptide sequencing.


Methods for determining whether a tumor or cancer comprises a G12C KRAS, HRAS or NRAS mutation can use a variety of samples. In some embodiments, the sample is taken from a subject having a tumor or cancer. In some embodiments, the sample is a fresh tumor/cancer sample. In some embodiments, the sample is a frozen tumor/cancer sample. In some embodiments, the sample is a formalin-fixed paraffin-embedded sample. In some embodiments, the sample is a circulating tumor cell (CTC) sample. In some embodiments, the sample is processed to a cell lysate. In some embodiments, the sample is processed to DNA or RNA.


The disclosure also relates to a method of treating a hyperproliferative disorder in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound as disclosed herein, or a pharmaceutically acceptable salt thereof. In some embodiments, said method relates to the treatment of a subject who suffers from a cancer such as acute myeloid leukemia, cancer in adolescents, adrenocortical carcinoma childhood, AIDS-related cancers (e.g. Lymphoma and Kaposi's Sarcoma), anal cancer, appendix cancer, astrocytomas, atypical teratoid, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumors, Burkitt lymphoma, carcinoid tumor, atypical teratoid, embryonal tumors, germ cell tumor, primary lymphoma, cervical cancer, childhood cancers, chordoma, cardiac tumors, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myleoproliferative disorders, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, extrahepatic ductal carcinoma in situ (DCIS), embryonal tumors, CNS cancer, endometrial cancer, ependymoma, esophageal cancer, esthesioneuroblastoma, ewing sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor, eye cancer, fibrous histiocytoma of bone, gall bladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumors (GIST), germ cell tumor, gestational trophoblastic tumor, hairy cell leukemia, head and neck cancer, heart cancer, liver cancer, Hodgkin lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumors, pancreatic neuroendocrine tumors, kidney cancer, laryngeal cancer, lip and oral cavity cancer, liver cancer,custom characterlobular carcinoma in situ (LCIS),custom character lung cancer, lymphoma, metastatic squamous neck cancer with occult primary,custom character midline tract carcinoma,custom charactermouth cancer, multiple endocrine neoplasia syndromes,custom charactermultiple myeloma/plasma cell neoplasm,custom charactermycosis fungoides, myelodysplastic syndromes,custom charactermyelodysplastic/myeloproliferative neoplasms, multiple myeloma, merkel cell carcinoma, malignant mesothelioma, malignant fibrous histiocytoma of bone and osteosarcoma, nasal cavity and paranasal sinus cancer,custom character nasopharyngeal cancer, neuroblastoma,custom character non-hodgkin lymphoma,custom character non-small cell lung cancer (NSCLC), oral cancer, lip and oral cavity cancer, oropharyngeal cancer, ovarian cancer, pancreatic cancer, papillomatosis, paraganglioma,custom character paranasal sinus and nasal cavity cancer,custom characterparathyroid cancer,custom characterpenile cancer, pharyngeal cancer,custom character pleuropulmonary blastoma, primary central nervous system (CNS) lymphoma, prostate cancer, rectal cancer, transitional cell cancer,custom characterretinoblastoma,custom characterrhabdomyosarcoma, salivary gland cancer, skin cancer, stomach (gastric) cancer, small cell lung cancer, small intestine cancer, soft tissue sarcoma, T-Cell lymphoma, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer, transitional cell cancer of the renal pelvis and ureter, trophoblastic tumor, unusual cancers of childhood,custom character urethral cancer, uterine sarcoma, vaginal cancer, vulvar cancer, or viral-induced cancer. In some embodiments, said method relates to the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e.g., psoriasis), restenosis, or prostate (e.g., benign prostatic hypertrophy (BPH)).


In some embodiments, the methods for treatment are directed to treating lung cancers, the methods comprise administering an effective amount of any of the above described compound (or a pharmaceutical composition comprising the same) to a subject in need thereof. In certain embodiments the lung cancer is a non-small cell lung carcinoma (NSCLC), for example adenocarcinoma, squamous-cell lung carcinoma or large-cell lung carcinoma. In some embodiments, the lung cancer is a small cell lung carcinoma. Other lung cancers treatable with the disclosed compounds include, but are not limited to, glandular tumors, carcinoid tumors and undifferentiated carcinomas.


The disclosure further provides methods of modulating a G12C Mutant KRAS, HRAS or NRAS protein activity by contacting the protein with an effective amount of a compound of the disclosure. Modulation can be inhibiting or activating protein activity. In some embodiments, the disclosure provides methods of inhibiting protein activity by contacting the G12C Mutant KRAS, HRAS or NRAS protein with an effective amount of a compound of the disclosure in solution. In some embodiments, the disclosure provides methods of inhibiting the G12C Mutant KRAS, HRAS or NRAS protein activity by contacting a cell, tissue, or organ that expresses the protein of interest. In some embodiments, the disclosure provides methods of inhibiting protein activity in subject including but not limited to rodents and mammal (e.g., human) by administering into the subject an effective amount of a compound of the disclosure. In some embodiments, the percentage modulation exceeds 25%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%. In some embodiments, the percentage of inhibiting exceeds 25%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%.


In some embodiments, the disclosure provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in a cell by contacting said cell with an amount of a compound of the disclosure sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said cell. In some embodiments, the disclosure provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in a tissue by contacting said tissue with an amount of a compound of the disclosure sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said tissue. In some embodiments, the disclosure provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in an organism by contacting said organism with an amount of a compound of the disclosure sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said organism. In some embodiments, the disclosure provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in an animal by contacting said animal with an amount of a compound of the disclosure sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said animal. In some embodiments, the disclosure provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in a mammal by contacting said mammal with an amount of a compound of the disclosure sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said mammal. In some embodiments, the disclosure provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in a human by contacting said human with an amount of a compound of the disclosure sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said human. The present disclosure provides methods of treating a disease mediated by KRAS, HRAS or NRAS G12C activity in a subject in need of such treatment.


Combination Therapy

The present disclosure also provides methods for combination therapies in which an agent known to modulate other pathways, or other components of the same pathway, or even overlapping sets of target enzymes are used in combination with a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. In one aspect, such therapy includes but is not limited to the combination of one or more compounds of the disclosure with chemotherapeutic agents, therapeutic antibodies, and radiation treatment, to provide a synergistic or additive therapeutic effect.


Many chemotherapeutics are presently known in the art and can be used in combination with the compounds of the disclosure. In some embodiments, the chemotherapeutic is selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, anti-hormones, angiogenesis inhibitors, and anti-androgens. Non-limiting examples are chemotherapeutic agents, cytotoxic agents, and non-peptide small molecules such as Gleevec® (Imatinib Mesylate), Kyprolis® (carfilzomib), Velcade® (bortezomib), Casodex (bicalutamide), Iressa® (gefitinib), Venclexta™ (venetoclax) and Adriamycin™, (docorubicin) as well as a host of chemotherapeutic agents. Non-limiting examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (Cytoxan™); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamine; nitrogen mustards such as chlorambucil, chlomaphazine, chlorocyclophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, calicheamicin, carabicin, carminomycin, carzinophilin, Casodex™, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine, androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfomithine; elliptinium acetate; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK; razoxane; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2′,2″-trichlorotriethylamine; urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (“Ara-C”); cyclophosphamide; thiotepa; taxanes, e.g. paclitaxel and docetaxel; retinoic acid; esperamicins; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above.


Also included as suitable chemotherapeutic cell conditioners are anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens including for example tamoxifen, (Nolvadex™), raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY 117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; camptothecin-11 (CPT-11); topoisomerase inhibitor RFS 2000; difluoromethylomithine (DMFO).


Where desired, the compounds or pharmaceutical composition of the present disclosure can be used in combination with commonly prescribed anti-cancer drugs such as Herceptin®, Avastin®, Erbitux®, Rituxan®, Taxol®, Arimidex®, Taxotere®, ABVD, AVICINE, Abagovomab, Acridine carboxamide, Adecatumumab, 17-N-Allylamino-17-demethoxygeldanamycin, Alpharadin, Alvocidib, 3-Aminopyridine-2-carboxaldehyde thiosemicarbazone, Amonafide, Anthracenedione, Anti-CD22 immunotoxins, Antineoplastic, Antitumorigenic herbs, Apaziquone, Atiprimod, Azathioprine, Belotecan, Bendamustine, BIBW 2992, Biricodar, Brostallicin, Bryostatin, Buthionine sulfoximine, CBV (chemotherapy), Calyculin, cell-cycle nonspecific antineoplastic agents, Dichloroacetic acid, Discodermolide, Elsamitrucin, Enocitabine, Epothilone, Eribulin, Everolimus, Exatecan, Exisulind, Ferruginol, Forodesine, Fosfestrol, ICE chemotherapy regimen, IT-101, Imexon, Imiquimod, Indolocarbazole, Irofulven, Laniquidar, Larotaxel, Lenalidomide, Lucanthone, Lurtotecan, Mafosfamide, Mitozolomide, Nafoxidine, Nedaplatin, Olaparib, Ortataxel, PAC-1, Pawpaw, Pixantrone, Proteasome inhibitor, Rebeccamycin, Resiquimod, Rubitecan, SN-38, Salinosporamide A, Sapacitabine, Stanford V, Swainsonine, Talaporfin, Tariquidar, Tegafur-uracil, Temodar, Tesetaxel, Triplatin tetranitrate, Tris(2-chloroethyl)amine, Troxacitabine, Uramustine, Vadimezan, Vinflunine, ZD6126 or Zosuquidar.


This disclosure further relates to a method for using the compounds or pharmaceutical compositions provided herein, in combination with radiation therapy for inhibiting abnormal cell growth or treating the hyperproliferative disorder in the mammal. Techniques for administering radiation therapy are known in the art, and these techniques can be used in the combination therapy described herein. The administration of the compound of the disclosure in this combination therapy can be determined as described herein.


Radiation therapy can be administered through one of several methods, or a combination of methods, including without limitation external-beam therapy, internal radiation therapy, implant radiation, stereotactic radiosurgery, systemic radiation therapy, radiotherapy and permanent or temporary interstitial brachytherapy. The term “brachytherapy,” as used herein, refers to radiation therapy delivered by a spatially confined radioactive material inserted into the body at or near a tumor or other proliferative tissue disease site. The term is intended without limitation to include exposure to radioactive isotopes (e.g. At-211, I-131, I-125, Y-90, Re-186, Re-188, Sm-153, Bi-212, P-32, and radioactive isotopes of Lu). Suitable radiation sources for use as a cell conditioner of the present disclosure include both solids and liquids. By way of non-limiting example, the radiation source can be a radionuclide, such as I-125, I-131, Yb-169, Ir-192 as a solid source, 1-125 as a solid source, or other radionuclides that emit photons, beta particles, gamma radiation, or other therapeutic rays. The radioactive material can also be a fluid made from any solution of radionuclide(s), e.g., a solution of I-125 or I-131, or a radioactive fluid can be produced using a slurry of a suitable fluid containing small particles of solid radionuclides, such as Au-198, Y-90. Moreover, the radionuclide(s) can be embodied in a gel or radioactive micro spheres.


The compounds or pharmaceutical compositions of the disclosure can be used in combination with an amount of one or more substances selected from anti-angiogenesis agents, signal transduction inhibitors, antiproliferative agents, glycolysis inhibitors, or autophagy inhibitors.


Anti-angiogenesis agents, such as MMP-2 (matrix-metalloproteinase 2) inhibitors, MMP-9 (matrix-metalloproteinase 9) inhibitors, and COX-11 (cyclooxygenase 11) inhibitors, can be used in conjunction with a compound of the disclosure and pharmaceutical compositions described herein. Anti-angiogenesis agents include, for example, rapamycin, temsirolimus (CCI-779), everolimus (RAD001), sorafenib, sunitinib, and bevacizumab. Examples of useful COX-II inhibitors include alecoxib, valdecoxib, and rofecoxib. Examples of useful matrix metalloproteinase inhibitors are described in WO 96/33172 WO 96/27583 European Patent Publication EP0818442, European Patent Publication EP1004578, WO 98/07697, WO 98/03516, WO 98/34918, WO 98/34915, WO 98/33768, WO 98/30566, European Patent Publication 606046, European Patent Publication 931 788, WO 90/05719, WO 99/52910, WO 99/52889, WO 99/29667, WO1999007675, European Patent Publication EP1786785, European Patent Publication No. EP1181017, United States Publication No. US20090012085, United States Publication U.S. Pat. No. 5,863,949, United States Publication U.S. Pat. No. 5,861,510, and European Patent Publication EP0780386, all of which are incorporated herein in their entireties by reference. Preferred MMP-2 and MMP-9 inhibitors are those that have little or no activity inhibiting MMP-1. More preferred, are those that selectively inhibit MMP-2 and/or AMP-9 relative to the other matrix-metalloproteinases (i.e., MAP-1, MMP-3, MMP-4, MMP-5, MMP-6, MMP-7, MMP-8, MMP-10, MMP-11, MMP-12, and MMP-13). Some specific examples of MMP inhibitors useful in the disclosure are AG-3340, RO 32-3555, and RS 13-0830.


The present compounds may also be used in co-therapies with other anti-neoplastic agents, such as acemannan, aclarubicin, aldesleukin, alemtuzumab, alitretinoin, altretamine, amifostine, aminolevulinic acid, amrubicin, amsacrine, anagrelide, anastrozole, ANCER, ancestim, ARGLABIN, arsenic trioxide, BAM 002 (Novelos), bexarotene, bicalutamide, broxuridine, capecitabine, celmoleukin, cetrorelix, cladribine, clotrimazole, cytarabine ocfosfate, DA 3030 (Dong-A), daclizumab, denileukin diftitox, deslorelin, dexrazoxane, dilazep, docetaxel, docosanol, doxercalciferol, doxifluridine, doxorubicin, bromocriptine, carmustine, cytarabine, fluorouracil, HIT diclofenac, interferon alfa, daunorubicin, doxorubicin, tretinoin, edelfosine, edrecolomab, eflomithine, emitefur, epirubicin, epoetin beta, etoposide phosphate, exemestane, exisulind, fadrozole, filgrastim, finasteride, fludarabine phosphate, formestane, fotemustine, gallium nitrate, gemcitabine, gemtuzumab zogamicin, gimeracil/oteracil/tegafur combination, glycopine, goserelin, heptaplatin, human chorionic gonadotropin, human fetal alpha fetoprotein, ibandronic acid, idarubicin, (imiquimod, interferon alfa, interferon alfa, natural, interferon alfa-2, interferon alfa-2a, interferon alfa-2b, interferon alfa-N1, interferon alfa-n3, interferon alfacon-1, interferon alpha, natural, interferon beta, interferon beta-1a, interferon beta-1b, interferon gamma, natural interferon gamma-1a, interferon gamma-1b, interleukin-1 beta, iobenguane, irinotecan, irsogladine, lanreotide, LC 9018 (Yakult), leflunomide, lenograstim, lentinan sulfate, letrozole, leukocyte alpha interferon, leuprorelin, levamisole+fluorouracil, liarozole, lobaplatin, lonidamine, lovastatin, masoprocol, melarsoprol, metoclopramide, mifepristone, miltefosine, mirimostim, mismatched double stranded RNA, mitoguazone, mitolactol, mitoxantrone, molgramostim, nafarelin, naloxone+pentazocine, nartograstim, nedaplatin, nilutamide, noscapine, novel erythropoiesis stimulating protein, NSC 631570 octreotide, oprelvekin, osaterone, oxaliplatin, paclitaxel, pamidronic acid, pegaspargase, peginterferon alfa-2b, pentosan polysulfate sodium, pentostatin, picibanil, pirarubicin, rabbit antithymocyte polyclonal antibody, polyethylene glycol interferon alfa-2a, porfimer sodium, raloxifene, raltitrexed, rasburiembodiment, rhenium Re 186 etidronate, RII retinamide, rituximab, romurtide, samarium (153 Sm) lexidronam, sargramostim, sizofiran, sobuzoxane, sonermin, strontium-89 chloride, suramin, tasonermin, tazarotene, tegafur, temoporfin, temozolomide, teniposide, tetrachlorodecaoxide, thalidomide, thymalfasin, thyrotropin alfa, topotecan, toremifene, tositumomab-iodine 131, trastuzumab, treosulfan, tretinoin, trilostane, trimetrexate, triptorelin, tumor necrosis factor alpha, natural, ubenimex, bladder cancer vaccine, Maruyama vaccine, melanoma lysate vaccine, valrubicin, verteporfin, vinorelbine, VIRULIZIN, zinostatin stimalamer, or zoledronic acid; abarelix; AE 941 (Aeterna), ambamustine, antisense oligonucleotide, bcl-2 (Genta), APC 8015 (Dendreon), cetuximab, decitabine, dexaminoglutethimide, diaziquone, EL 532 (Elan), EM 800 (Endorecherche), eniluracil, etanidazole, fenretinide, filgrastim SD01 (Amgen), fulvestrant, galocitabine, gastrin 17 immunogen, HLA-B7 gene therapy (Vical), granulocyte macrophage colony stimulating factor, histamine dihydrochloride, ibritumomab tiuxetan, ilomastat, IM 862 (Cytran), interleukin-2, iproxifene, LDI 200 (Milkhaus), leridistim, lintuzumab, CA 125 MAb (Biomira), cancer MAb (Japan Pharmaceutical Development), HER-2 and Fc MAb (Medarex), idiotypic 105AD7 MAb (CRC Technology), idiotypic CEA MAb (Trilex), LYM-1-iodine 131 MAb (Techniclone), polymorphic epithelial mucin-yttrium 90 MAb (Antisoma), marimastat, menogaril, mitumomab, motexafin gadolinium, MX 6 (Galderma), nelarabine, nolatrexed, P 30 protein, pegvisomant, pemetrexed, porfiromycin, prinomastat, RL 0903 (Shire), rubitecan, satraplatin, sodium phenylacetate, sparfosic acid, SRL 172 (SR Pharma), SU 5416 (SUGEN), TA 077 (Tanabe), tetrathiomolybdate, thaliblastine, thrombopoietin, tin ethyl etiopurpurin, tirapazamine, cancer vaccine (Biomira), melanoma vaccine (New York University), melanoma vaccine (Sloan Kettering Institute), melanoma oncolysate vaccine (New York Medical College), viral melanoma cell lysates vaccine (Royal Newcastle Hospital), or valspodar.


The compounds of the invention may further be used with VEGFR inhibitors. Other compounds described in the following patents and patent applications can be used in combination therapy: U.S. Pat. No. 6,258,812, US 2003/0105091, WO 01/37820, U.S. Pat. No. 6,235,764, WO 01/32651, U.S. Pat. Nos. 6,630,500, 6,515,004, 6,713,485, 5,521,184, 5,770,599, 5,747,498, WO 02/68406, WO 02/66470, WO 02/55501, WO 04/05279, WO 04/07481, WO 04/07458, WO 04/09784, WO 02/59110, WO 99/45009, WO 00/59509, WO 99/61422, U.S. Pat. No. 5,990,141, WO 00/12089, and WO 00/02871.


In some embodiments, the combination comprises a composition of the present invention in combination with at least one anti-angiogenic agent. Agents are inclusive of, but not limited to, in vitro synthetically prepared chemical compositions, antibodies, antigen binding regions, radionuclides, and combinations and conjugates thereof. An agent can be an agonist, antagonist, allosteric modulator, toxin or, more generally, may act to inhibit or stimulate its target (e.g., receptor or enzyme activation or inhibition), and thereby promote cell death or arrest cell growth.


Exemplary anti-angiogenic agents include ERBITUX™ (IMC-C225), KDR (kinase domain receptor) inhibitory agents (e.g., antibodies and antigen binding regions that specifically bind to the kinase domain receptor), anti-VEGF agents (e.g., antibodies or antigen binding regions that specifically bind VEGF, or soluble VEGF receptors or a ligand binding region thereof) such as AVASTIN™ or VEGF-TRAP™, and anti-VEGF receptor agents (e.g., antibodies or antigen binding regions that specifically bind thereto), EGFR inhibitory agents (e.g., antibodies or antigen binding regions that specifically bind thereto) such as Vectibix (panitumumab), IRESSA™ (gefitinib), TARCEVA™ (erlotinib), anti-Ang1 and anti-Ang2 agents (e.g., antibodies or antigen binding regions specifically binding thereto or to their receptors, e.g., Tie2/Tek), and anti-Tie2 kinase inhibitory agents (e.g., antibodies or antigen binding regions that specifically bind thereto). The pharmaceutical compositions of the present invention can also include one or more agents (e.g., antibodies, antigen binding regions, or soluble receptors) that specifically bind and inhibit the activity of growth factors, such as antagonists of hepatocyte growth factor (HGF, also known as Scatter Factor), and antibodies or antigen binding regions that specifically bind its receptor “c-met”.


Other anti-angiogenic agents include Campath, IL-8, B-FGF, Tek antagonists (Ceretti et al., U.S. Publication No. 2003/0162712; U.S. Pat. No. 6,413,932), anti-TWEAK agents (e.g., specifically binding antibodies or antigen binding regions, or soluble TWEAK receptor antagonists; see, Wiley, U.S. Pat. No. 6,727,225), ADAM distintegrin domain to antagonize the binding of integrin to its ligands (Fanslow et al., U.S. Publication No. 2002/0042368), specifically binding anti-eph receptor and/or anti-ephrin antibodies or antigen binding regions (U.S. Pat. Nos. 5,981,245; 5,728,813; 5,969,110; 6,596,852; 6,232,447; 6,057,124 and patent family members thereof), and anti-PDGF-BB antagonists (e.g., specifically binding antibodies or antigen binding regions) as well as antibodies or antigen binding regions specifically binding to PDGF-BB ligands, and PDGFR kinase inhibitory agents (e.g., antibodies or antigen binding regions that specifically bind thereto).


Additional anti-angiogenic/anti-tumor agents include: SD-7784 (Pfizer, USA); cilengitide. (Merck KGaA, Germany, EPO 770622); pegaptanib octasodium, (Gilead Sciences, USA); Alphastatin, (BioActa, UK); M-PGA, (Celgene, USA, U.S. Pat. No. 5,712,291); ilomastat, (Arriva, USA, U.S. Pat. No. 5,892,112); emaxanib, (Pfizer, USA, U.S. Pat. No. 5,792,783); vatalanib, (Novartis, Switzerland); 2-methoxyestradiol, (EntreMed, which changed their name to CASI Pharmaceuticals, USA); TLC ELL-12, (formerly Elan, Ireland); anecortave acetate, (Alcon, USA); alpha-D148 Mab, (Amgen, USA); CEP-7055, (Cephalon, USA); anti-Vn Mab, (Crucell, Netherlands) DAC:antiangiogenic, (ConjuChem, Canada); Angiocidin, (InKine Pharmaceutical, USA); KM-2550, (Kyowa Hakko, Japan); SU-0879, (Pfizer, USA); CGP-79787, (Novartis, Switzerland, EP 970070); ARGENT technology, (Ariad, USA); YIGSR-Stealth, (Johnson & Johnson, USA); fibrinogen-E fragment, (BioActa, UK); angiogenesis inhibitor, (Trigen, UK); TBC-1635, (Encysive Pharmaceuticals, USA); SC-236, (Pfizer, USA); ABT-567, (Abbott, USA); Metastatin, (EntreMed, USA); angiogenesis inhibitor, (Tripep, Sweden); maspin, (Sosei, Japan); 2-methoxyestradiol, (Oncology Sciences Corporation, USA); ER-68203-00, (IVAX, USA); Benefin, (Lane Labs, USA); Tz-93, (Tsumura, Japan); TAN-1120, (Takeda, Japan); FR-111142, (Fujisawa, Japan, JP 02233610); platelet factor 4, (RepliGen, USA, EP 407122); vascular endothelial growth factor antagonist, (Borean, Denmark); bevacizumab (plNN), (Genentech, USA); angiogenesis inhibitors, (SUGEN, USA); XL 784, (Exelixis, USA); XL 647, (Exelixis, USA); MAb, alpha5beta3 integrin, second generation, (Applied Molecular Evolution, USA and MedImmune, USA); gene therapy, retinopathy, (Oxford BioMedica, UK); enzastaurin hydrochloride (USAN), (Lilly, USA); CEP 7055, (Cephalon, USA and Sanofi-Synthelabo, France); BC 1, (Genoa Institute of Cancer Research, Italy); angiogenesis inhibitor, (Alchemia, Australia); VEGF antagonist, (Regeneron, USA); rBPI 21 and BPI-derived antiangiogenic, (XOMA, USA); PI 88, (Progen, Australia); cilengitide (plNN), (Merck KGaA, German; Munich Technical University, Germany, Scripps Clinic and Research Foundation, USA); cetuximab (INN), (Aventis, France); AVE 8062, (Ajinomoto, Japan); AS 1404, (Cancer Research Laboratory, New Zealand); SG 292, (Telios, USA); Endostatin, (Boston Childrens Hospital, USA); ATN 161, (Attenuon, USA); ANGIOSTATIN, (Boston Childrens Hospital, USA); 2-methoxyestradiol, (Boston Childrens Hospital, USA); ZD 6474, (AstraZeneca, UK); ZD 6126, (Angiogene Pharmaceuticals, UK); PPI 2458, (Praecis, USA); AZD 9935, (AstraZeneca, UK); AZD 2171, (AstraZeneca, UK); vatalanib (pINN), (Novartis, Switzerland and Schering AG, Germany); tissue factor pathway inhibitors, (EntreMed, USA); pegaptanib (Pinn), (Gilead Sciences, USA); xanthorrhizol, (Yonsei University, South Korea); vaccine, gene-based, VEGF-2, (Scripps Clinic and Research Foundation, USA); SPV5.2, (Supratek, Canada); SDX 103, (University of California at San Diego, USA); PX 478, (ProlX, USA); METASTATIN, (EntreMed, USA); troponin I, (Harvard University, USA); SU 6668, (SUGEN, USA); OXI 4503, (OXiGENE, USA); o-guanidines, (Dimensional Pharmaceuticals, USA); motuporamine C, (British Columbia University, Canada); CDP 791, (Celltech Group, UK); atiprimod (pINN), (GlaxoSmithKline, UK); E 7820, (Eisai, Japan); CYC 381, (Harvard University, USA); AE 941, (Aeterna, Canada); vaccine, angiogenesis, (EntreMed, USA); urokinase plasminogen activator inhibitor, (Dendreon, USA); oglufanide (pINN), (Melmotte, USA); HIF-lalfa inhibitors, (Xenova, UK); CEP 5214, (Cephalon, USA); BAY RES 2622, (Bayer, Germany); Angiocidin, (InKine, USA); A6, (Angstrom, USA); KR 31372, (Korea Research Institute of Chemical Technology, South Korea); GW 2286, (GlaxoSmithKline, UK); EHT 0101, (ExonHit, France); CP 868596, (Pfizer, USA); CP 564959, (OSI, USA); CP 547632, (Pfizer, USA); 786034, (GlaxoSmithKline, UK); KRN 633, (Kirin Brewery, Japan); drug delivery system, intraocular, 2-methoxyestradiol, (EntreMed, USA); anginex, (Maastricht University, Netherlands, and Minnesota University, USA); ABT 510, (Abbott, USA); AAL 993, (Novartis, Switzerland); VEGI, (ProteomTech, USA); tumor necrosis factor-alpha inhibitors, (National Institute on Aging, USA); SU 11248, (Pfizer, USA and SUGEN USA); ABT 518, (Abbott, USA); YH16, (Yantai Rongchang, China); S-3APG, (Boston Childrens Hospital, USA and EntreMed, USA); MAb, KDR, (ImClone Systems, USA); MAb, alpha5 beta1, (Protein Design, USA); KDR kinase inhibitor, (Celltech Group, UK, and Johnson & Johnson, USA); GFB 116, (South Florida University, USA and Yale University, USA); CS 706, (Sankyo, Japan); combretastatin A4 prodrug, (Arizona State University, USA); chondroitinase AC, (IBEX, Canada); BAY RES 2690, (Bayer, Germany); AGM 1470, (Harvard University, USA, Takeda, Japan, and TAP, USA); AG 13925, (Agouron, now Pfizer, USA); Tetrathiomolybdate, (University of Michigan, USA); GCS 100, (Wayne State University, USA) CV 247, (Ivy Medical, UK); CKD 732, (Chong Kun Dang, South Korea); MAb, vascular endothelium growth factor, (Xenova, UK); irsogladine (INN), (Nippon Shinyaku, Japan); RG 13577, (Aventis, France); WX 360, (Wilex, Germany); squalamine (plNN), (Genaera, USA); RPI 4610, (Sima, USA); cancer therapy, (Marinova, Australia); heparanase inhibitors, (InSight, Israel); KL 3106, (Kolon, South Korea); Honokiol, (Emory University, USA); ZK CDK, (Schering AG, Germany); ZK Angio, (Schering AG, Germany); ZK 229561, (Novartis, Switzerland, and Schering AG, Germany); XMP 300, (XOMA, USA); VGA 1102, (Taisho, Japan); VEGF receptor modulators, (Pharmacopeia, USA); VE-cadherin-2 antagonists, (ImClone Systems, USA); Vasostatin, (National Institutes of Health, USA); vaccine, Flk-1, (ImClone Systems, USA); TZ 93, (Tsumura, Japan); TumStatin, (Beth Israel Hospital, USA); truncated soluble FLT 1 (vascular endothelial growth factor receptor 1), (Merck & Co, USA); Tie-2 ligands, (Regeneron, USA); and, thrombospondin 1 inhibitor, (Allegheny Health, Education and Research Foundation, USA).


Autophagy inhibitors include, but are not limited to chloroquine, 3-methyladenine, hydroxychloroquine (Plaquenil™), bafilomycin A1, 5-amino-4-imidazole carboxamide riboside (AICAR), okadaic acid, autophagy-suppressive algal toxins which inhibit protein phosphatases of type 2A or type 1, analogues of cAMP, and drugs which elevate cAMP levels such as adenosine, LY204002, N6-mercaptopurine riboside, and vinblastine. In addition, antisense or siRNA that inhibits expression of proteins including but not limited to ATG5 (which are implicated in autophagy), may also be used.


Additional pharmaceutically active compounds/agents that can be used in the treatment of cancers and that can be used in combination with one or more compound of the present invention include: epoetin alfa; darbepoetin alfa; panitumumab; pegfilgrastim; palifermin; filgrastim; denosumab; ancestim; AMG 102; AMG 176; AMG 386; AMG 479; AMG 655; AMG 745; AMG 951; and AMG 706, or a pharmaceutically acceptable salt thereof.


In certain embodiments, a composition provided herein is conjointly administered with a chemotherapeutic agent. Suitable chemotherapeutic agents may include, natural products such as vinca alkaloids (e.g., vinblastine, vincristine, and vinorelbine), paclitaxel, epidipodophyllotoxins (e.g., etoposide and teniposide), antibiotics (e.g., dactinomycin (actinomycin D), daunorubicin, doxorubicin, and idarubicin), anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin), mitomycin, enzymes (e.g., L-asparaginase which systemically metabolizes L-asparagine and deprives cells which do not have the capacity to synthesize their own asparagine), antiplatelet agents, antiproliferative/antimitotic alkylating agents such as nitrogen mustards (e.g., mechlorethamine, cyclophosphamide and analogs, melphalan, and chlorambucil), ethylenimines and methylmelamines (e.g., hexaamethylmelaamine and thiotepa), CDK inhibitors (e.g., seliciclib, UCN-01, P1446A-05, PD-0332991, dinaciclib, P27-00, AT-7519, RGB286638, and SCH727965), alkyl sulfonates (e.g., busulfan), nitrosoureas (e.g., carmustine (BCNU) and analogs, and streptozocin), trazenes-dacarbazinine (DTIC), antiproliferative/antimitotic antimetabolites such as folic acid analogs (e.g., methotrexate), pyrimidine analogs (e.g., fluorouracil, floxuridine, and cytarabine), purine analogs and related inhibitors (e.g., mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine), aromatase inhibitors (e.g., anastrozole, exemestane, and letrozole), and platinum coordination complexes (e.g., cisplatin and carboplatin), procarbazine, hydroxyurea, mitotane, aminoglutethimide, histone deacetylase (HDAC) inhibitors (e.g., trichostatin, sodium butyrate, apicidan, suberoyl anilide hydroamic acid, vorinostat, LBH 589, romidepsin, ACY-1215, and panobinostat), mTor inhibitors (e.g., temsirolimus, everolimus, ridaforolimus, and sirolimus), KSP(Eg5) inhibitors (e.g., Array 520), DNA binding agents (e.g., Zalypsis), PI3K delta inhibitor (e.g., GS-1101 and TGR-1202), PI3K delta and gamma inhibitor (e.g., CAL-130), multi-kinase inhibitor (e.g., TG02 and sorafenib), hormones (e.g., estrogen) and hormone agonists such as leutinizing hormone releasing hormone (LHRH) agonists (e.g., goserelin, leuprolide and triptorelin), BAFF-neutralizing antibody (e.g., LY2127399), IKK inhibitors, p38MAPK inhibitors, anti-IL-6 (e.g., CNTO328), telomerase inhibitors (e.g., GRN 163L), aurora kinase inhibitors (e.g., MLN8237), cell surface monoclonal antibodies (e.g., anti-CD38 (HUMAX-CD38), anti-CS1 (e.g., elotuzumab), HSP90 inhibitors (e.g., 17 AAG and KOS 953), P13K/Akt inhibitors (e.g., perifosine), Akt inhibitor (e.g., GSK-2141795), PKC inhibitors (e.g., enzastaurin), FTIs (e.g., Zamestra™), anti-CD138 (e.g., BT062), Torcl/2 specific kinase inhibitor (e.g., INK128), kinase inhibitor (e.g., GS-1101), ER/UPR targeting agent (e.g., MKC-3946), cFMS inhibitor (e.g., ARRY-382), JAK1/2 inhibitor (e.g., CYT387), PARP inhibitor (e.g., olaparib and veliparib (ABT-888)), BCL-2 antagonist. Other chemotherapeutic agents may include mechlorethamine, camptothecin, ifosfamide, tamoxifen, raloxifene, gemcitabine, navelbine, sorafenib, or any analog or derivative variant of the foregoing.


The compounds of the present invention may also be used in combination with radiation therapy, hormone therapy, surgery and immunotherapy, which therapies are well known to those skilled in the art.


In certain embodiments, a pharmaceutical composition provided herein is conjointly administered with a steroid. Suitable steroids may include, but are not limited to, 21-acetoxypregnenolone, alclometasone, algestone, amcinonide, beclomethasone, betamethasone, budesonide, chloroprednisone, clobetasol, clocortolone, cloprednol, corticosterone, cortisone, cortivazol, deflazacort, desonide, desoximetasone, dexamethasone, diflorasone, diflucortolone, difuprednate, enoxolone, fluazacort, flucloronide, flumethasone, flunisolide, fluocinolone acetonide, fluocinonide, fluocortin butyl, fluocortolone, fluorometholone, fluperolone acetate, fluprednidene acetate, fluprednisolone, flurandrenolide, fluticasone propionate, formocortal, halcinonide, halobetasol propionate, halometasone, hydrocortisone, loteprednol etabonate, mazipredone, medrysone, meprednisone, methylprednisolone, mometasone furoate, paramethasone, prednicarbate, prednisolone, prednisolone 25-diethylaminoacetate, prednisolone sodium phosphate, prednisone, prednival, prednylidene, rimexolone, tixocortol, triamcinolone, triamcinolone acetonide, triamcinolone benetonide, triamcinolone hexacetonide, and salts and/or derivatives thereof. In a particular embodiment, the compounds of the present invention can also be used in combination with additional pharmaceutically active agents that treat nausea. Examples of agents that can be used to treat nausea include: dronabinol; granisetron; metoclopramide; ondansetron; and prochlorperazine; or a pharmaceutically acceptable salt thereof.


The compounds of the present invention may also be used in combination with an additional pharmaceutically active compound that disrupts or inhibits RAS-RAF-ERK or PI3K-AKT-TOR signaling pathways. In other such combinations, the additional pharmaceutically active compound is a PD-1 and PD-L1 antagonist. The compounds or pharmaceutical compositions of the disclosure can also be used in combination with an amount of one or more substances selected from EGFR inhibitors, MEK inhibitors, PI3K inhibitors, AKT inhibitors, TOR inhibitors, Mcl-1 inhibitors, BCL-2 inhibitors, SHP2 inhibitors, proteasome inhibitors, and immune therapies, including monoclonal antibodies, immunomodulatory imides (IMiDs), anti-PD-1, anti-PDL-1, anti-CTLA4, anti-LAG1, and anti-OX40 agents, GITR agonists, CAR-T cells, and BiTEs.


EGFR inhibitors include, but are not limited to, small molecule antagonists, antibody inhibitors, or specific antisense nucleotide or siRNA. Useful antibody inhibitors of EGFR include cetuximab (Erbitux), panitumumab (Vectibix), zalutumumab, nimotuzumab, and matuzumab. Small molecule antagonists of EGFR include gefitinib, erlotinib (Tarceva), and most recently, lapatinib (TykerB). See e.g., Yan L, et. al., Pharmacogenetics and Pharmacogenomics In Oncology Therapeutic Antibody Development, BioTechniques 2005; 39(4): 565-8, and Paez J G, et. al., EGFR Mutations In Lung Cancer Correlation With Clinical Response To Gefitinib Therapy, Science 2004; 304(5676): 1497-500.


Non-limiting examples of small molecule EGFR inhibitors include any of the EGFR inhibitors described in the following patent publications, and all pharmaceutically acceptable salts and solvates of said EGFR inhibitors: European Patent Application EP 520722, published Dec. 30, 1992; European Patent Application EP 566226, published Oct. 20, 1993; PCT International Publication WO 96/33980, published Oct. 31, 1996; U.S. Pat. No. 5,747,498, issued May 5, 1998; PCT International Publication WO 96/30347, published Oct. 3, 1996; European Patent Application EP 787772, published Aug. 6, 1997; PCT International Publication WO 97/30034, published Aug. 21, 1997; PCT International Publication WO 97/30044, published Aug. 21, 1997; PCT International Publication WO 97/38994, published Oct. 23, 1997; PCT International Publication WO 97/49688, published Dec. 31, 1997; European Patent Application EP 837063, published Apr. 22, 1998; PCT International Publication WO 98/02434, published Jan. 22, 1998; PCT International Publication WO 97/38983, published Oct. 23, 1997; PCT International Publication WO 95/19774, published Jul. 27, 1995; PCT International Publication WO 95/19970, published Jul. 27, 1995; PCT International Publication WO 97/13771, published Apr. 17, 1997; PCT International Publication WO 98/02437, published Jan. 22, 1998; PCT International Publication WO 98/02438, published Jan. 22, 1998; PCT International Publication WO 97/32881, published Sep. 12, 1997; German Application DE 19629652, published Jan. 29, 1998; PCT International Publication WO 98/33798, published Aug. 6, 1998; PCT International Publication WO 97/32880, published Sep. 12, 1997; PCT International Publication WO 97/32880 published Sep. 12, 1997; European Patent Application EP 682027, published Nov. 15, 1995; PCT International Publication WO 97/02266, published Jan. 23, 1997; PCT International Publication WO 97/27199, published Jul. 31, 1997; PCT International Publication WO 98/07726, published Feb. 26, 1998; PCT International Publication WO 97/34895, published Sep. 25, 1997; PCT International Publication WO 96/31510′, published Oct. 10, 1996; PCT International Publication WO 98/14449, published Apr. 9, 1998; PCT International Publication WO 98/14450, published Apr. 9, 1998; PCT International Publication WO 98/14451, published Apr. 9, 1998; PCT International Publication WO 95/09847, published Apr. 13, 1995; PCT International Publication WO 97/19065, published May 29, 1997; PCT International Publication WO 98/17662, published Apr. 30, 1998; U.S. Pat. No. 5,789,427, issued Aug. 4, 1998; U.S. Pat. No. 5,650,415, issued Jul. 22, 1997; U.S. Pat. No. 5,656,643, issued Aug. 12, 1997; PCT International Publication WO 99/35146, published Jul. 15, 1999; PCT International Publication WO 99/35132, published Jul. 15, 1999; PCT International Publication WO 99/07701, published Feb. 18, 1999; and PCT International Publication WO 92/20642 published Nov. 26, 1992. Additional non-limiting examples of small molecule EGFR inhibitors include any of the EGFR inhibitors described in Traxler, P., 1998, Exp. Opin. Ther. Patents 8(12): 1599-1625.


Antibody-based EGFR inhibitors include any anti-EGFR antibody or antibody fragment that can partially or completely block EGFR activation by its natural ligand. Non-limiting examples of antibody-based EGFR inhibitors include those described in Modjtahedi, H., et al., 1993, Br. J. Cancer 67:247-253; Teramoto, T., et al., 1996, Cancer 77:639-645; Goldstein et al., 1995, Clin. Cancer Res. 1:1311-1318; Huang, S. M., et al., 1999, Cancer Res. 15:59(8):1935-40; and Yang, X., et al., 1999, Cancer Res. 59:1236-1243. Thus, the EGFR inhibitor can be monoclonal antibody Mab E7.6.3 (Yang, 1999 supra), or Mab C225 (ATCC Accession No. HB-8508), or an antibody or antibody fragment having the binding specificity thereof.


MEK inhibitors include, but are not limited to, CI-1040, AZD6244, PD318088, PD98059, PD334581, RDEA119, ARRY-142886, ARRY-438162, and PD-325901.


PI3K inhibitors include, but are not limited to, wortmannin, 17-hydroxywortmannin analogs described in WO 06/044453, 4-[2-(1H-Indazol-4-yl)-6-[[4-(methylsulfonyl)piperazin-1-yl]methyl]thieno[3,2-d]pyrimidin-4-yl]morpholine (also known as GDC 0941 and described in PCT Publication Nos. WO 09/036,082 and WO 09/055,730), 2-Methyl-2-[4-[3-methyl-2-oxo-8-(quinolin-3-yl)-2,3-dihydroimidazo[4,5-c]quinolin-1-yl]phenyl]propionitrile (also known as BEZ 235 or NVP-BEZ 235, and described in PCT Publication No. WO 06/122806), (S)-1-(4-((2-(2-aminopyrimidin-5-yl)-7-methyl-4-morpholinothieno[3,2-d]pyrimidin-6-yl)methyl)piperazin-1-yl)-2-hydroxypropan-1-one (described in PCT Publication No. WO 2008/070740), LY294002 (2-(4-Morpholinyl)-8-phenyl-4H-1-benzopyran-4-one available from Axon Medchem), PI 103 hydrochloride (3-[4-(4-morpholinylpyrido-[3′,2′:4,5]furo[3,2-d]pyrimidin-2-yl]phenol hydrochloride available from Axon Medchem), PIK 75 (N′-[(1E)-(6-bromoimidazo[1,2-a]pyridin-3-yl)methylene]-N,2-dimethyl-5-nitrobenzenesulfono-hydrazide hydrochloride available from Axon Medchem), PIK 90 (N-(7,8-dimethoxy-2,3-dihydroimidazo[1,2-c]quinazolin-5-yl)-nicotinamide available from Axon Medchem), GDC-0941 bismesylate (2-(1H-Indazol-4-yl)-6-(4-methanesulfonyl-piperazin-1-ylmethyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidine bismesylate available from Axon Medchem), AS-252424 (5-[1-[5-(4-Fluoro-2-hydroxy-phenyl)-furan-2-yl]-meth-(Z)-ylidene]-thiazolidine-2,4-dione available from Axon Medchem), and TGX-221 (7-Methyl-2-(4-morpholinyl)-9-[1-(phenylamino)ethyl]-4H-pyrido-[1,2-a]pyrimidin-4-one available from Axon Medchem), XL-765, and XL-147. Other PI3K inhibitors include demethoxyviridin, perifosine, CAL101, PX-866, BEZ235, SF1126, INK1117, IPI-145, BKM120, XL147, XL765, Palomid 529, GSK1059615, ZSTK474, PWT33597, IC87114, TG100-115, CAL263, PI-103, GNE-477, CUDC-907, and AEZS-136.


AKT inhibitors include, but are not limited to, Akt-1-1 (inhibits Akt1) (Bamett et al. (2005) Biochem. J., 385 (Pt. 2), 399-408); Akt-1-1,2 (inhibits Ak1 and 2) (Barnett et al. (2005) Biochem. J. 385 (Pt. 2), 399-408); API-59CJ-Ome (e.g., Jin et al. (2004) Br. J. Cancer 91, 1808-12); 1-H-imidazo[4,5-c]pyridinyl compounds (e.g., WO05011700); indole-3-carbinol and derivatives thereof (e.g., U.S. Pat. No. 6,656,963; Sarkar and Li (2004) JNutr. 134(12 Suppl), 3493S-3498S); perifosine (e.g., interferes with Akt membrane localization; Dasmahapatra et al. (2004) Clin. Cancer Res. 10(15), 5242-52, 2004); phosphatidylinositol ether lipid analogues (e.g., Gills and Dennis (2004) Expert. Opin. Investig. Drugs 13, 787-97); and triciribine (TCN or API-2 or NCI identifier: NSC 154020; Yang et al. (2004) Cancer Res. 64, 4394-9).


TOR inhibitors include, but are not limited to, AP-23573, CCI-779, everolimus, RAD-001, rapamycin, temsirolimus, ATP-competitive TORC1/TORC2 inhibitors, including PI-103, PP242, PP30 and Torin 1. Other TOR inhibitors in FKBP12 enhancer; rapamycins and derivatives thereof, including: CCI-779 (temsirolimus), RAD001 (Everolimus; WO 9409010) and AP23573; rapalogs, e.g. as disclosed in WO 98/02441 and WO 01/14387, e.g. AP23573, AP23464, or AP23841; 40-(2-hydroxyethyl)rapamycin, 40-[3-hydroxy(hydroxymethyl)methylpropanoate]-rapamycin (also called CC1779), 40-epi-(tetrazolyt)-rapamycin (also called ABT578), 32-deoxorapamycin, 16-pentynyloxy-32(S)-dihydrorapanycin, and other derivatives disclosed in WO 05005434; derivatives disclosed in U.S. Pat. No. 5,258,389, WO 94/090101, WO 92/05179, U.S. Pat. Nos. 5,118,677, 5,118,678, 5,100,883, 5,151,413, 5,120,842, WO 93/111130, WO 94/02136, WO 94/02485, WO 95/14023, WO 94/02136, WO 95/16691, WO 96/41807, WO 96/41807 and U.S. Pat. No. 5,256,790; phosphorus-containing rapamycin derivatives (e.g., WO 05016252); 4H-1-benzopyran-4-one derivatives (e.g., U.S. Provisional Application No. 60/528,340).


MCl-1 inhibitors include, but are not limited to, AMG-176, MIK665, and S63845. The myeloid cell leukemia-1 (MCL-1) protein is one of the key anti-apoptotic members of the B-cell lymphoma-2 (BCL-2) protein family. Over-expression of MCL-1 has been closely related to tumor progression as well as to resistance, not only to traditional chemotherapies but also to targeted therapeutics including BCL-2 inhibitors such as ABT-263.


SHP inhibitors include, but are not limited to, SHP099.


Proteasome inhibitors include, but are not limited to, Kyprolis® (carfilzomib), Velcade® (bortezomib), and oprozomib.


Immune therapies include, but are not limited to, anti-PD-1 agents, anti-PDL-1 agents, anti-CTLA-4 agents, anti-LAG1 agents, and anti-OX40 agents.


Monoclonal antibodies include, but are not limited to, Darzalex® (daratumumab), Herceptin® (trastuzumab), Avastin® (bevacizumab), Rituxan® (rituximab), Lucentis® (ranibizumab), and Eylea® (aflibercept).


Immunomodulatory imide drugs (IMiDs) are a class of immunomodulatory drugs (drugs that adjust immune responses) containing an imide group. The IMiD class includes thalidomide and its analogues (lenalidomide, pomalidomide, and apremilast).


Exemplary anti-PD-1 antibodies and methods for their use are described by Goldberg et al., Blood 110(1):186-192 (2007), Thompson et al., Clin. Cancer Res. 13(6):1757-1761 (2007), and Korman et al., International Application No. PCT/JP2006/309606 (publication no. WO 2006/121168 A1), each of which are expressly incorporated by reference herein. include: Yervoy™ (ipilimumab) or Tremelimumab (to CTLA-4), galiximab (to B7.1), BMS-936558 (to PD-1), MK-3475 (pembrolizumab)(to PD-1), AMG 404 (to PD-1), AMP224 (to B7DC), BMS-936559 (nivolumab) (to B7-H1), MPDL3280A (to B7-H1), MEDI-570 (to ICOS), AMG557 (to B7H2), MGA271 (to B7H3), IMP321 (to LAG-3), BMS-663513 (to CD137), PF-05082566 (to CD137), CDX-1127 (to CD27), anti-OX40 (Providence Health Services), huMAbOX40L (to OX40L), Atacicept (to TACI), CP-870893 (to CD40), Lucatumumab (to CD40), Dacetuzumab (to CD40), Muromonab-CD3 (to CD3), Ipilumumab (to CTLA-4). Immune therapies also include genetically engineered T-cells (e.g., CAR-T cells) and bispecific antibodies (e.g., BiTEs).


GITR agonists include, but are not limited to, GITR fusion proteins and anti-GITR antibodies (e.g., bivalent anti-GITR antibodies), such as, a GITR fusion protein described in U.S. Pat. No. 6,111,090box.c, European Patent No.: 090505B1, U.S. Pat. No. 8,586,023, PCT Publication Nos.: WO 2010/003118 and 2011/090754, or an anti-GITR antibody described, e.g., in U.S. Pat. No. 7,025,962, European Patent No.: 1947183B1, U.S. Pat. Nos. 7,812,135, 8,388,967, 8,591,886, European Patent No.: EP 1866339, PCT Publication No.: WO 2011/028683, PCT Publication No.: WO 2013/039954, PCT Publication No.: WO2005/007190, PCT Publication No.: WO 2007/133822, PCT Publication No.: WO2005/055808, PCT Publication No.: WO 99/40196, PCT Publication No.: WO 2001/03720, PCT Publication No.: WO99/20758, PCT Publication No.: WO2006/083289, PCT Publication No.: WO 2005/115451, U.S. Pat. No. 7,618,632, and PCT Publication No.: WO 2011/051726.


The compounds described herein can be used in combination with the agents disclosed herein or other suitable agents, depending on the condition being treated. Hence, in some embodiments the one or more compounds of the disclosure will be co-administered with other agents as described above. When used in combination therapy, the compounds described herein are administered with the second agent simultaneously or separately. This administration in combination can include simultaneous administration of the two agents in the same dosage form, simultaneous administration in separate dosage forms, and separate administration. That is, a compound described herein and any of the agents described above can be formulated together in the same dosage form and administered simultaneously. Alternatively, a compound of the disclosure and any of the agents described above can be simultaneously administered, wherein both the agents are present in separate formulations. In another alternative, a compound of the present disclosure can be administered just followed by and any of the agents described above, or vice versa. In some embodiments of the separate administration protocol, a compound of the disclosure and any of the agents described above are administered a few minutes apart, or a few hours apart, or a few days apart.


As one aspect of the present invention contemplates the treatment of the disease/conditions with a combination of pharmaceutically active compounds that may be administered separately, the invention further relates to combining separate pharmaceutical compositions in kit form. The kit comprises two separate pharmaceutical compositions: a compound of the present invention, and a second pharmaceutical compound. The kit comprises a container for containing the separate compositions such as a divided bottle or a divided foil packet. Additional examples of containers include syringes, boxes, and bags. In some embodiments, the kit comprises directions for the use of the separate components. The kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing health care professional.


Synthesis of Disclosed Compounds

Compounds as disclosed herein can be synthesized via a number of specific methods. The examples which outline specific synthetic routes, and the generic schemes below are meant to provide guidance to the ordinarily skilled synthetic chemist, who will readily appreciate that the solvent, concentration, reagent, protecting group, order of synthetic steps, time, temperature, and the like can be modified as necessary, well within the skill and judgment of the ordinarily skilled artisan.


Section 1—General Procedures
Method 1
Example 1-1: 2-(5-Chloro-2-ethyl-3-((5-methoxy-3,4-dihydro-2,6-naphthyridin-2(1H)-yl)carbonyl)-7-methyl-1H-indol-1-yl)-N-(1-(2-propenoyl)-3-azetidinyl)acetamide



embedded image


embedded image


Step 1: Methyl 5-chloro-2-ethyl-7-methyl-1H-indole-3-carboxylate

Cesium carbonate (8.87 g, 27.2 mmol), 2-(1H-1,2,3,4-tetraazol-1-yl)acetic acid (Apollo Scientific Ltd., 0.35 g, 2.72 mmol), copper(I) iodide (0.26 g, 1.36 mmol) and 2-bromo-4-chloro-6-methylaniline (3.00 g, 13.6 mmol) were weighed into a 50-mL vial. The solids were purged with argon, treated with DMSO (15 mL) and methyl 3-oxopentanoate (6.83 mL, 54.4 mmol) and the vial sealed and heated to 80° C. for 24 h. The reaction mixture was cooled to rt, treated with water and extracted with EtOAc, washed with brine and dried over MgSO4, filtered and concentrated in vacuo. The crude residue was purified by silica gel chromatography (eluent: 0-40% EtOAc/heptane) affording methyl 5-chloro-2-ethyl-7-methyl-1H-indole-3-carboxylate (2.39 g, 9.50 mmol, 70% yield) as a light yellow crystalline solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 11.82 (1H, br s), 7.72 (1H, s), 6.99 (1H, s), 3.82 (3H, s), 3.09 (2H, q, J=7.5 Hz), 2.46-2.47 (3H, m), 1.26 (3H, t, J=7.5 Hz). m/z (ESI, +ve ion): 252.1 (M+H)+.


Step 2: Methyl 1-(2-(tert-butoxy)-2-oxoethyl)-5-chloro-2-ethyl-7-methyl-1H-indole-3-carboxylate

Methyl 5-chloro-2-ethyl-7-methyl-1H-indole-3-carboxylate (1.02 g, 4.05 mmol) in N,N-dimethylformamide (10 mL) was treated with sodium hydride (60 wt % in mineral oil, 0.32 g, 8.10 mmol) in small portions and allowed to stir at rt for 15 min. tert-Butylbromoacetate (1.00 mL, 6.08 mmol) was then added slowly dropwise and allowed to stir at rt for 1 h. The reaction mixture was treated with brine, extracted with EtOAc, washed with brine, dried over MgSO4, filtered and concentrated in vacuo. The crude residue was purified by silica gel chromatography (eluent: 0-40% EtOAc/heptane) affording methyl 1-(2-(tert-butoxy)-2-oxoethyl)-5-chloro-2-ethyl-7-methyl-1H-indole-3-carboxylate (1.20 g, 3.28 mmol, 81% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 7.87 (d, J=1.45 Hz, 1H), 7.00-7.05 (m, 1H), 5.21 (s, 2H), 3.85 (s, 3H), 3.10-3.19 (m, 2H), 2.54-2.62 (m, 3H), 1.43 (s, 9H), 1.12 (t, J=7.46 Hz, 3H). m/z (ESI, +ve ion): 366.2 (M+H)+.


Step 3: 2-(5-Chloro-2-ethyl-3-(methoxycarbonyl)-7-methyl-1H-indol-1-yl)acetic Acid TFA Salt

Methyl 1-(2-(tert-butoxy)-2-oxoethyl)-5-chloro-2-ethyl-7-methyl-1H-indole-3-carboxylate (1.20 g, 3.28 mmol) in DCM (5 mL) was treated with TFA (2.5 mL, 32.4 mmol) and allowed to stir at rt for 1 h. The reaction mixture was concentrated to dryness under reduced pressure affording 2-(5-chloro-2-ethyl-3-(methoxycarbonyl)-7-methyl-1H-indol-1-yl)acetic acid TFA salt as white solid. The crude material was used in the subsequent step without further purification. 1H NMR (400 MHz, DMSO-d6) δ ppm 7.86-7.89 (m, 1H), 7.00-7.03 (m, 1H), 5.22 (s, 2H), 3.85 (s, 3H), 3.14 (q, J=7.39 Hz, 2H), 2.58-2.64 (m, 3H), 1.13 (t, J=7.46 Hz, 3H). 19F NMR (376 MHz, DMSO-d6) δ ppm −74.53 (3F, s). m/z (ESI, +ve ion): 310.1 (M+H)+.


Step 4: Methyl 1-(2-((1-(tert-butoxycarbonyl)azetidin-3-yl)amino)-2-oxoethyl)-5-chloro-2-ethyl-7-methyl-1H-indole-3-carboxylate

2-(5-Chloro-2-ethyl-3-(methoxycarbonyl)-7-methyl-1H-indol-1-yl)acetic acid TFA salt (1.39 g, 3.28 mmol) in N,N-dimethylformamide (5 mL) was treated with diisopropylethylamine (2.3 mL, 13.1 mmol) and 1-Boc-3-aminoazetidine (Advanced ChemBlocks Inc., Burlingame, Calif., USA, 0.62 mL, 3.61 mmol) and the stirring solution was treated with T3P (50 wt. % in EtOAc, 3.91 mL, 6.56 mmol). The reaction mixture was then allowed to stir at rt for 1 h then treated with brine, extracted with EtOAc and washed with brine, dried over MgSO4, filtered and concentrated in vacuo. The crude residue was purified by silica gel chromatography (eluent: 0-70% EtOAc/heptane) affording methyl 1-(2-((1-(tert-butoxycarbonyl)azetidin-3-yl)amino)-2-oxoethyl)-5-chloro-2-ethyl-7-methyl-1H-indole-3-carboxylate (1.01 g, 2.18 mmol, 66% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 9.06 (1H, d, J=7.0 Hz), 7.95 (1H, s), 7.08 (1H, s), 5.16 (2H, s), 4.32-4.56 (m, 1H), 4.17 (2H, br t, J=7.9 Hz), 3.94 (3H, s), 3.72-3.88 (2H, m), 3.13-3.20 (2H, m), 2.66 (3H, s), 1.47 (9H, s), 1.16-1.31 (3H, m). m/z (ESI, +ve ion): 486.2 (M+Na)+.


Step 5: 1-(2-((1-(tert-Butoxycarbonyl)azetidin-3-yl)amino)-2-oxoethyl)-5-chloro-2-ethyl-7-methyl-1H-indole-3-carboxylic Acid

To a solution of methyl 1-(2-((1-(tert-butoxycarbonyl)azetidin-3-yl)amino)-2-oxoethyl)-5-chloro-2-ethyl-7-methyl-1H-indole-3-carboxylate (1.01 g, 2.18 mmol) in MeOH (20 mL) and THF (10 mL) was added 5 N sodium hydroxide (6.54 mL, 32.7 mmol). The resulting mixture was then heated at 90° C. for 4 h. The reaction mixture was concentrated under reduced pressure and cooled to 0° C. in an ice bath and slowly treated with 2.5 N hydrochloric acid (ca. 14 mL, 35 mmol). The resulting white suspension was collected by filtration affording crude 1-(2-((1-(tert-butoxycarbonyl)azetidin-3-yl)amino)-2-oxoethyl)-5-chloro-2-ethyl-7-methyl-1H-indole-3-carboxylic acid (771 mg, 1.71 mmol, 79% yield) as an off-white solid. The material was used in the subsequent step without further purification. m/z (ESI, +ve ion): 472.1 (M+Na)+.


Step 6: tert-Butyl 3-(2-(5-chloro-2-ethyl-3-(5-methoxy-1,2,3,4-tetrahydro-2,6-naphthyridine-2-carbonyl)-7-methyl-1H-indol-1-yl)acetamido)azetidine-1-carboxylate

1-(2-((1-(tert-Butoxycarbonyl)azetidin-3-yl)amino)-2-oxoethyl)-5-chloro-2-ethyl-7-methyl-1H-indole-3-carboxylic acid (250 mg, 0.56 mmol) and 5-methoxy-1,2,3,4-tetrahydro-2,6-naphthyridine hydrochloride (123 mg, 0.61 mmol) were treated with N,N-dimethylformamide (2.5 mL) and DIPEA (0.49 mL, 2.78 mmol) followed by HATU (264 mg, 0.70 mmol) in one portion. The reaction mixture was allowed to stir at rt for 1.5 h then the reaction mixture was treated with sat'd NaHCO3 and extracted with EtOAc, washed with brine and dried over MgSO4, filtered and concentrated in vacuo. The crude residue was purified by silica gel chromatography (eluent: 0-40% [20% MeOH in DCM]/DCM) affording tert-butyl 3-(2-(5-chloro-2-ethyl-3-(5-methoxy-1,2,3,4-tetrahydro-2,6-naphthyridine-2-carbonyl)-7-methyl-1H-indol-1-yl)acetamido)azetidine-1-carboxylate (227 mg, 0.38 mmol, 68% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 8.93 (1H, d, J=7.0 Hz), 7.90-7.97 (1H, m), 7.16 (1H, br s), 6.92 (1H, s), 6.70-6.84 (1H, m), 4.99 (2H, s), 4.56-4.78 (2H, m), 4.39-4.50 (1H, m), 4.08 (2H, br t, J=7.9 Hz), 3.87 (3H, s), 3.62-3.79 (3H, m), 2.62-2.75 (5H, m), 2.58 (3H, s), 1.38 (9H, s), 1.04 (3H, br t, J=7.4 Hz). m/z (ESI, +ve ion): 496.2 (M-100)+.


Step 7: N-(1-Acryloylazetidin-3-yl)-2-(5-chloro-2-ethyl-3-(5-methoxy-1,2,3,4-tetrahydro-2,6-naphthyridine-2-carbonyl)-7-methyl-1H-indol-1-yl)acetamide

tert-Butyl 3-(2-(5-chloro-2-ethyl-3-(5-methoxy-1,2,3,4-tetrahydro-2,6-naphthyridine-2-carbonyl)-7-methyl-1H-indol-1-yl)acetamido)azetidine-1-carboxylate (226 mg, 0.38 mmol) in DCM (3 mL) was treated with TFA (1 mL) and allowed to stir at rt for 25 min. The reaction mixture was concentrated in vacuo, and this crude reaction mixture was treated with DCM (3 mL), cooled in an ice bath then treated with DIPEA (0.33 mL, 1.90 mmol) and acryloyl chloride (0.046 mL, 0.57 mmol) and allowed to stir at 0° C. for 30 min. The reaction mixture was treated with sat'd NaHCO3, the reaction mixture extracted with DCM, dried over MgSO4, filtered and concentrated in vacuo. The crude residue was purified by silica gel chromatography (eluent: 0-50% [20% MeOH in DCM]/DCM) affording N-(1-acryloylazetidin-3-yl)-2-(5-chloro-2-ethyl-3-(5-methoxy-1,2,3,4-tetrahydro-2,6-naphthyridine-2-carbonyl)-7-methyl-1H-indol-1-yl)acetamide (118 mg, 0.22 mmol, 57% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 8.98 (1H, br d, J=6.4 Hz), 7.93 (1H, br d, J=4.8 Hz), 7.16 (1H, br s), 6.92 (1H, s), 6.76 (1H, br s), 6.32 (1H, dd, J=17.1, 10.3 Hz), 6.10 (1H, br d, J=17.0 Hz), 5.67 (1H, br d, J=10.4 Hz), 5.01 (2H, s), 4.66 (2H, br s), 4.45-4.58 (2H, m), 4.14-4.23 (1H, m), 4.02 (1H, br d, J=3.3 Hz), 3.87 (3H, s), 3.74-3.84 (2H, m), 3.69 (1H, br s), 2.60-2.76 (4H, m), 2.58 (3H, s), 1.04 (3H, br t, J=7.4 Hz). m/z (ESI, +ve ion): 550.3 (M+H)+.









TABLE 1







Compounds 1-2 to 1-7 were prepared following the procedure described in Method 1, Steps 1-7, above as follows:














Method



Ex. #
Chemical structure
Name
Changes
Reagent





1-2


embedded image


2-(5-Chloro-2- ethyl-3-((5- methoxy-3,4- dihydro-2(1H)- isoquinolinyl) carbonyl)-7- methyl-1H- indol-1-yl)-N- (1-(2-propenoyl)- 3- azetidinyl) acetamide
Use 5- methoxy- 1,2,3,4- tetrahydro- isoquinoline hydrochloride in Step 6
5-Methoxy- 1,2,3,4- tetrahydro- isoquinoline hydrochloride (J&W Pharmlab)





1-3


embedded image


2-(5-Chloro-3- (((2R)-4-(5- cyano-2- pyridinyl)-2- methyl-1- piperazinyl) carbonyl)- 2-ethyl-7- methyl-1H-indol- 1-yl)-N-(1-(2- propenoyl)-3- azetidinyl) acetamide
Use (R)-6-(3- methyl- piperazin-1- yl)nicotino- nitrile in Step 6
(R)-6-(3- Methyl- piperazin-1- yl)nicotino- nitrile (Ark Pharm, Inc.)





1-4


embedded image


2-(5-Chloro-2- cyclopropyl-3- ((5-methoxy-3,4- dihydro-2,6- naphthyridin- 2(1H)- yl)carbonyl)-7- methyl-1H-indol- 1-yl)-N-(1-(2- propenoyl)-3- azetidinyl) acetamide
Use methyl 3- cyclopropyl- 3- oxopropanoate in Step 1
Methyl 3- cyclopropyl- 3- oxopropanoate (Accela)





1-5


embedded image


2-(5-Chloro-2- (cyclopropyl- methyl)-3-((5- methoxy-3,4- dihydro-2(1H)- isoquinolinyl) carbonyl)-7- methyl- 1H-indol-1-yl)-N- (1-(2-propenoyl)- 3- azetidinyl) acetamide
Use ethyl 4- cyclopropyl- 3- oxobutanoate in Step 1 and 5-methoxy- 1,2,3,4- tetrahydro- isoquinoline hydrochloride in Step 6
Ethyl 4- cyclopropyl- 3- oxobutanoate (Life Chemicals) and 5- methoxy- 1,2,3,4- tetrahydro- isoquinoline hydrochloride (J&W Pharmlab)





1-6


embedded image


2-(5-Chloro-2- cyclopropyl-7- methyl-3-(5-oxa- 8- azaspiro[3.5] nonan-8- ylcarbonyl)-1H- indol-1-yl)-N- (1-(2-propenoyl)- 3- azetidinyl) acetamide
Use methyl 3- cyclopropyl- 3- oxopropanoate in Step 1 and 5-oxa-8- azaspiro[3.5] nonane and TBTU in Step 6
Methyl 3- cyclopropyl- 3-oxoprop- anoate (Accela) and 5-oxa-8- azaspiro[3.5] nonane (FSSI)





1-7


embedded image


2-(5-Chloro-2- cyclopropyl-3- ((5-methoxy-3,4- dihydro-2(1H)- isoquinolinyl) carbonyl)-7- methyl-1H- indol-1-yl)-N- (1-(2-propenoyl)- 3- azetidinyl) acetamide
Use methyl 3- cyclopropyl- 3- oxopropanoate in Step 1 and use 5- methoxy- 1,2,3,4- tetrahydro- isoquinoline hydrochloride in Step 6
Methyl 3- cyclopropyl- 3- oxopropanoate (Accela) and 5- methoxy- 1,2,3,4- tetrahydro- isoquinoline hydrochloride (J&W Pharmlab)









Method 2
Example 2-1: 2-(5-Bromo-3-((5-methoxy-3,4-dihydro-2(1H)-isoquinolinyl)carbonyl)-2,7-dimethyl-1H-indol-1-yl)-N-(1-(2-propenoyl)-3-azetidinyl)acetamide



embedded image


embedded image


Step 1: Methyl 5-bromo-2,7-dimethyl-1H-indole-3-carboxylate

Carried out using the conditions in Example 1, Step 1 with 2,4 dibromo-6-methylaniline (Oakwood Products, Inc., Estill, N.C., USA, 4.50 mL, 16.98 mmol) to provide methyl 5-bromo-2,7-dimethyl-1H-indole-3-carboxylate (2.91 g, 10.3 mmol, 61% yield) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) □: 11.88 (br s, 1H), 7.86 (s, 1H), 7.10 (s, 1H), 3.81, (s, 3H), 2.66 (s, 3H), 2.46 (s, 3H); m/z (ESI, +ve ion): 282.0, 284.0 (M+H)+.


Step 2: Methyl 5-bromo-1-(2-(tert-butoxy)-2-oxoethyl)-2,7-dimethyl-1H-indole-3-carboxylate

To a solution of methyl 5-bromo-2,7-dimethyl-1H-indole-3-carboxylate (500 mg, 1.77 mmol) in N,N-dimethylformamide (5 mL) was added cesium carbonate (1.16 g, 3.54 mmol) and tert-butylbromoacetate (0.58 mL, 3.54 mmol). The resulting mixture was then subjected to a microwave irradiation at 120° C. for 30 min. The mixture was diluted with H2O and was extracted with EtOAc. The organic layer was washed with brine, dried over MgSO4, and concentrated in vacuo. The crude residue was purified by silica gel chromatography (eluent: 0%-100% EtOAc/heptane) affording methyl 5-bromo-1-(2-(tert-butoxy)-2-oxoethyl)-2,7-dimethyl-1H-indole-3-carboxylate as yellow solid that was used in the next step. m/z (ESI, +ve ion): 396.0, 398.0 (M+H)+.


Step 3: 2-(5-Bromo-3-(methoxycarbonyl)-2,7-dimethyl-1H-indol-1-yl)acetic Acid

To a solution of methyl 5-bromo-1-(2-(tert-butoxy)-2-oxoethyl)-2,7-dimethyl-1H-indole-3-carboxylate (702 mg, 1.77 mmol) in DCM (4 mL) was added hydrochloric acid (4.0 N in dioxane, 4.43 mL, 17.71 mmol). After the addition, the mixture was stirred at rt for 16 h. The mixture was then diluted with H2O and was extracted with EtOAc. The combined organic extracts were then washed with brine, dried over MgSO4, filtered, and concentrated in vacuo. The crude residue was purified by silica gel chromatography (eluent: 0%-100% EtOAc/heptane) affording 2-(5-bromo-3-(methoxycarbonyl)-2,7-dimethyl-1H-indol-1-yl)acetic acid (599 mg, 1.76 mmol, 99% yield) as a yellow solid. m/z (ESI, +ve ion): 340.0, 342.0 (M+H)+.


Step 4: Methyl 5-bromo-1-(2-((1-(tert-butoxycarbonyl)azetidin-3-yl)amino)-2-oxoethyl)-2,7-dimethyl-1H-indole-3-carboxylate

To a solution of 2-(5-bromo-3-(methoxycarbonyl)-2,7-dimethyl-1H-indol-1-yl)acetic acid (599 mg, 1.76 mmol) and tert-butyl 3-aminoazetidine-1-carboxylate (607 mg, 3.52 mmol) in an ice bath at 0° C. was added propylphosphonic anhydride solution, 50 wt. % in DMF (3.14 mL, 5.28 mmol). After the addition, the mixture was then stirred at 0° C. for 5 min and at rt for 16 h. Then, DMF (5 mL), propylphosphonic anhydride solution, 50 wt. % in DMF (1.4 mL), and tert-butyl 3-aminoazetidine-1-carboxylate (607 mg, 3.52 mmol) were added and the mixture became clear. The resulting mixture was then stirred at rt for 4 h. The mixture was then quenched with sat'd NaHCO3 and then extracted with EtOAc. The organic extract was dried over Na2SO4 and concentrated in vacuo. The crude material was purified by silica gel chromatography (elutent: 0%-10% MeOH/DCM) affording methyl 5-bromo-1-(2-((1-(tert-butoxycarbonyl)azetidin-3-yl)amino)-2-oxoethyl)-2,7-dimethyl-1H-indole-3-carboxylate (871 mg, 1.76 mmol, 100% yield) as a light yellow solid. m/z (ESI, +ve ion): 516.0, 518.0 (M+Na)+.


Step 5: 5-Bromo-1-(2-((1-(tert-butoxycarbonyl)azetidin-3-yl)amino)-2-oxoethyl)-2,7-dimethyl-1H-indole-3-carboxylic Acid

To a solution of methyl 5-bromo-1-(2-((1-(tert-butoxycarbonyl)azetidin-3-yl)amino)-2-oxoethyl)-2,7-dimethyl-1H-indole-3-carboxylate (851 mg, 1.72 mmol) in methanol (20 mL) and water (5.0 mL) was added sodium hydroxide (1.38 g, 34.4 mmol). The resulting mixture was subjected to microwave irradiation at 80° C. for 2 h. The mixture was concentrated and H2O (40 mL) was added. The mixture was then adjusted to pH 2 using 5 N HCl solution. The mixture was extracted with EtOAc. The combined organic extracts were dried over MgSO4 and concentrated in vacuo. Then, DCM was added to the mixture and a white precipitate was observed. The mixture was filtered and the white solid was collected and dried in vacuo affording 5-bromo-1-(2-((1-(tert-butoxycarbonyl)azetidin-3-yl)amino)-2-oxoethyl)-2,7-dimethyl-1H-indole-3-carboxylic acid (540 mg, 1.12 mmol, 65% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.94 (br d, J=6.8 Hz, 1H), 8.05 (s, 1H), 7.06 (s, 1H), 5.02 (s, 2H), 4.44 (br d, J=7.2 Hz, 1H), 4.07 (br t, J=7.8 Hz, 2H), 3.66-3.73 (m, 2H), 2.62 (s, 3H), 2.56 (s, 3H), 1.37 (s, 9H). m/z (ESI, +ve ion): 502.1, 504.0 (M+Na)+.


Step 6: tert-Butyl 3-(2-(5-bromo-3-(5-methoxy-1,2,3,4 tetrahydroisoquinoline-2-carbonyl)-2,7-dimethyl-1H-indol-1-yl)acetamido)azetidine-1-carboxylate

To a solution of 5-bromo-1-(2-((1-(tert-butoxycarbonyl)azetidin-3-yl)amino)-2-oxoethyl)-2,7-dimethyl-1H-indole-3-carboxylic acid (104 mg, 0.22 mmol) in DCM (2 mL) and N,N-dimethylformamide (1 mL) was added 5-methoxy-1,2,3,4-tetrahydroisoquinoline hydrochloride (J&W Pharmlab LLC., 108 mg, 0.54 mmol), propylphosphonic anhydride solution, 50 wt. % in DMF (0.64 mL, 1.08 mmol) followed by DIPEA (0.19 mL, 1.08 mmol). The resulting mixture was stirred at 50° C. for 16 h. The mixture was quenched with sat'd NaHCO3(30 mL) and was extracted with EtOAc. The organic layer was dried over Na2SO4, filtered, and concentrated in vacuo. The crude residue was purified by silica gel chromatography (elutent: 0%-100% EtOAc/heptane) affording tert-butyl 3-(2-(5-bromo-3-(5-methoxy-1,2,3,4-tetrahydroisoquinoline-2-carbonyl)-2,7-dimethyl-1H-indol-1-yl)acetamido)azetidine-1-carboxylate as light yellow solid that was used in the next step. m/z (ESI, +ve ion): 625.0 (M+H)+.


Step 7: N-(1-Acryloylazetidin-3-yl)-2-(5-bromo-3-(5-methoxy-1,2,3,4-tetrahydroisoquinoline-2-carbonyl)-2,7-dimethyl-1H-indol-1-yl)acetamide

To a solution of tert-butyl 3-(2-(5-bromo-3-(5-methoxy-1,2,3,4-tetrahydroisoquinoline-2-carbonyl)-2,7-dimethyl-1H-indol-1-yl)acetamido)azetidine-1-carboxylate (135 mg, 0.22 mmol) in DCM (2 mL) was added trifluoroacetic acid (0.61 mL, 8.20 mmol). After the addition, the mixture was stirred at rt for 30 min. The mixture was concentrated and dried in vacuo. The residue was dissolved in DCM (4 mL) and was cooled to −78° C. under N2. Then, potassium carbonate (0.52 mL, 8.60 mmol) was added to the solution followed by acryloyl chloride (0.035 mL, 0.43 mmol). After the addition, the mixture was stirred at 0° C. for 30 min. Acryloyl chloride (0.035 mL, 0.430 mmol) and DIPEA (0.19 mL, 1.08 mmol) were added. The mixture was stirred at rt for 20 min. Then, H2O was added and the mixture was extracted with EtOAc. The organic layer was collected, washed with brine, dried over Na2SO4, and concentrated in vacuo. The crude residue was purified by silica gel chromatography (eluent: 0%-15% MeOH/DCM) affording N-(1-acryloylazetidin-3-yl)-2-(5-bromo-3-(5-methoxy-1,2,3,4-tetrahydroisoquinoline-2-carbonyl)-2,7-dimethyl-1H-indol-1-yl)acetamide (74 mg, 0.13 mmol, 59% yield) as an off-white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.98 (br d, J=6.7 Hz, 1H), 7.32 (s, 1H), 7.15 (br t, J=7.4 Hz, 1H), 7.03 (s, 1H), 6.82 (d, J=8.0 Hz, 1H), 6.58-6.78 (m, 1H), 6.32 (dd, J=16.9, 10.3 Hz, 1H), 6.10 (dd, J=16.8, 2.0 Hz, 1H), 5.67 (dd, J=10.5, 1.9 Hz, 1H), 5.00 (s, 2H), 4.43-4.72 (m, 4H), 4.18 (br t, J=8.9 Hz, 1H), 3.98-4.07 (m, 1H), 3.66-3.87 (m, 6H), 2.67 (br s, 2H), 2.57 (s, 3H), 2.29 (s, 3H). m/z (ESI, +ve ion): 579.0, 581.2 (M+H)+.









TABLE 2







Compounds 2-2 to 2-3 were prepared following the procedure described in Method 2, Steps 1-7, above as follows:














Method



Ex. #
Chemical structure
Name
Changes
Reagent





2-2


embedded image


2-(5-Bromo-2,7-dimethyl-3- (((1R)-1-methyl-3,4-dihydro- 2(1H)-isoquinolinyl) carbonyl)-1H-indol-1-yl)-N- (1-(2-propenoyl)-3-azetidinyl) acetamide|2-(5-bromo-2,7- dimethyl-3-(((1S)-1-methyl- 3,4-dihydro-2(1H)- isoquinolinyl)carbonyl)-1H- indol-1-yl)-N-(1-(2- propenoyl)-3-azetidinyl) acetamide
Use 1-methyl- 1,2,3,4- tetrahydo- isoquinoline hydrochloride in Step 6
1-methyl- 1,2,3,4- tetrahydo- isoquinoline hydrochloride (Enamine, Monmouth Junction, NJ, USA)








embedded image










2-3


embedded image

embedded image

2-(5-Bromo-3-(((1R,3R)-6- methoxy-1,3-dimethyl-3,4- dihydro-2(1H)-isoquinolinyl) carbonyl)-2,7-dimethyl-1H-indol- 1-yl)-N-(1-(2-propenoyl)-3- azetidinyl)acetamide|2-(5- bromo-3-(((1R,3S)-6-methoxy- 1,3-dimethyl-3,4-dihydro- 2(1H)-isoquinolinyl)carbonyl)- 2,7-dimethyl-1H-indol-1- yl)-N-(1-(2-propenoyl)-3- azetidinyl)acetamide|2-(5- bromo-3-(((1S,3R)-6-methoxy- 1,3-dimethyl-3,4-dihydro- 2(1H)-isoquinolinyl)carbonyl)- 2,7-dimethyl-1H-indol-1-yl)- N-(1-(2-propenoyl)-3- azetidinyl)acetamide|2-(5- bromo-3-(((1S,3S)-6-methoxy- 1,3-dimethyl-3,4-dihydro-2(1H)- isoquinolinyl)carbonyl)-2,7- dimethyl-1H-indol-1-yl)-N-(1- (2-propenoyl)-3-azetidinyl) acetamide
Use 6- methoxy-1,3- dimethyl- 1,2,3,4- tetrahydro- isoquinoline hydrochloride in Step 6
6-methoxy- 1,3-dimethyl- 1,2,3,4- tetrahydro- isoquinoline hydrochloride (FSSI)








embedded image













embedded image











Method 3
Example 3-1: 2-(5-Chloro-3-((5-methoxy-3,4-dihydro-2(1H)-isoquinolinyl)carbonyl)-2,7-dimethyl-1H-indol-1-yl)-N-(1-(2-propenoyl)-3-azetidinyl)acetamide



embedded image


Step 1: Methyl 5-chloro-2,7-dimethyl-1H-indole-3-carboxylate

A mixture of cesium carbonate (1478 mg, 4.54 mmol), 2-(1H-1,2,3,4-tetraazol-1-yl)acetic acid (Apollo Scientific Ltd., 58.1 mg, 0.454 mmol), copper(i) iodide (43.2 mg, 0.227 mmol) and 2-bromo-4-chloro-6-methylaniline (Oakwood Products, Inc., 500 mg, 2.268 mmol) was purged with argon and treated with acetoacetic acid methyl ester (1.476 mL, 13.61 mmol), the vial sealed and heated to 95° C. overnight. The reaction mixture was treated with water and extracted with EtOAc, washed with brine, dried over MgSO4, filtered and concentrated in vacuo. The crude residue was purified by silica gel chromatography (eluent: 0-60% EtOAc/heptane) affording methyl 5-chloro-2,7-dimethyl-1H-indole-3-carboxylate (279.6 mg, 1.176 mmol, 51.9% yield) as an off-white amorphous solid. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 8.21 (1H, br s), 7.90 (1H, s), 6.99 (1H, s), 3.93 (3H, s), 2.76 (3H, s), 2.45 (3H, s). m/z (ESI, +ve ion): 238.1 (M+H)+.


Step 2: 5-Chloro-2,7-dimethyl-1H-indole-3-carboxylic Acid

Methyl 5-chloro-2,7-dimethyl-1H-indole-3-carboxylate (128 mg, 0.54 mmol) in MeOH (1.0 mL) and THF (1.0 mL) was treated with 5 N sodium hydroxide (0.54 mL, 2.69 mmol) and the reaction mixture allowed to stir at 60° C. for 3-d. The reaction mixture was treated with 5 N HCl (0.6 mL) and extracted with EtOAc (25 mL), washed with brine and dried over MgSO4, filtered and concentrated affording crude 5-chloro-2,7-dimethyl-1H-indole-3-carboxylic acid (108 mg, 0.48 mmol, 89% yield) as a light yellow solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 12.02 (1H, s), 11.76 (1H, br s), 7.73 (1H, d, J=1.8 Hz), 6.95 (1H, s), 2.64-2.66 (3H, s), 2.36-2.46 (3H, s). m/z (ESI, +ve ion): 224.0 (M+H)+.


Step 3: (5-Chloro-2,7-dimethyl-1H-indol-3-yl)(5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl)methanone

5-Methoxy-1,2,3,4-tetrahydroisoquinoline hydrochloride (J&W Pharmlab LLC., 105 mg, 0.53 mmol) and 5-chloro-2,7-dimethyl-1H-indole-3-carboxylic acid (107 mg, 0.48 mmol) were treated with N,N-dimethylformamide (3.0 mL) and Hunig's base (0.50 mL, 2.87 mmol) followed by HATU (218 mg, 0.57 mmol) in one portion. The reaction mixture was then allowed to stir at rt for 3.5 h. The reaction mixture was treated with a sat'd solution of NaHCO3, extracted with EtOAc (30 mL) and washed with brine (2×20 mL), dried over MgSO4, filtered and concentrated. The crude residue was purified by silica gel chromatography (eluent: 0-100% EtOAc/heptane) affording (5-chloro-2,7-dimethyl-1H-indol-3-yl)(5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl)methanone (71.3 mg, 0.19 mmol, 40% yield) as a light yellow tar. 1H NMR (400 MHz, CDCl3) δ ppm 8.10 (1H, br s), 7.14 (1H, br t, J=7.6 Hz), 6.94 (1H, s), 6.71 (1H, d, J=8.0 Hz), 6.65 (1H, br s), 4.76 (2H, br s), 3.82 (3H, s), 3.74-3.90 (3H, m), 2.73-2.91 (2H, m), 2.49 (3H, s), 2.44 (3H, s). m/z (ESI, +ve ion): 369.1 (M+H)+.


Step 4: N-(1-Acryloylazetidin-3-yl)-2-(5-chloro-3-(5-methoxy-1,2,3,4-tetrahydroisoquinoline-2-carbonyl)-2,7-dimethyl-1H-indol-1-yl)acetamide

(5-Chloro-2,7-dimethyl-1H-indol-3-yl)(5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl)methanone (71 mg, 0.19 mmol) in THF (3 mL) was treated with sodium hydride (60 wt % in mineral oil, 24 mg, 0.62 mmol) and allowed to stir at rt for 15 min. The reaction mixture was treated with N-(1-acryloylazetidin-3-yl)-2-bromoacetamide (Intermediate A, 138 mg, 0.21 mmol) in a single portion and allowed to stir at rt for 7 h. The reaction mixture was then quenched with a sat'd solution of NaHCO3, extracted with EtOAc (30 mL) washed with brine and dried over MgSO4, filtered and concentrated. The crude residue was purified by silica gel chromatography (eluent: 0-50% [20% MeOH in DCM]/DCM) affording N-(1-acryloylazetidin-3-yl)-2-(5-chloro-3-(5-methoxy-1,2,3,4-tetrahydroisoquinoline-2-carbonyl)-2,7-dimethyl-1H-indol-1-yl)acetamide (9.2 mg, 0.017 mmol, 9% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 8.98 (1H, br d, J=6.7 Hz), 7.18 (1H, s), 7.14 (1H, br t, J=7.8 Hz), 6.91 (1H, s), 6.82 (1H, d, J=8.0 Hz), 6.58-6.74 (1H, m), 6.32 (1H, dd, J=17.0, 10.4 Hz), 6.10 (1H, dd, J=16.9, 2.2 Hz), 5.67 (1H, dd, J=10.4, 2.2 Hz), 5.00 (2H, s), 4.66 (2H, br s), 4.45-4.61 (2H, m), 4.14-4.22 (1H, m), 4.03 (1H, br dd, J=7.7, 4.8 Hz), 3.75-3.82 (4H, m), 3.57-3.75 (2H, m), 2.62-2.73 (2H, m), 2.54-2.61 (3H, m), 2.29 (3H, s). m/z (ESI, +ve ion): 535.3 (M+H)+.









TABLE 3







Compound 3-2 was prepared following the procedure described in Method 3, Steps 1-4, above as follows:














Method



Ex. #
Chemical structure
Name
Changes
Reagent





3-2


embedded image


2-(5-Bromo-2- cyclopropyl-3- ((5-methoxy-3,4- dihydro-2(1H)- isoquinolinyl) carbonyl)-1H- indol-1-yl)-N-(1- (2-propenoyl)-3- azetidinyl) acetamide
Use 4-bromo- 2-iodoaniline and tert-butyl 3- cyclopropyl- 3-oxo- propaoate in Step 1, Use TFA/DCM in Step 2
4-bromo-2- iodoaniline (Sigma- Aldrich Corporation) and tert-butyl 3-cyclo- propyl-3- oxopropanoate (Enamine)









Method 4
Example 4-1: 2-(5-Chloro-2-ethyl-3-((5-methoxy-3,4-dihydro-2,6-naphthyridin-2(1H)-yl)carbonyl)-7-methyl-1H-indol-1-yl)-N-methyl-N-(1-(2-propenoyl)-3-azetidinyl)acetamide



embedded image


Step 1: N-(1-Acryloylazetidin-3-yl)-2-(5-chloro-2-ethyl-3-(5-methoxy-1,2,3,4-tetrahydro-2,6-naphthyridine-2-carbonyl)-7-methyl-1H-indol-1-yl)-N-methylacetamide

N-(1-Acryloylazetidin-3-yl)-2-(5-chloro-2-ethyl-3-(5-methoxy-1,2,3,4-tetrahydro-2,6-naphthyridine-2-carbonyl)-7-methyl-1H-indol-1-yl)acetamide (61 mg, 0.11 mmol) in THF (3 mL) at rt was treated with iodomethane (0.035 mL, 0.55 mmol) and sodium hydride (60 wt % in mineral oil, 5.8 mg, 0.14 mmol) and allowed to stir at rt for 30 min. The reaction mixture was quenched with water, extracted with EtOAc (20 mL), washed with brine and dried over MgSO4, filtered and concentrated. The crude residue was purified by silica gel chromatography (eluent: 0-40% [20% MeOH in DCM]/DCM) affording N-(1-acryloylazetidin-3-yl)-2-(5-chloro-2-ethyl-3-(5-methoxy-1,2,3,4-tetrahydro-2,6-naphthyridine-2-carbonyl)-7-methyl-1H-indol-1-yl)-N-methylacetamide (47.7 mg, 0.085 mmol, 76% yield) as a white amorphous solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 7.93 (1H, br d, J=5.2 Hz), 7.15 (1H, s), 6.90 (1H, s), 6.77 (1H, br s), 6.25-6.47 (1H, m), 6.03-6.22 (1H, m), 5.62-5.76 (2H, m), 5.35 (1H, br s), 5.28 (1H, br s), 4.95-5.21 (2H, m), 4.66 (2H, br s), 4.50-4.62 (1H, m), 4.36-4.50 (1H, m), 4.30 (1H, br d, J=9.1 Hz), 3.94-4.20 (3H, m), 3.87 (3H, s), 3.78 (1H, br s), 3.53-3.74 (2H, m), 2.68-2.76 (1H, m), 2.61-2.66 (1H, m), 2.34-2.46 (2H, m), 2.22-2.32 (1H, m), 1.01 (3H, br t, J=7.4 Hz) two rotomers observed. m/z (ESI, +ve ion): 564.2 (M+H)+.









TABLE 4







Compounds 4-2 to 4-6 were prepared following the procedure described in Method 4, above as follows:














Method



Ex. #
Chemical structure
Name
Changes
Reagent





4-2


embedded image


2-(5-Chloro-2-ethyl-3- ((5-methoxy-3,4-dihydro- 2(1H)-isoquinolinyl) carbonyl)-7-methyl-1H- indol-1-yl)-N-methyl- N-(1-(2-propenoyl)-3- azetidinyl)acetamide
Using the product of Example 1-2






4-3


embedded image


2-(5-Chloro-2-cyclopropyl- 3-((5-methoxy-3,4- dihydro-2(1H)- isoquinolinyl)carbonyl)- 7-methyl-1H-indol-1- yl)-N-methyl-N-(1-(2- propenoyl)-3- azetidinyl)acetamide
Using the product of Example 1-7






4-4


embedded image


2-(5-Chloro-2- (cyclopropylmethyl)- 3-((5-methoxy-3,4- dihydro-2(1H)- isoquinolinyl) carbonyl)-7-methyl- 1H-indol-1-yl)-N- methyl-N-(1-(2- propenoyl)-3- azetidinyl)acetamide
Using the product of Example 1-5






4-5


embedded image


2-(5-Bromo-3-((5- methoxy-3,4-dihydro- 2(1H)-isoquinolinyl) carbonyl)-2,7- dimethyl-1H-indol- 1-yl)-N-methyl-N- (1-(2-propenoyl)- 3-azetidinyl)acetamide
Using the product of Example 2-1






4-6


embedded image


2-(5-Chloro-3-(((2R)- 4-(5-cyano-2-pyridinyl)- 2-methyl-1-piperazinyl) carbonyl)-2-ethyl-7- methyl-1H-indol-1-yl)- N-methyl-N-(1-(2- propenoyl)-3- azetidinyl)acetamide
Using the product of Example 1-3










Method 5
Example 5-1: 2-(5-Chloro-2-methyl-3-(4-(2-propanyl)-3-pyridinyl)-1H-indol-1-yl)-N-(1-(2-propenoyl)-3-azetidinyl)acetamide



embedded image


Step 1: N-(1-Acryloylazetidin-3-yl)-2-(5-chloro-3-iodo-2-methyl-1H-indol-1-yl)acetamide

A solution of 5-chloro-3-iodo-2-methyl-1H-indole (2.0 g, 6.86 mmol, Sigma-Aldrich Corporation) in THF (40 mL) was cooled to 0° C. and treated with sodium hydride (60 wt % in mineral oil, 0.82 g, 20.6 mmol) and allowed to stir at rt for 30 min. The reaction mixture was treated with N-(1-acryloylazetidin-3-yl)-2-bromoacetamide (Intermediate A, 1.87 g, 7.55 mmol) and stirred at rt for 2 h. The reaction mixture was then quenched with a sat'd solution of NaHCO3, extracted with EtOAc washed with brine and dried over MgSO4, filtered and concentrated. The crude residue was purified by silica gel chromatography (eluent: 0-50% acetone/heptane) affording N-(1-acryloylazetidin-3-yl)-2-(5-chloro-3-iodo-2-methyl-1H-indol-1-yl)acetamide (1.85 g, 4.04 mmol, 59% yield) as a brown solid. 1H NMR (400 MHz, CDCl3) δ ppm 7.43 (d, J=1.8 Hz, 1H), 7.22 (dd, J=8.6, 1.8 Hz, 1H), 7.09 (d, J=8.6 Hz, 1H), 6.26 (dd, J=17.0, 1.2 Hz, 1H), 6.01-6.11 (m, 1H), 5.84 (br d, J=6.7 Hz, 1H), 5.66 (dd, J=10.3, 1.3 Hz, 1H), 4.79 (s, 2H), 4.65-4.75 (m, 1H), 4.46 (br t, J=8.1 Hz, 1H), 4.31 (br t, J=9.6 Hz, 1H), 3.81-3.89 (m, 1H), 3.70 (br dd, J=5.5, 4.5 Hz, 1H), 2.48 (s, 3H). m/z (ESI, +ve ion): 458.0 (M+H)+.


Step 2: 2-(5-Chloro-2-methyl-3-(4-(2-propanyl)-3-pyridinyl)-1H-indol-1-yl)-N-(1-(2-propenoyl)-3-azetidinyl)acetamide

A mixture of N-(1-acryloylazetidin-3-yl)-2-(5-chloro-3-iodo-2-methyl-1H-indol-1-yl)acetamide (0.040 g, 0.087 mmol), 4-(iso-propyl)pyridine-3-boronic acid (CombiPhos Catalysts, 0.043 g, 0.26 mmol), potassium phosphate (0.074 g, 0.350 mmol), tetrakis(triphenylphosphine)palladium (0.020 g, 0.017 mmol), 1,4-dioxane (0.5 mL), and water (0.13 mL) was heated at 80° C. for 1 h. The reaction mixture was cooled to rt and partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc. The combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo. The crude residue was purified by silica gel chromatography (eluent: DCM:EtOAc:MeOH=75%:22%:3%) affording 2-(5-chloro-2-methyl-3-(4-(2-propanyl)-3-pyridinyl)-1H-indol-1-yl)-N-(1-(2-propenoyl)-3-azetidinyl)acetamide (0.005 g, 0.011 mmol, 13% yield) as a white solid. 1H NMR (400 MHz, CDCl3) δ ppm 8.52-8.65 (m, 1H), 8.36 (br s, 1H), 7.37 (br d, J=3.1 Hz, 1H), 7.17-7.23 (m, 3H), 6.24 (br dd, J=16.7, 8.8 Hz, 1H), 5.95-6.13 (m, 2H), 5.66 (br d, J=10.6 Hz, 1H), 4.83 (s, 2H), 4.68-4.80 (m, 1H), 4.49 (br t, J=8.1 Hz, 1H), 4.35 (br t, J=8.3 Hz, 1H), 3.80-3.91 (m, 1H), 3.65-3.78 (m, 1H), 2.84 (dt, J=13.7, 7.0 Hz, 1H), 2.25 (s, 3H), 1.16 (d, J=6.8 Hz, 3H), 1.08 (d, J=7.0 Hz, 3H). m/z (ESI, +ve ion): 451.2 (M+H)+.









TABLE 5







Compounds 5-2 to 5-4 were prepared following the procedure described in Method 5, Steps 1-2, above as follows:














Method



Ex. #
Chemical structure
Name
Changes
Reagent





5-2


embedded image


2-(5-Chloro-2- methyl-3-(4-(2- propanyl)-5- pyrimidinyl)-1H- indol-1-yl)-N-(1- (2-propenoyl)-3- azetidinyl)acetamide
Use (4- isopropyl-5- pyrimidinyl) boronic acid in Step 2
(4-Isopropyl- 5-pyrimidinyl) boronic acid (Combi- Blocks Inc.)





5-3


embedded image


2-(5-Chloro-2- methyl-3-(2-(2- propanyl)phenyl)- 1H-indol-1-yl)-N- (1-(2-propenoyl)-3- azetidinyl)acetamide
Use 2- isopropylphenyl- boronic acid in Step 2
2- Isopropyl- phenylboronic acid (Alfa Aesar)





5-4


embedded image


2-(5-Chloro-2- methyl-3-(2-(1- pyrrolidinyl)phenyl)- 1H-indol-1-yl)-N-(1- (2-propenoyl)-3- azetidinyl)acetamide
Use (2- (pyrrolidin-1- yl)phenyl)boronic acid in Step 2
(2- (Pyrrolidin-1- yl)phenyl) boronic acid (Sigma- Aldrich Corporation)









Synthesis of Intermediate A
N-(1-Acryloylazetidin-3-yl)-2-bromoacetamide



embedded image


Step 1: tert-Butyl (1-acryloylazetidin-3-yl)carbamate

To a stirred solution of tert-butyl azetidin-3-ylcarbamate hydrochloride (Suzhou Sibian Chemical Technology Co., Ltd., Suzhou, China, 135 g, 647 mmol) in DCM (1.35 L) was added TEA (225 mL, 1.62 mol) at −10° C. under a nitrogen atmosphere. The reaction mixture was stirred for 10 min. Acryloyl chloride (63.1 mL, 776 mmol) was added to the above reaction mixture slowly and stirred for 1 h at −10° C. The reaction mixture was quenched with water (1.0 L) and extracted with DCM. The organic extract was washed with brine, dried over Na2SO4, filtered and concentrated in vacuo. The crude residue was purified by silica gel chromatography (eluent: 0-70% EtOAc/hexanes) affording tert-butyl (1-acryloylazetidin-3-yl)carbamate (75 g, 51% yield) as an off-white solid. 1H NMR (400 MHz, CDCl3) δ 6.35 (dd, J=17.0, 1.8 Hz, 1H), 6.17 (dd, J=17.0, 10.4 Hz, 1H), 5.69 (dd, J=10.4, 1.8 Hz, 1H), 5.03 (b s, 1H), 4.55-4.38 (m, 3H), 4.03 (d, J=3.6 Hz, 1H), 3.88 (dd, J=10.7, 4.1 Hz, 1H), 1.47 (s, 9H). m/z (ESI, +ve ion): 227.2 (M+H)+.


Step 2: 1-(3-Aminoazetidin-1-yl)prop-2-en-1-one 2,2,2-trifluoroacetate

To a solution of tert-butyl (1-acryloylazetidin-3-yl)carbamate (44 g, 194 mmol, 1.0 equiv) in DCM (660 mL) was added TFA (74.9 mL, 972 mmol) at rt and stirred for 24 h. The reaction mixture was concentrated under reduced pressure to give 1-(3-aminoazetidin-1-yl)prop-2-en-1-one 2,2,2-trifluoroacetate as pale yellow oil. The crude material was taken for next step without further purification. 1H NMR (400 MHz, DMSO-d6) δ 8.34 (s, 3H), 6.34 (dd, J=16.9, 10.3 Hz, 1H), 6.13 (dd, J=17.0, 2.2 Hz, 1H), 5.71 (dd, J=10.3, 2.2 Hz, 1H), 4.48 (dd, J=9.9, 7.6 Hz, 1H), 4.20-4.11 (m, 3H), 3.89 (dd, J=11.0, 4.7 Hz, 1H). m/z (ESI, +ve ion): 127.2 (M+H)+.


Step 3: N-(1-Acryloylazetidin-3-yl)-2-bromoacetamide

To a solution of 1-(3-aminoazetidin-1-yl)prop-2-en-1-one 2,2,2-trifluoroacetate (60 g, 250 mmol) in DCM (1.5 L) was added DIPEA (109 mL, 625 mmol) slowly at 0° C. The reaction mixture was cooled to −70° C. and added 2-bromoacetyl bromide (50.4 g, 250 mmol) dropwise. The reaction mixture was stirred at the same temperature for 45 min. The reaction mixture was quenched with water and concentrated in vacuo. The crude residue was purified by reversed-phase purification using 10-13% of acetonitrile in water affording N-(1-acryloylazetidin-3-yl)-2-bromoacetamide (9.0 g, 15% yield) as a pale brown solid. 1H NMR (400 MHz, DMSO-d6) δ 9.00 (d, J=5.7 Hz, 1H), 6.39-6.25 (m, 1H), 6.11 (dq, J=17.0, 2.0 Hz, 1H), 5.68 (dq, J=10.3, 2.1 Hz, 1H), 4.55-4.41 (m, 2H), 4.18 (ddd, J=10.3, 6.6, 2.7 Hz, 1H), 4.07-3.97 (m, 1H), 3.86 (s, 2H), 3.77 (dd, J=10.2, 3.7 Hz, 1H). m/z (ESI, +ve ion): 247.1 (M+H)+.









TABLE 6







Separated Compound Examples













Separation





conditions for racemic


Ex. #
Chemical structure
Name
compounds





2-2-1


embedded image


2-(5-Bromo-2,7- dimethyl-3-(((1R)-1- methyl-3,4-dihydro- 2(1H)- isoquinolinyl)carbonyl)- 1H-indol-1-yl)-N- (1-(2-propenoyl)-3- azetidinyl)acetamide
Preparative Thar 200 SFC method: Column: OD (250 × 21 mm, 5 □) Mobile Phase: 60:40 (A:B) A: Liquid CO2 B: methanol (NH3) Flow Rate: 50 g/min Column/Oven temp.: ambient temperature 230 nm BPR = 102 bar SN: AF201





2-2-2


embedded image


2-(5-Bromo-2,7- dimethyl-3-(((1S)-1- methyl-3,4-dihydro- 2(1H)- isoquinolinyl)carbonyl)- 1H-indol-1-yl)-N- (1-(2-propenoyl)-3- azetidinyl)acetamide
Preparative Thar 200 SFC method: Column: OD (250 × 21 mm, 5 □□) Mobile Phase: 60:40 (A:B) A: Liquid CO2 B: methanol (NH3) Flow Rate: 50 g/min Column/Oven temp.: ambient temperature 230 nm BPR = 102 bar SN: AF201
















TABLE 7







Analytical Data










LCMS: (ESI,



Ex. #
+ve ion) m/z
NMR





1-1
550.3

1H NMR (400 MHz, DMSO-d6) δ ppm





8.98 (1 H, br d, J = 6.4 Hz), 7.93 (1 H, br d,




J = 4.8 Hz), 7.16 (1 H, br s), 6.92 (1 H, s),




6.76 (1 H, br s), 6.32 (1 H, dd, J = 17.1, 10.3




Hz), 6.10 (1 H, br d, J = 17.0 Hz), 5.67 (1 H,




br d, J = 10.4 Hz), 5.01 (2 H, s), 4.66 (2 H,




br s), 4.45-4.58 (2 H, m), 4.14-4.23 (1 H,




m), 4.02 (1 H, br d, J = 3.3 Hz), 3.87 (3 H,




s), 3.74-3.84 (2 H, m), 3.69 (1 H, br s),




2.60-2.76 (4 H, m), 2.58 (3 H, s), 1.04 (3




H, br t, J = 7.4 Hz)


1-2
549.2

1H NMR (400 MHz, DMSO-d6) δ ppm





8.98 (1 H, br d, J = 6.2 Hz), 7.09-7.22 (2




H, m), 6.92 (1 H, s), 6.82 (1 H, d, J = 8.3




Hz), 6.68 (1 H, br s), 6.32 (1 H, dd, J = 16.9,




10.3 Hz), 6.10 (1 H, dd, J = 16.9, 2.2 Hz),




5.67 (1 H, dd, J = 10.3, 2.2 Hz), 5.01 (2 H,




s), 4.66 (2 H, br s), 4.36-4.62 (2 H, m),




4.11-4.23 (1 H, m), 4.02 (1 H, br dd,




J = 8.0, 4.0 Hz), 3.72-3.85 (4 H, m), 3.67




(1 H, br s), 2.64-2.73 (3 H, m), 2.54-2.64




(2 H, m), 2.58 (3 H, s), 1.04 (3 H, br t,




J = 7.5 Hz)


1-3
587.9

1H NMR (400 MHz, DMSO-d6) δ ppm





8.98 (br d, J = 6.0 Hz, 1 H), 8.48 (s, 1




H), 7.86 (br d, J = 9.1 Hz, 1 H), 7.05-




7.34 (m, 1 H), 6.88-7.00 (m, 2 H),




6.32 (dd, J = 17.0, 10.2 Hz, 1 H), 6.10




(dd, J = 16.9, 2.0 Hz, 1 H), 5.67 (dd,




J = 10.3, 2.0 Hz, 1 H), 4.94-5.08 (m, 2




H), 4.13-4.64 (m, 6 H), 3.98-4.08 (m,




1 H), 3.80 (br dd, J = 10.2, 4.1 Hz, 2H),




3.34-3.48 (m, 1 H), 3.09-3.26 (m, 1




H), 2.63-3.02 (m, 2 H), 2.57 (s, 3 H),




1.02-1.29 (m, 7 H)


1-4
562.2

1H NMR (400 MHz, DMSO-d6) δ ppm





8.91-9.04 (m, 1 H), 7.85-8.04 (m, 1 H),




7.15-7.23 (m, 1 H), 6.90-6.99 (m, 1 H),




6.57-6.66 (m, 1 H), 6.28-6.38 (m, 1 H),




6.07-6.15 (m, 1 H), 5.65-5.72 (m, 1 H),




5.12-5.22 (m, 2 H), 4.80-4.90 (m, 1 H),




4.46-4.64 (m, 2 H), 4.15-4.25 (m, 1 H),




4.00-4.07 (m, 2 H), 3.85-3.92 (m, 3 H),




3.76-3.85 (m, 1 H), 3.55-3.65 (m, 1 H),




2.69-2.77 (m, 1 H), 2.55-2.63 (m, 3 H),




1.98-2.02 (m, 1 H), 1.71-1.92 (m, 1 H),




0.84-0.98 (m, 2 H), 0.59-0.77 (m, 1 H),




0.44-0.57 (m, 1 H), 0.32-0.40 (m, 1 H)


1-5
575.2

1H NMR (400 MHz, DMSO-d6) δ ppm





8.89 (br d, J = 6.6 Hz, 1 H), 7.04-7.14 (m,




2 H), 7.01 (br s, 1 H), 6.85 (s, 1 H), 6.73




(d, J = 8.1 Hz, 1 H), 6.38-6.68 (m, 1 H),




6.24 (dd, J = 17.0, 10.2 Hz, 1 H), 6.02 (dd,




J = 16.9, 2.0 Hz, 1 H), 5.59 (dd, J = 10.4, 1.9




Hz, 1 H), 4.87-5.06 (m, 2 H), 4.51-4.65




(m, 1 H), 4.37-4.51 (m, 2 H), 4.05-4.16




(m, 1 H), 3.90-3.98 (m, 1 H), 3.65-3.77




(m, 4 H), 3.49-3.62 (m, 1 H), 3.23 (s, 4




H), 2.57-2.68 (m, 3 H), 2.50 (s, 3 H), 0.69-




0.85 (m, 1 H), 0.06-0.28 (m, 1 H), -0.19-




0.06 (m, 1 H)


1-6
525.2

1H NMR (400 MHz, METHANOL-d4)





δ ppm 7.17-7.29 (m, 1 H), 6.91 (s, 1




H), 6.20-6.33 (m, 2 H), 5.73 (dd,




J = 8.9, 3.3 Hz, 1 H), 5.18-5.32 (m, 2




H), 4.63-4.72 (m, 1 H), 4.54-4.62 (m,




1 H), 4.31-4.40 (m, 1 H), 3.87-4.20




(m, 3 H), 3.59-3.79 (m, 2 H), 3.40-




3.57 (m, 3 H), 2.56-2.64 (m, 3 H),




1.58-2.21 (m, 7 H), 0.96-1.11 (m, 2




H), 0.74-0.89 (m, 1 H), 0.55-0.66 (m,




1H)


1-7
561.2

1H NMR (400 MHz, DMSO-d6) δ ppm





8.90-9.03 (m, 1 H), 7.19-7.26 (m, 1 H),




7.16 (s, 1 H), 7.05-7.13 (m, 1 H), 6.93 (br




s, 1 H), 6.78-6.91 (m, 1 H), 6.49-6.57




(m, 1 H), 6.28-6.38 (m, 1 H), 6.07-6.15




(m, 1 H), 5.65-5.72 (m, 1 H), 5.12-5.21




(m, 2 H), 4.80-4.87 (m, 1 H), 4.46-4.63




(m, 3 H), 4.16-4.24 (m, 1 H), 4.01-4.07




(m, 1 H), 3.87-3.98 (m, 1 H), 3.75-3.85




(m, 3 H), 3.52-3.62 (m, 1 H), 2.71-2.81




(m, 1 H), 2.55 -2.62 (m, 3 H), 1.71-1.91




(m, 1 H), 0.81-0.96 (m, 2 H), 0.60-0.73




(m, 1 H), 0.44-0.57 (m, 1 H), 0.32-0.40




(m, 1 H)


2-1
579.0/581.2

1H NMR (DMSO-d6) δ 8.98 (br d, J = 6.7





Hz, 1H), 7.32 (s, 1H), 7.15 (br t, J = 7.4 Hz,




1H), 7.03 (s, 1H), 6.82 (d, J = 8.0 Hz, 1H),




6.58-6.78 (m, 1H), 6.32 (dd, J = 16.9, 10.3




Hz, 1H), 6.10 (dd, J = 16.8, 2.0 Hz, 1H),




5.67 (dd, J = 10.5, 1.9 Hz, 1H), 5.00 (s, 2H),




4.43-4.72 (m, 4H), 4.18 (br t, J = 8.9 Hz,




1H), 3.98-4.07 (m, 1H), 3.66-3.87 (m, 6H),




2.67 (br s, 2H), 2.57 (s, 3H), 2.29 (s, 3H)


2-2
563.2/565.1

1H NMR (DMSO-d6) δ 8.90-9.08 (m, 1H),





7.11-7.42 (m, 5H), 7.02 (br d, J = 17.6 Hz,




1H), 6.24-6.40 (m, 1H), 6.05-6.16 (m, 1H),




5.67 (br d, J = 11.0 Hz, 1H), 4.92-5.08 (m,




2H), 4.51 (br t, J = 9.6 Hz, 2H), 4.12-4.28




(m, 1H), 4.04 (br s, 1H), 3.73-3.87 (m,




1H), 2.68-2.83 (m, 1H), 2.58 (d, J = 2.5 Hz,




3H), 2.48-2.49 (m, 3H), 2.33 (s, 2H), 1.46




(br d, J = 2.5 Hz, 2H), 1.15-1.33 (m, 3H)


2-2-1
563.2/565.1

1H NMR (DMSO-d6) δ 8.90-9.09 (m, 1H),





7.17 (br d, J = 3.1 Hz, 5H), 7.02 (br d,




J = 17.4 Hz, 1H), 6.32 (br dd, J = 16.6, 10.4




Hz, 1H), 6.03-6.16 (m, 1H), 5.67 (br d,




J = 10.6 Hz, 1H), 5.00 (br d, J = 8.3 Hz, 2H),




4.43-4.60 (m, 2H), 4.13-4.27 (m, 1H), 4.03




(br d, J = 3.3 Hz, 1H), 3.73-3.88 (m, 1H),




3.33-3.51 (m, 1H), 2.63-2.94 (m, 2H), 2.57




(br s, 3H), 2.33 (br s, 3H), 1.40-1.55 (m,




2H), 1.25 (br s, 3H)


2-2-2
563.0/565.1

1H NMR (DMSO-d6) δ 9.00 (br t, J = 7.2





Hz, 1H), 7.10-7.41 (m, 5H), 7.02 (br d,




J = 17.8 Hz, 1H), 6.24-6.41 (m, 1H), 6.04-




6.15 (m, 1H), 5.67 (br d, J = 11.0 Hz, 1H),




4.92-5.06 (m, 2H), 4.44-4.62 (m, 2H),




4.13-4.25 (m, 1H), 4.03 (br d, J = 4.6 Hz,




1H), 3.70-3.86 (m, 1H), 3.33-3.50 (m, 1H),




2.65-2.89 (m, 2H), 2.58 (d, J = 2.7 Hz, 3H),




2.33 (s, 3H), 1.34-1.59 (m, 3H), 1.17-1.33




(m, 2H)


2-3
606.8/608.8

1H NMR (DMSO-d6) δ 9.00 (br d, J = 6.8





Hz, 1H), 7.07-7.37 (m, 2H), 7.02 (br d,




J = 11.6 Hz, 1H), 6.68-6.86 (m, 2H), 6.24-




6.42 (m, 1H), 6.01-6.18 (m, 1H), 5.63-5.72




(m, 1H), 4.96-5.05 (m, 2H), 4.45-4.62 (m,




2H), 4.15-4.25 (m, 1H), 4.04 (br s, 1H),




3.76-3.86 (m, 1H), 3.74 (d, J = 2.5 Hz, 3H),




2.63-2.74 (m, 1H), 2.59 (br s, 3H), 2.52-




2.55 (m, 3H), 2.26-2.37 (m, 2H), 1.02-1.34




(m, 5H)


3-1
535.3

1H NMR (400 MHz, DMSO-d6) δ ppm





8.99 (br d, J = 6.7 Hz, 1 H), 7.19 (s, 1 H),




7.15 (br t, J = 7.9 Hz, 1 H), 6.90-6.94 (m, 1




H), 6.83 (d, J = 8.2 Hz, 1 H), 6.60-6.79 (m,




1 H), 6.32 (dd, J = 17.0, 10.4 Hz, 1 H), 6.11




(dd, J = 16.9, 2.2 Hz, 1 H), 5.68 (dd, J = 10.4,




2.2 Hz, 1 H), 5.01 (s, 2 H), 4.62-4.74 (m,




2 H), 4.46-4.60 (m, 2 H), 4.15-4.24 (m,




1 H), 4.00-4.07 (m, 1 H), 3.66-3.86 (m,




6 H), 2.63-2.73 (m, 2 H), 2.58 (s, 3 H),




2.22-2.32 (m, 3 H)


3-2
591.2/593.3

1H NMR (400 MHz, DMSO-d6) δ ppm





8.90-9.04 (m, 1 H), 7.46-7.51 (m, 1 H),




7.37-7.43 (m, 1 H), 7.26-7.31 (m, 1 H),




7.03-7.26 (m, 2 H), 6.79-6.86 (m, 1 H),




6.50-6.58 (m, 1 H), 6.28-6.37 (m, 1 H),




6.07-6.15 (m, 1 H), 5.68 (br d, J = 9.8 Hz,




1 H), 4.92-5.05 (m, 2 H), 4.53-4.64 (m,




2 H), 4.14-4.23 (m, 1 H), 4.02-4.07 (m,




1 H), 3.75-3.81 (m, 3 H), 3.53-3.68 (m,




2 H), 2.71-2.83 (m, 1 H), 1.78-1.95 (m,




2 H), 0.82-0.94 (m, 2 H), 0.48-0.76 (m,




3 H), 0.39-0.45 (m, 1 H)


4-1
564.2

1H NMR (400 MHz, DMSO-d6) δ ppm





7.93 (1 H, br d, J = 5.2 Hz), 7.15 (1 H, s),




6.90 (1 H, s), 6.77 (1 H, br s), 6.25-6.47




(1 H, m), 6.03-6.22 (1 H, m), 5.62-5.76




(2 H, m), 5.35 (1 H, br s), 5.28 (1 H, br s),




4.95-5.21 (2 H, m), 4.66 (2 H, br s), 4.50-




4.62 (1 H, m), 4.36-4.50 (1 H, m), 4.30 (1




H, br d, J = 9.1 Hz), 3.94-4.20 (3 H, m),




3.87 (3 H, s), 3.78 (1 H, br s), 3.53-3.74




(2 H, m), 2.68-2.76 (1 H, m), 2.61-2.66




(1 H, m), 2.34-2.46 (2 H, m), 2.22-2.32




(1 H, m), 1.01 (3 H, br t, J = 7.4 Hz) two




rotomers observed


4-2
563.2

1H NMR (400 MHz, DMSO-d6) δ ppm





7.11-7.19 (m, 2 H), 6.86-6.93 (m, 1 H),




6.83 (d, J = 8.1 Hz, 1 H), 6.64-6.78 (m, 1




H), 6.28-6.43 (m, 1 H), 6.13 (td, J = 16.4,




1.9 Hz, 1 H), 5.65-5.74 (m, 1 H), 5.25-




5.39 (m, 2 H), 5.03-5.19 (m, 2 H), 4.53-




4.72 (m, 2 H), 4.39-4.49 (m, 1 H), 4.22-




4.36 (m, 1 H), 4.00-4.21 (m, 2 H), 3.75-




3.83 (m, 3 H), 3.57-3.75 (m, 2 H), 3.21 (s,




2 H), 2.95-3.01 (m, 1 H), 2.60-2.76 (m,




4 H), 2.40-2.46 (m, 2 H), 0.95-1.07 (m,




3 H)


4-3
575.2

1H NMR (400 MHz, DMSO-d6) δ ppm





7.04-7.26 (m, 2 H), 6.90 (br s, 1 H), 6.79-




6.87 (m, 1 H), 6.46-6.57 (m, 1 H), 6.28-




6.44 (m, 1 H), 6.07-6.19 (m, 1 H), 5.64-




5.75 (m, 1 H), 5.38-5.54 (m, 2 H), 5.04-




5.25 (m, 1 H), 4.76-4.91 (m, 1 H), 4.39-




4.62 (m, 3 H), 4.27-4.36 (m, 1 H), 4.09-




4.23 (m, 1 H), 3.97-4.09 (m, 1 H), 3.71-




3.89 (m, 4 H), 3.50-3.62 (m, 1 H), 3.19-




3.26 (m, 2 H), 2.96-3.04 (m, 1 H), 2.72-




2.82 (m, 1 H), 2.43-2.48 (m, 1 H), 1.68-




1.90 (m, 2 H), 0.76-0.95 (m, 2 H), 0.55-




0.71 (m, 1 H), 0.38-0.53 (m, 1 H), 0.23-




0.34 (m, 1 H)


4-4
589.3

1H NMR (400 MHz, DMSO-d6) δ ppm





6.99-7.11 (m, 2 H), 6.81 (s, 1 H), 6.72 (d,




J = 8.3 Hz, 1 H), 6.46-6.67 (m, 1 H), 6.18-




6.34 (m, 1 H), 6.03 (td, J = 16.6, 2.0 Hz, 1




H), 5.51-5.69 (m, 1 H), 5.19-5.35 (m, 2




H), 4.93-5.10 (m, 1 H), 4.43-4.67 (m, 2




H), 4.28-4.40 (m, 1 H), 4.16-4.24 (m, 1




H), 3.90-4.08 (m, 4 H), 3.62-3.73 (m, 3




H), 3.50-3.62 (m, 1 H), 3.11 (s, 2 H), 3.08




(d, J = 5.2 Hz, 2 H), 2.84-2.91 (m, 1 H),




2.48-2.67 (m, 3 H), 2.31-2.38 (m, 1 H),




0.65-0.80 (m, 1 H), 0.05-0.25 (m, 2 H), -




0.21-0.05 (m, 2 H)


4-5
593.2/595.1

1H NMR (DMSO-d6) δ 7.31 (br s, 1H),





7.11-7.19 (m, 1H), 7.00 (s, 1H), 6.82 (br d,




J = 8.0 Hz, 1H), 6.58-6.75 (m, 1H), 6.25-




6.44 (m, 1H), 6.12 (br t, J = 16.7 Hz, 1H),




5.61-5.78 (m, 1H), 5.24-5.39 (m, 2H),




5.00-5.19 (m, 1H), 4.40-4.75 (m, 3H),




4.24-4.35 (m, 1H), 4.08-4.20 (m, 1H),




3.98-4.07 (m, 1H), 3.64-3.84 (m, 5H), 3.19




(s, 3H), 2.67 (br s, 2H), 2.46 (br s, 3H),




2.23-2.27 (m, 3H)


4-6
602.0

1H NMR (400 MHz, DMSO-d6) δ ppm





8.49 (d, J = 1.7 Hz, 1 H), 7.86 (dd,




J = 9.0, 2.2 Hz, 1 H), 7.04-7.32 (m, 1




H), 6.86-6.99 (m, 2 H), 6.26-6.43 (m,




1 H), 6.12 (td, J = 16.7, 1.9 Hz, 1 H),




5.64-5.74 (m, 1 H), 5.25-5.40 (m, 2




H), 5.00-5.19 (m, 1 H), 3.65-4.65 (m,




9 H), 3.31-3.49 (m, 2 H), 2.62-3.27




(m, 7 H), 1.04-1.23 (m, 7 H)


5-1
451.2

1H NMR (400 MHz, CHLOROFORM-





d) δ ppm 8.52-8.65 (m, 1 H), 8.36 (br




s, 1 H), 7.37 (br d, J = 3.1 Hz, 1 H), 7.17-




7.23 (m, 3 H), 6.24 (br dd, J = 16.7, 8.8




Hz, 1 H), 5.95-6.13 (m, 2 H), 5.66 (br




d, J = 10.6 Hz, 1 H), 4.83 (s, 2 H), 4.68-




4.80 (m, 1 H), 4.49 (br t, J = 8.1 Hz, 1




H), 4.35 (br t, J = 8.3 Hz, 1 H), 3.80-




3.91 (m, 1 H), 3.65-3.78 (m, 1 H),




2.84 (dt, J = 13.7, 7.0 Hz, 1 H), 2.25 (s,




3 H), 1.16 (d, J = 6.8 Hz, 3 H), 1.08 (d,




J = 7.0 Hz, 3 H)


5-2
452.2

1H NMR (400 MHz, CHLOROFORM-





d) δ ppm 9.22 (s, 1 H), 8.50 (s, 1 H),




7.18-7.25 (m, 3 H), 6.01-6.27 (m, 3




H), 5.66 (br d, J = 9.7 Hz, 1 H), 4.83 (s,




2 H), 4.71-4.81 (m, 1 H), 4.50 (br d,




J = 2.1 Hz, 1 H), 4.29-4.40 (m, 1 H),




3.69-3.91 (m, 2 H), 2.98 (dt, J = 13.4,




6.6 Hz, 1 H), 2.27 (s, 3 H), 1.21 (d,




J = 6.8 Hz, 3 H), 1.16 (d, J = 6.6 Hz, 3 H)


5-3
450.2

1H NMR (400 MHz, CHLOROFORM-





d) δ ppm 7.38-7.51 (m, 3 H), 7.13-




7.26 (m, 4 H), 6.24-6.33 (m, 1 H),




6.03-6.13 (m, 1 H), 5.77 (br s, 1 H),




5.67 (dd, J = 10.4, 1.2 Hz, 1 H), 4.81 (s,




2 H), 4.72 (br d, J = 5.1 Hz, 1 H), 4.48




(br t, J = 8.4 Hz, 1 H), 4.34 (br t, J = 9.4




Hz, 1 H), 3.82 (br d, J = 5.3 Hz, 1 H),




3.68 (br d, J = 1.6 Hz, 1 H), 2.82 (dt,




J = 13.7, 6.9 Hz, 1 H), 2.23 (s, 3 H), 1.11




(dd, J = 13.3, 6.8 Hz, 6 H)


5-4
477.2

1H NMR (400 MHz, CHLOROFORM-





d) δ ppm 7.48 (s, 1 H), 7.28-7.32 (m,




1 H), 7.12-7.22 (m, 3 H), 6.96 (d,




J = 8.2 Hz, 1 H), 6.89 (t, J = 7.3 Hz, 1 H),




6.24-6.33 (m, 1 H), 6.03-6.13 (m, 1




H), 5.74 (br s, 1 H), 5.67 (dd, J = 10.4,




1.6 Hz, 1 H), 4.79 (s, 2 H), 4.66-4.76




(m, 1 H), 4.47 (br t, J = 7.9 Hz, 1 H),




4.33 (br t, J = 9.4 Hz, 1 H), 3.76-3.86




(m, 1 H), 3.57-3.68 (m, 1 H), 2.77-




2.93 (m, 4 H), 2.24 (s, 3 H), 1.62-1.78




(m, 4 H)









The following compounds were prepared by methods similar to those detailed in methods 1-5:









TABLE 8







Mass Data











LRMS: m/z


Ex.

(ESI, +ve ion):


#
Chemical structure
(M + H)+





 6


embedded image


515.2





 7


embedded image


556.1/558.2





 8


embedded image


617.9/619.8





 9


embedded image


594.8/596.8





 10


embedded image


647.0/649.1





 11


embedded image


511.2





 12


embedded image


566.0/568.1





 13


embedded image


565.0/567.2





 14


embedded image


586.2





 15


embedded image


542.2/544.1





 16


embedded image


603.8/605.7





 17


embedded image


532.1





 18


embedded image


449.0





 19


embedded image


600.2





 20


embedded image


497.2





 21


embedded image


659.0





 22


embedded image


617.9/619.8





 23


embedded image


513.2





 24


embedded image


549.2





 25


embedded image


549.2





 26


embedded image


530.8/532.8





 27


embedded image


604.1/606.0





 28


embedded image


522.0





 29


embedded image


579.0/581.0





 30


embedded image


450.2





 31


embedded image


575.2





 32


embedded image


465.2





 33


embedded image


535.1





 34


embedded image


644.9





 35


embedded image


577.1/579.2





 36


embedded image


448.2





 37


embedded image


543.2





 38


embedded image


624.0/626.2





 39


embedded image


563.2/565.1





 40


embedded image


633.0/635.0





 41


embedded image


521.2





 42


embedded image


591.1/593.2





 43


embedded image


501.8/503.9





 44


embedded image


579.0/581  





 45


embedded image


565.1/567.0





 46


embedded image


610.1/612.0





 47


embedded image


593.2/595.1





 48


embedded image


565.1/567.0





 49


embedded image


493.3





 50


embedded image


591.0/593.2





 51


embedded image


627.0





 52


embedded image


617.9/619.8





 53


embedded image


563.3





 54


embedded image


436.2





 55


embedded image


506.0/508.1





 56


embedded image


498.2





 57


embedded image


506.0/508.1





 58


embedded image


550.8/553.0





 59


embedded image


577.1/579.2





 60


embedded image


551.0/553.0





 61


embedded image


565.1/567.0





 62


embedded image


528.2/530.2





 63


embedded image


535.5





 64


embedded image


517.0





 65


embedded image


534.0





 66


embedded image


549.7





 67


embedded image


484.2





 68


embedded image


553.2





 69


embedded image


541.2





 70


embedded image


489.9/490.0





 71


embedded image


412.0/414.0





 72


embedded image


590.8/592.8





 73


embedded image


576.8/578.9





 74


embedded image


525.2





 75


embedded image


446.9/449.0





 76


embedded image


551.2





 77


embedded image


579.0





 78


embedded image


408.2





 79


embedded image


565.1/567.0





 80


embedded image


627.0





 81


embedded image


604.0/605.9





 82


embedded image


565.1





 83


embedded image


590.0/592.1





 84


embedded image


368.0





 85


embedded image


487.9/489.8





 86


embedded image


535.2





 87


embedded image


515.9/517.8





 88


embedded image


552.2





 89


embedded image


 552.0/5540.0





 90


embedded image


470.0





 91


embedded image


410.0/411.8





 92


embedded image


603.1





 93


embedded image


581.1/583.1





 94


embedded image


549.2





 95


embedded image


501.8/503.9





 96


embedded image


591.0





 97


embedded image


555.0/557.0





 98


embedded image


451.2





 99


embedded image


596.0/598.1





100


embedded image


417.1 (as des- Boc)





101


embedded image


473.2





102


embedded image


430.1/432.0





103


embedded image


551.0/553.1





104


embedded image


601.0





105


embedded image


496.9/498.8





106


embedded image


599.0





107


embedded image


565.1/567.0





108


embedded image


420.0





109


embedded image


555.0





110


embedded image


502.0/504.0





111


embedded image


545.9





112


embedded image


470.2





113


embedded image


466.2





114


embedded image


539.2/541.2





115


embedded image


511.0/512.9





116


embedded image


536.0





117


embedded image


412.1





118


embedded image


539.1/541.1





119


embedded image


494.0/496.0





120


embedded image


613.0





121


embedded image


422.2





122


embedded image


535.2





123


embedded image


516.0





124


embedded image


579.2





125


embedded image


438.2





126


embedded image


535.1





127


embedded image


572.2





128


embedded image


455.0





129


embedded image


521.1/523.1





130


embedded image


492.1/494.1





131


embedded image


549.2





132


embedded image


402.0/404.0





133


embedded image


492.2





134


embedded image


546.2/548.2





135


embedded image


397.3





136


embedded image


481.1





137


embedded image


474.0/476.0





138


embedded image


551.0/553.1





139


embedded image


443.9





140


embedded image


509.1





141


embedded image


551.2





142


embedded image


448.1





143


embedded image


383.1





144


embedded image


428.0





145


embedded image


507.0/509.0





146


embedded image


434.1





147


embedded image


459.0





148


embedded image


563.0/565.1





149


embedded image


608.2/610.1





150


embedded image


520.0





151


embedded image


468.0/470.1





152


embedded image


587.2





153


embedded image


502.0





154


embedded image


398.0





155


embedded image


509.1





156


embedded image


539.1





157


embedded image


631.0





158


embedded image


654.0





159


embedded image


608.2/610.0





160


embedded image


416.0/418.1





161


embedded image


577.0





162


embedded image


591.1





163


embedded image


495.0





164


embedded image


495.0





165


embedded image


352.1





166


embedded image


595.1





167


embedded image


580.1





168


embedded image


589.2





169


embedded image


580.1





170


embedded image


602.2





171


embedded image


570.1





172


embedded image


544.1





173


embedded image


552.2





174


embedded image


563.2





175


embedded image


524.0/526.0





176


embedded image


544.0





177


embedded image


594.2





178


embedded image


567.0





179


embedded image


594.2





180


embedded image


362.1





181


embedded image


420.0





182


embedded image


362.1





183


embedded image


284.3





184


embedded image


390.0





185


embedded image


390.0





186


embedded image


376.1





187


embedded image


318.2





188


embedded image


410.0





189


embedded image


509.1





190


embedded image


376.1





191


embedded image


309.0





192


embedded image


621.0





193


embedded image


491.9





194


embedded image


376.0/377.9





195


embedded image


334.0





196


embedded image


396.1





197


embedded image


398.1





198


embedded image


302.2





199


embedded image


318.1





200


embedded image


384.2





201


embedded image


356.1





202


embedded image


318.2





203


embedded image


318.2





204


embedded image


462.0





205


embedded image


544.2





206


embedded image


439.1/441.1





207


embedded image


438.2





208


embedded image


552.2





209


embedded image


496.3





210


embedded image


508.2





211


embedded image


386.9





212


embedded image


410.0





213


embedded image


374.1





214


embedded image


401.0/403.0





215


embedded image


361.3





216


embedded image


463.0/465.0





217


embedded image


324.2





218


embedded image


447.0/449.0





219


embedded image


461.1





220


embedded image


410.1





221


embedded image


484.1/486.1 (M + Na)+





222


embedded image


530.0/532.1





223


embedded image


590.2/592.0





224


embedded image


528.0/530.2





225


embedded image


545.0





226


embedded image


509.1





227


embedded image


551.2





228


embedded image


575.0/577.0





229


embedded image


460.1





230


embedded image


528.0/530.2





231


embedded image


452.1/454.1





232


embedded image


329.1





233


embedded image


407.0





234


embedded image


523.0/525.0





235


embedded image


352.1





236


embedded image


314.1





237


embedded image


360.0





238


embedded image


415.1





239


embedded image


507.2





240


embedded image


363.2





241


embedded image


523.1





242


embedded image


445.1








embedded image








243


embedded image


438.0





244


embedded image


547.3





245


embedded image


580.1





246


embedded image


502.3





247


embedded image


518.3





248


embedded image


417.2 (as dec- Boc)





249


embedded image


417.2





250


embedded image


417.1





251


embedded image


459.1





252


embedded image


459.2





253


embedded image


539.2





254


embedded image


403.1 (as des- Boc)





255


embedded image


380.1





256


embedded image


574.1/576.0





257


embedded image


478.0





258


embedded image


352.1





259


embedded image


413.0/415.0





260


embedded image


474.0/476.0





261


embedded image


425.9/428.0





262


embedded image


430.0





263


embedded image


550.2





264


embedded image


501.8/504.0





265


embedded image


528.0/530.0





266


embedded image


330.2





267


embedded image


380.0/382.0





268


embedded image


494.0/496.0





269


embedded image


499.0





270


embedded image


498.2





271


embedded image


526.2





272


embedded image


517.2





273


embedded image


499.0





274


embedded image


510.2





275


embedded image


499.2





276


embedded image


499.2





277


embedded image


312.2





278


embedded image


465.2 (M + Na+)





279


embedded image


551.0/553.1





280


embedded image


369.2





281


embedded image


444.0





282


embedded image


523.0





283


embedded image


567.0





284


embedded image


539.0





285


embedded image


539.0





286


embedded image


552.0





287


embedded image


563.0/565.0





288


embedded image


549.0/551.0





289


embedded image


551.2/553.2





290


embedded image


577.2/579.2





291


embedded image


537.2/539.2





292


embedded image


580.2/582.2





293


embedded image


516.2/518.0





294


embedded image


531.2/533.2





295


embedded image


510.2/512.2





296


embedded image


534.2/536.2





297


embedded image


499.0/501.2





298


embedded image


539.1/541.1





299


embedded image


523.0/525.0





300


embedded image


536.0/538.0





301


embedded image


536.0/538.0





302


embedded image


536.0/538.0





303


embedded image


503.0/505.0





304


embedded image


512.9/514.8





305


embedded image


487.0/489.0





306


embedded image


443.0/445.0





307


embedded image


489.0/491.0





308


embedded image


531.0/533.0





309


embedded image


503.0/505.0





310


embedded image


591.1/593.2





311


embedded image


549.2





312


embedded image


531.0/533.0





313


embedded image


609.0/611.0





314


embedded image


434.1





315


embedded image


576.0/578.0





316


embedded image


527.2





317


embedded image


352.0





318


embedded image


464.2





319


embedded image


398.2





320


embedded image


509.9





321


embedded image


601.8/604.0





322


embedded image


570.2





323


embedded image


573.0





324


embedded image


491.2





325


embedded image


465.2





326


embedded image


584.2





327


embedded image


553.2





328


embedded image


543.2





329


embedded image


586.2





330


embedded image


579.1





331


embedded image


578.2





332


embedded image


525.1





333


embedded image


499.1





334


embedded image


517.2





335


embedded image


503.2





336


embedded image


512.0





337


embedded image


517.2





338


embedded image


466.0





339


embedded image


480.0





340


embedded image


517.2





341


embedded image


505.2





342


embedded image


506.0





343


embedded image


482.0





344


embedded image


520.0





345


embedded image


495.0





346


embedded image


376.1/378.0





347


embedded image


376.0





348


embedded image


390.0/392.1





349


embedded image


376.1/378.0





350


embedded image


390.0/392.1





351


embedded image


390.0/392.0





352


embedded image


376.1





353


embedded image


392.0





354


embedded image


406.1





355


embedded image


390.0/392.0





356


embedded image


390.0





357


embedded image


390.0/392.1





358


embedded image


332.1





359


embedded image


386.1/388.1





360


embedded image


323.1





361


embedded image


376.0





362


embedded image


376.0





363


embedded image


376.0





364


embedded image


390.0









Biological Analysis

For compounds in Table 9, the following assay conditions were employed:


Coupled Nucleotide Exchange Assay:


Purified GDP-bound KRAS protein (aa 1-169), containing both G12C and C118A amino acid substitutions and an N-terminal His-tag, was pre-incubated in assay buffer (25 mM HEPES pH 7.4, 10 mM MgCl2, and 0.01% Triton X-100) with a compound dose-response titration for either 5 min, 2 hours or 20 hours (see Table 15). Following compound pre-incubation, purified SOS protein (aa 564-1049) and GTP (Roche 10106399001) were added to the assay wells and incubated for an additional 30 min (for 5 min compound pre-incubation) or 1 hour (for 2 hour compound pre-incubation). To determine the extent of inhibition of SOS-mediated nucleotide exchange, purified GST-tagged cRAF (aa 1-149), nickel chelate AlphaLISA acceptor beads (PerkinElmer AL108R), and AlphaScreen glutathione donor beads (PerkinElmer 6765302) were added to the assay wells and incubated for 10 minutes. The assay plates were then read on a PerkinElmer EnVision Multilabel Reader, using AlphaScreen® technology, and data were analyzed using a 4-parameter logistic model to calculate IC50 values.


Phospho-ERK1/2 MSD Assay:


MIA PaCa-2 (ATCC® CRL-1420™) cells were cultured in RPMI 1640 Medium (ThermoFisher Scientific 11875093) containing 10% fetal bovine serum (ThermoFisher Scientific 16000044) and 1× penicillin-streptomycin-glutamine (ThermoFisher Scientific 10378016). Sixteen hours prior to compound treatment, MIA PaCa-2 cells were seeded in 96-well cell culture plates at a density of 25,000 cells/well and incubated at 37° C., 5% CO2. A compound dose-response titration was diluted in growth media, added to appropriate wells of a cell culture plate, and then incubated at 37° C., 5% C02 for 2 or 4 hours. Following compound treatment, cells were stimulated with 10 ng/mL EGF (Roche 11376454001) for 10 min, washed with ice-cold Dulbecco's phosphate-buffered saline, no Ca2+ or Mg2+ (ThermoFisher Scientific 14190144), and then lysed in RIPA buffer (50 mM Tris-HCl pH 7.5, 1% Igepal, 0.5% sodium deoxycholate, 150 mM NaCl, and 0.5% sodium dodecyl sulfate) containing protease inhibitors (Roche 4693132001) and phosphatase inhibitors (Roche 4906837001). Phosphorylation of ERK1/2 in compound-treated lysates was assayed using Phospho-ERK1/2 Whole Cell Lysate kits (Meso Scale Discovery K151DWD) according to the manufacturer's protocol. Assay plates were read on a Meso Scale Discovery Sector Imager 6000, and data were analyzed using a 4-parameter logistic model to calculate IC50 values.









TABLE 9







Biochemical and cellular activity of compounds













Coupled
Coupled
Coupled
pERK
pERK



Exchange
Exchange
Exchange
IC50
IC50



IC50
IC50
IC50
(4 h,
(4 h,



(5 min;
(2 h;
(20 h;
MiaPaCa-
A549;


Ex.#
□M)
□M)
□M)
2; □M)
□M)















 1-1
0.20
0.07

0.09
>100.0


 1-2

0.06

0.13
>100.0


 1-3

0.15

1.31
>100.0


 1-4
0.27
0.08
0.01
0.15
>100.0


 1-5

0.08

0.15
>100.0


 1-6

0.72

0.67
>100.0


 1-7

0.11
0.11
0.22
60.40


 2-1

0.11
0.11
0.30
>100.0


 2-2

0.25

2.28
40.90


 2-2-1

0.16

0.56
47.00


 2-2-2

0.42

3.01
61.00


 2-3

0.16

0.67
51.70


 3-1

0.15
0.10
0.68
>100.0


 3-2

0.14

0.60
52.60


 4-1

0.12

0.13
>100.0


 4-2

0.15

0.21
>100.0


 4-3

0.12

0.15
74.10


 4-4

0.10

0.18
29.30


 4-5

0.09

0.27
>100.0


 4-6

0.13

0.73
86.20


 5-1

0.78

1.32
>100.0


 5-2

1.05

1.49
>100.0


 5-3
4.11
0.63
0.22
2.06
>33.3


 5-4

0.38

5.49
>100.0


 6

0.34

1.12
>100.0


 7

0.45

1.33



 8

0.22

1.56
>100.0


 9

0.20

1.56
41.60


 10

0.82

1.58
43.40


 11

0.66

1.60
36.40


 12

0.51
0.17
1.65
>100.0


 13

0.30
0.11
1.68
>100.0


 14

0.58

1.70
>100.0


 15

0.22

1.79
>100.0


 16

0.33

1.79
>100.0


 17

1.22
0.30
1.83
>100.0


 18

0.97

1.85
>100.0


 19

0.61

1.87
>100.0


 20

1.27

1.94
>100.0


 21

0.49
0.10
1.95
27.30


 22

0.19

2.02
>100.0


 23

0.95

2.35
>100.0


 24

2.44
0.62
2.44
>100.0


 25

0.54
0.09
2.47
>100.0


 26

1.59

2.48
>33.3


 27

0.77
0.24
2.50
>100.0


 28

0.59
0.18
2.63
>100.0


 29

2.47
0.53
2.66
>100.0


 30

2.32

2.68
>100.0


 31

1.18

2.70
>100.0


 32
202.00
6.05

2.73
>100.0


 33

0.21
0.13
2.80
>100.0


 34

1.18
0.17
3.06
>100.0


 35

0.29

3.06
55.70


 36

0.71
0.20
3.31
>33.3


 37

1.09
0.17
3.33
>100.0


 38

0.76
0.22
3.33
>100.0


 39

0.33

3.35
60.30


 40

2.08

3.42
44.60


 41

0.37
0.12
3.55
>100.0


 42

0.97
0.28
3.78
>33.3


 43
36.60
4.65
0.44
3.82
>33.3


 44

3.94
0.95
3.88
>100.0


 45

5.18
1.07
3.96
>100.0


 46

1.65
0.36
4.08
>100.0


 47

1.21
0.30
4.16
>100.0


 48

5.31
1.05
4.56
>33.3


 49

0.72

4.67
7.68


 50

0.66

4.81
>33.3


 51

0.75
0.18
4.82
>100.0


 52

0.53

4.83
>100.0


 53

0.85
0.18
5.00
>100.0


 54
4.37
0.65
0.20
5.88
>33.3


 55

0.95
0.23
5.93
>100.0


 56
4.77
0.88

6.17
>100.0


 57

0.99
0.14
6.81
>100.0


 58

0.28
0.11
6.81
>33.3


 59

0.81

6.84
45.90


 60

7.07
0.96
6.94
>100.0


 61

7.94
1.59
7.06
>33.3


 62

1.18
0.25
7.06
>33.3


 63
>250.0
2.79
0.45
7.57
>100.0


 64

2.57
0.54
7.74
>100.0


 65

2.91

7.99
3.47


 66
19.50
2.41
0.10
8.48
>33.3


 67
3.90
0.83
0.24
8.80
>100.0


 68

2.58
0.56
8.81
>100.0


 69

0.19
0.13
9.39
>100.0


 70

8.96
1.70
9.94
>100.0


 71

3.51
0.52
10.60
>100.0


 72

3.25

10.60
63.40


 73

2.06

10.60
28.30


 74

0.22
0.17
11.10
>100.0


 75

3.73
0.43
11.30
>100.0


 76

0.64
0.17
11.40
>100.0


 77

2.14
0.48
11.59
10.20


 78
33.10
5.49
1.20
11.70
>100.0


 79

8.97
1.38
11.80
>100.0


 80

0.33
0.15
12.20
25.70


 81

1.03

12.50
>100.0


 82

0.17
0.13
12.80
>100.0


 83

1.22
0.23
13.60
>100.0


 84

3.12
0.63
13.90
61.00


 85
23.60
3.84
0.49
13.99
>100.0


 86

0.68
0.17
14.12
>100.0


 87

4.81
0.91
14.70
>100.0


 88

3.26
0.72
14.90
>100.0


 89

1.32
0.28
14.90
>100.0


 90
61.10
4.30
0.48
16.00
>100.0


 91

3.18
0.37
16.30
>100.0


 92

0.39
0.18
16.50
>100.0


 93

4.29
0.57
16.50
>100.0


 94

0.21
0.13
16.60
>100.0


 95

5.79
1.09
16.70
>100.0


 96

1.68
0.31
17.40
30.50


 97

2.00
0.57
17.90



 98

1.12

18.20
13.20


 99

1.88
0.33
18.90
>100.0


100

5.46
0.64
19.38
>33.3


101

1.11

20.30
56.80


102

4.90
0.90
21.20
>100.0


103

3.36
0.70
21.60
>100.0


104

2.32
0.66
21.70
14.60


105

6.45

22.00
11.30


106

0.63
0.18
22.20
44.70


107

0.59
0.18
23.10
>100.0


108

22.00
3.55
23.50
>100.0


109

1.29
0.31
23.90
21.30


110

4.39
1.20
24.90
42.10


111

1.98
0.35
25.40
>100.0


112
3.95
2.36
0.71
25.90
>100.0


113

0.90

26.20
>100.0


114

1.37
0.31
28.00
>100.0


115

2.78

28.10
>100.0


116

3.35
0.66
28.50
>100.0


117

2.05
0.43
29.60
>100.0


118

2.12
0.51
30.00
>100.0


119

1.71
0.39
30.60
>100.0


120

1.02
0.28
30.80
28.40


121
12.40
1.57
0.42
31.20
>100.0


122

0.69
0.17
31.40
>100.0


123

7.91
1.70
32.40
>100.0


124

1.17
0.26
32.90
>100.0


125

2.75

33.60
>100.0


126
11.20
1.54
0.11
33.90
48.40


127

2.98
0.66
34.30
>100.0


128
>250.0
7.24
1.18
35.60
>100.0


129

1.27
0.19
36.50
>100.0


130

4.49
1.02
37.00
50.10


131

5.54
1.04
37.00
50.60


132

9.52
1.71
37.90
>100.0


133

7.06
1.33
37.90
>100.0


134

1.88
0.42
38.00
>100.0


135

4.25
0.63
38.70
>100.0


136

9.85
1.91
39.60
36.60


137

6.90
1.13
40.40
16.50


138

6.37
0.89
40.50
>100.0


139
>250.0
4.15
0.52
40.60
>100.0


140

4.26
0.68
41.20
>100.0


141

7.25
1.51
42.40
19.30


142

3.71
0.83
44.10
>100.0


143

4.53
0.85
44.90
>100.0


144

3.84
0.75
46.80
>100.0


145

5.70
1.12
46.90
50.90


146

3.80
0.74
47.10
74.20


147

2.60
0.52
49.60
>100.0


148

3.02
0.47
50.20
>100.0


149

3.60
0.68
50.70
>100.0


150

2.92
0.59
50.80
>100.0


151

4.33
0.92
51.20
14.60


152

1.80
0.47
51.60
68.80


153

1.58
0.26
51.80
>100.0


154
56.20
5.12
0.86
52.10
>100.0


155

6.63
0.78
52.70
>100.0


156

2.55
0.44
53.40
>100.0


157

3.04

53.60
42.50


158

0.44
0.16
53.90
>100.0


159

2.29
0.53
57.40
>100.0


160

4.95
1.07
58.30
>100.0


161

0.99
0.23
79.50
>100.0


162

0.76
0.26
81.50
>100.0


163

14.20
1.91
>100.0
>100.0


164

10.70
1.11
>100.0
>100.0


165

13.20
2.65
>100.0
>100.0


166

1.21
0.28
>100.0
>100.0


167

1.08
0.26
>100.0
>100.0


168

2.28
0.51
>100.0
>100.0


169

2.96
0.68
>100.0
>100.0


170

3.00
0.65
>100.0
>100.0


171

4.64
1.05
>100.0
>100.0


172

4.27
0.93
>100.0
>100.0


173

2.94
0.67
>100.0
>100.0


174

7.69
1.65
>100.0
>100.0


175

3.55
0.69
>100.0
>100.0


176

2.80
0.60
>100.0
>100.0


177

0.43
0.17
>100.0
>100.0


178

0.63
0.22
>100.0
>100.0


179

8.83
1.11
>100.0
>100.0


180
201.00
>250.0
5.50




181

>250.0
6.47




182

>250.0
>250.0




183

>250.0
>250.0




184

32.50
4.41




185

>250.0
15.90




186
>250.0
26.60
4.47




187
>250.0
>250.0
7.27




188

22.20
3.71




189

15.60
1.62




190
193.00
16.00
1.76




191

>250.0
>250.0




192

107.00
61.70




193

>250.0
0.39




194
151.00
30.10
2.23




195

>250.0
33.50




196

45.60
6.09




197

24.50
5.51




198

>250.0
>250.0




199

>250.0
21.60




200

>250.0
>250.0




201

>250.0
>250.0




202

>250.0
>250.0




203

>250.0
>250.0




204

22.00
5.85




205

80.50
11.40




206

14.10
2.84




207

164.00
18.20




208

20.50
13.00




209

14.10
3.01




210

>250.0
10.90




211

22.90
5.20




212
>250.0
27.40
6.80




213

31.10
5.09




214

33.80
4.48




215

>250.0
>250.0




216

12.90
3.00




217

>250.0
>125.0




218

>250.0
>250.0




219

>250.0
2.87




220

12.20
1.89




221

26.30
6.52




222

36.50
6.41




223

16.10
2.64




224

16.60
2.83




225

>250.0
19.80




226

8.23
1.66




227

12.30
2.61




228

14.60
4.65




229

18.90
5.40




230

23.20
4.25




231

10.30
2.05




232

>250.0
94.10




233
>250.0
11.40
1.58




234

>250.0
0.32




235

>250.0
5.45




236

>250.0
>250.0




237

>250.0
>250.0




238

73.10
9.89




239

>250.0
44.00




240

13.10
1.33




241

>15.6
1.78




242

>250.0
7.66




243

18.20
2.82




244

5.16
0.75




245

2.19
0.27




246

3.09
0.48




247

3.23
0.41




248

43.90
3.22




249

54.60
21.70




250

89.40
5.69




251

>250.0
7.91




252

19.40
2.49




253

19.40
2.31




254

38.20
4.92




255

>250.0
4.89




256

23.80
3.01




257
64.70
9.12
0.97




258

104.00
5.22




259

26.80
2.88




260

36.20
26.40




261

15.40
2.17




262

>250.0
3.31




263

>250.0
16.30




264

9.60
2.75




265

>250.0
3.24




266

>250.0
>62.5




267

15.50
2.25




268

6.81
1.62




269

21.10
4.40




270

11.50
2.18




271

21.50
3.51




272

23.70
4.81




273

41.40
8.51




274

>250.0
5.87




275

33.20
6.60




276

37.70
7.74




277

>250.0
>250.0




278

105.00
>250.0




279

14.50
2.96




280

>250.0
>62.5




281

13.60
2.32




282

15.70
3.10




283

11.00
1.95




284

27.90
3.97




285

10.30
1.80




286

46.00
6.03




287

121.00





288

>250.0
4.09




289

10.10
1.86




290

100.00
5.25




291

12.30
2.16




292

45.60
7.44




293

>250.0
14.10




294

50.80
8.12




295

15.20
3.48




296

34.80
6.58




297

21.00
3.36




298

9.92
1.78




299

5.77
1.04




300

7.13
1.36




301

17.40
4.09




302

12.30
3.24




303

9.56
1.57




304

7.24
1.24




305

2.98
0.81




306

9.53
1.64




307

20.60
3.81




308

11.60
2.14




309

8.35
1.61




310

7.36





311
43.50
7.24
0.77




312

4.45





313

>250.0





314

11.70





315

6.22





316

7.85





317

15.50





318
16.20
98.90





319

34.70





320

6.43





321

35.90





322

2.07





323

93.90





324
2.42






325
1.55






326
26.10






327
45.90






328
31.00






329
85.80






330
36.60






331
22.40






332
43.30






333
80.10






334
0.37






335
0.80






336
2.11






337
2.02






338
2.87






339
2.88






340
0.89






341
2.44






342
0.66






343
13.60






344
0.88






345
4.43






346
>250.0

2.73




347
>250.0

1.47




348
235.00

1.22




349
>250.0

3.21




350
237.00

1.53




351
>250.0

3.32




352
>250.0

25.00




353
182.00

92.60




354
>250.0

>62.5




355
155.00

1.70




356
>250.0

>31.3




357
>250.0

6.78




358
>250.0

3.20




359
>250.0

7.66




360
>250.0

>250.0




361
231.00

3.04




362
>250.0

>250.0




363
>250.0

2.22




364
>250.0

8.14











The present invention is described in connection with preferred embodiments. However, it should be appreciated that the invention is not limited to the disclosed embodiments. It is understood that, given the description of the embodiments of the invention herein, various modifications can be made by a person skilled in the art. Such modifications are encompassed by the claims below.

Claims
  • 1. A compound having a structure of formula (I)
  • 2. A compound of claim 1 having a structure of formula (Ia)
  • 3. The compound of claim 1 wherein R1 and R1A is independently H, —C1-6alkyl, —C1-6alkylene-OH, or C1-6alkylene-NR10R11.
  • 4. The compound of claim 3 wherein R1 is H.
  • 5. The compound of claim 3 wherein R1A is H.
  • 6. The compound of claim 1 wherein R2 is H.
  • 7. The compound of claim 1 wherein R3 is independently H or —C1-6alkyl.
  • 8. The compound of claim 7 wherein R3 is H.
  • 9. The compound of claim 1 wherein R4 is H or —C1-6alkyl.
  • 10. The compound of claim 9 wherein R4 is H.
  • 11. The compound of claim 9 wherein R4 is —C1-6alkyl.
  • 12. The compound of claim 11 wherein R4 is —CH3.
  • 13. The compound of claim 1 wherein R4A is independently H, or —C1-6alkyl.
  • 14. The compound of claim 13 wherein R4A is H.
  • 15. The compound of claim 1 wherein R4 and R4A, together with the atoms to which they are attached, form a 6- to 12-membered aryl or heteroaryl, a 3- to 8-membered monocyclic or bicyclic cycloalkyl, a 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl, or a 3- to 8-membered monocyclic or bicyclic heterocycloalkyl group, wherein the heteroaryl and heterocycloalkyl groups have 1, 2, 3 or 4 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl group may include a S═O or SO2.
  • 16. The compound of claim 15 wherein R4 and R4A, together with the atoms to which they are attached, form a heterocycloalkyl ring.
  • 17. The compound of claim 16 wherein R4 and R4A, together with the atoms to which they are attached, form
  • 18. The compound of claim 1 wherein R4B is —C1-6alkyl.
  • 19. The compound of claim 1 wherein R4B is H.
  • 20. The compound of claim 1 wherein R5 is H.
  • 21. The compound of claim 1 wherein R5 is —CH3.
  • 22. The compound of claim 1 wherein R6 is independently H, halo or —C1-6alkyl, a 6- to 12-membered aryl, a 5- to 12-membered heteroaryl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, a 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl group, wherein the heteroaryl, spiroheterocycloalkyl and heterocycloalkyl groups have 1, 2, or 3 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl group may include a S═O or SO2.
  • 23. The compound of claim 22 wherein R6 is H.
  • 24. The compound of claim 1 wherein R7 is independently H, halo or —C1-6alkyl, —O—C1-6alkyl, —S—C1-6alkyl, —NO2, —CN, —CF4, a 6- to 12-membered aryl, 5- to 12-membered heteroaryl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, a 5- to 12-membered spirocycloalkyl or spiroheterocycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl group, wherein the heteroaryl, and heterocycloalkyl groups have 1, 2, or 3 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl, spiroheterocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2.
  • 25. The compound of claim 24 wherein R7 is halo.
  • 26. The compound of claim 25 wherein R7 is Cl.
  • 27. The compound of claim 25 wherein R7 is Br.
  • 28. The compound of claim 1 wherein R8 is —C1-6alkyl.
  • 29. The compound of claim 1 wherein R8 is H.
  • 30. The compound of claim 1, wherein R9 is independently selected from H, OH, —C1-6alkyl, —OC1-6alkyl, —C1-6alkyl-O—C1-6alkyl, halo, —O-haloC1-6alkyl, —CN, —C1-6alkyl-C(═O)—NRaRb, —NRaRb, —(NRaRbRc)n, —OSO2Ra, —SO2Ra, —(CH2CH2O)nCH3, —(═O), —C(═O), —C(═O)Ra, —OC(═O)Ra, —C(═O)ORa, —C(═O)C(═O)ORa, —C(═O)NRaRb, —O—SiRaRbRc, —SiRaRbRc, —O-(3- to 12-membered cycloakyl), —O-(3- to 12-membered heterocycloakyl), —C(═O)-6- to 12-membered aryl, —C(═O)-5- to 12-membered heteroaryl, —C(═O)-3- to 12-membered cycloalkenyl, —C(═O)-3- to 12-membered monocyclic or bicyclic cycloalkyl, —C(═O)-3- to 12-membered monocyclic or bicyclic heterocycloalkyl, —C(═O)-5- to 12-membered spirocycloalkyl, —C(═O)-5- to 12-membered spiroheterocycloalkyl, —C1-6alkyl-C(═O)-6- to 12-membered aryl, —C1-6alkyl-C(═O)-5- to 12-membered heteroaryl, a —C1-6alkyl-C(═O)-3- to 12-membered monocyclic or bicyclic cycloalkyl, a —C1-6alkyl-C(═O)-3- to 12-membered monocyclic or bicyclic heterocycloalkyl, —C1-6alkyl-C(═O)-5- to 12-membered monocyclic or bicyclic spirocycloalkyl, —C1-6alkyl-C(═O)-5- to 12-membered spiroheterocycloalkyl group, 6- to 12-membered aryl, 5- to 12-membered heteroaryl, a 3- to 12-membered cycloalkenyl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl group, a 5- to 12-membered monocyclic or bicyclic spirocycloalkyl, or a 5- to 12-membered monocyclic or bicyclic spiroheterocycloalkyl group, wherein the heteroaryl, heterocycloalkyl and spiroheterocycloalkyl groups have 1, 2, or 3 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2; and still further wherein two adjacent carbon atoms or an adjacent carbon atom and adjacent N atom on a R9 group that includes a cycloalkyl, heterocycloalkyl, aryl, heteroaryl, spirocycloalkyl, or a spiroheterocycloalkyl group may join together to form a 5-6 membered cycloalkyl, heterocycloalkyl, aryl, heteroaryl spirocycloalkyl, or a spiroheterocycloalkyl ring that is unsubstituted or is substituted with 1, 2, or 3 R11 groups.
  • 31. The compound of claim 30, wherein R9 is
  • 32. The compound of claim 1, wherein R10 is independently H, halo, OH, —NH—C1-8alkyl, —N(C1-8alkyl)2, —NH—C0-3alkylene-C6-14aryl, —NH—C0-3alkylene-C2-14heteroaryl, —NH—C0-3alkylene-C3-14cycloalkyl, —NH—C0-3 alkylene-C2-14heterocycloalkyl, cyano, or C1-6alkylene-amine, —C1-6alkyl, —C(═O)NRaRb, —C(═O)ORa, —C(═O), —C0-3alkylene-C3-14cycloalkyl, —Co-3alkylene-C6-14aryl, —C0-3alkylene-C3-14heteroaryl, —C0-3 alkylene-C3-14cycloalkyl, —Co-3alkylene-C2-14heterocycloalkyl, —C1-6alkoxy, —O—C0-3 alkylene-C6-14aryl, —O—C0-3alkylene-C3-14heteroaryl, —O—C0-3 alkylene-C3-14cycloalkyl, —O—C0-3 alkylene-C2-14heterocycloalkyl, —C(═O)-(3- to 10-membered heterocycloalkyl), —C(═O)-6- to 12-membered aryl, —C(═O)-5- to 12-membered heteroaryl, —C(═O)-3- to 12-membered cycloalkenyl, —C(═O)-3- to 12-membered monocyclic or bicyclic cycloalkyl, —C(═O)-3- to 12-membered monocyclic or bicyclic heterocycloalkyl, a —C(═O)-5- to 12-membered bicyclic spirocycloalkyl, a —C(═O)-5- to 12-membered spiroheterocycloalkyl, a 6- to 12-membered aryl, 5- to 12-membered heteroaryl, a 3- to 12-membered cycloalkenyl, a 3- to 12-membered monocyclic or bicyclic cycloalkyl, or a 3- to 12-membered monocyclic or bicyclic heterocycloalkyl, a 5- to 12-membered monocyclic or bicyclic spirocycloalkyl, or a 5- to 12-membered monocyclic or bicyclic spiroheterocycloalkyl group, wherein the heteroaryl, heterocycloalkyl and spiroheterocycloalkyl groups have 1, 2, or 3 heteroatoms independently selected from O, N or S, wherein the cycloalkyl, spirocycloalkyl and heterocycloalkyl groups may include a C═O group, and further wherein the spiroheterocycloalkyl and heterocycloalkyl groups may include a S═O or SO2.
  • 33. The compound of claim 32, wherein R10 is —CH3.
  • 34. The compound of claim 32, wherein R10 is —CH2CH3.
  • 35. The compound of claim 32, wherein R10 is a —C0-3alkylene-cycloalkyl.
  • 36. The compound of claim 32, wherein R10 is —CH2-cycloalkyl.
  • 37. A compound having a structure selected from the formula
  • 38. A compound having a structure selected from the formula
  • 39. The compound of claim 1 in the form of a pharmaceutically acceptable salt.
  • 40. A pharmaceutical formulation comprising the compound of claim 1 and a pharmaceutically acceptable excipient.
  • 41. A method of inhibiting KRAS G12C in a cell, comprising contacting the cell with the compound of claim 1.
  • 42. A method of treating cancer mediated by a KRAS, HRAS or NRAS G12C mutation in a subject comprising administering to the subject a therapeutically effective amount of the compound of claim 1.
  • 43. The method of claim 42, wherein the cancer is lung cancer, pancreatic cancer, endometrial cancer, appendix cancer, small intestine cancer or colorectal cancer.
  • 44. The method of claim 42, wherein the lung cancer is non-small cell lung cancer.
  • 45. The method of claim 42, further comprising administering to the patient in need thereof a therapeutically effective amount of an additional pharmaceutically active compound.
  • 46. The method of claim 45, wherein the additional pharmaceutically active compound is selected from an anti-PD-1 agent and a MEK inhibitor.
  • 47. A kit for treating cancer mediated by a KRAS, HRAS or NRAS G12C mutation, the kit comprising: a compound of claim 1, or the stereoisomer thereof, the atropisomer thereof, the pharmaceutically acceptable salt thereof, the pharmaceutically acceptable salt of the stereoisomer thereof, or the pharmaceutically acceptable salt of the atropisomer thereof; and an additional pharmaceutical active compound.
  • 48. The kit of claim 47, wherein the cancer is non-small cell lung cancer.
CROSS-REFERENCE TO RELATED APPLICATIONS

The present application claims priority to and the benefit of U.S. Provisional Application No. 62/769,497, filed Nov. 19, 2018, which is incorporated herein by reference in its entirety for all purposes.

US Referenced Citations (70)
Number Name Date Kind
4232027 Turk et al. Nov 1980 A
5100883 Schiehser Mar 1992 A
5118677 Caufield Jun 1992 A
5118678 Kao et al. Jun 1992 A
5120842 Failli et al. Jun 1992 A
5151413 Caufield et al. Sep 1992 A
5256790 Nelson Oct 1993 A
5258389 Goulet et al. Nov 1993 A
5521184 Zimmermann May 1996 A
5650415 Tang et al. Jul 1997 A
5656643 Spada et al. Aug 1997 A
5712291 D'Amato Jan 1998 A
5728813 Lyman et al. Mar 1998 A
5747498 Schnur et al. May 1998 A
5770599 Gibson Jun 1998 A
5789427 Chen et al. Aug 1998 A
5792783 Tang et al. Aug 1998 A
5861510 Piscopio et al. Jan 1999 A
5863949 Robinson et al. Jan 1999 A
5892112 Levy et al. Apr 1999 A
5969110 Beckmann et al. Oct 1999 A
5981245 Fox et al. Nov 1999 A
5990141 Hirth et al. Nov 1999 A
6057124 Bartley et al. May 2000 A
6111090 Gorman et al. Aug 2000 A
6232447 Cerretti May 2001 B1
6235764 Larson et al. May 2001 B1
6258812 Bold et al. Jul 2001 B1
6413932 Cerretti et al. Jul 2002 B1
6515004 Misra et al. Feb 2003 B1
6596852 Cerretti et al. Jul 2003 B2
6630500 Gingrich et al. Oct 2003 B2
6656963 Firestone et al. Dec 2003 B2
6713485 Carter et al. Mar 2004 B2
6727225 Wiley Apr 2004 B2
7025962 Gorman et al. Apr 2006 B1
7361760 Sircar et al. Apr 2008 B2
7618632 Collins et al. Nov 2009 B2
7812135 Smith et al. Oct 2010 B2
8388967 Smith et al. Mar 2013 B2
8586023 Shiku et al. Nov 2013 B2
8591886 Ponath et al. Nov 2013 B2
10519146 Lanman et al. Dec 2019 B2
10532042 Lanman et al. Jan 2020 B2
10640504 Lanman et al. May 2020 B2
20020042368 Fanslow, III et al. Apr 2002 A1
20030105091 Riedl et al. Jun 2003 A1
20030162712 Cerretti et al. Aug 2003 A1
20090012085 Baum et al. Jan 2009 A1
20140288045 Ren et al. Sep 2014 A1
20150239900 Li et al. Aug 2015 A1
20160159738 Ren et al. Jun 2016 A1
20160166571 Janes et al. Jun 2016 A1
20160297774 Li et al. Oct 2016 A1
20180015087 Liu et al. Jan 2018 A1
20180072723 Blake et al. Mar 2018 A1
20190336514 Wurz et al. Nov 2019 A1
20190343838 Allen et al. Nov 2019 A1
20190345169 Minatti et al. Nov 2019 A1
20190374542 Allen et al. Dec 2019 A1
20190375749 Chen et al. Dec 2019 A1
20200030324 Booker et al. Jan 2020 A1
20200055845 Lanman et al. Feb 2020 A1
20200069657 Lanman et al. Mar 2020 A1
20200207766 Lanman et al. Jul 2020 A1
20200216446 Parsons et al. Jul 2020 A1
20200222407 Lipford et al. Jul 2020 A1
20200360374 Henary et al. Nov 2020 A1
20200369662 Chaves et al. Nov 2020 A1
20210009577 Lanman et al. Jan 2021 A1
Foreign Referenced Citations (159)
Number Date Country
19629652 Jan 1998 DE
0090505 Oct 1983 EP
0520722 Dec 1992 EP
0566226 Oct 1993 EP
0606046 Jul 1994 EP
0682027 Nov 1995 EP
0407122 Oct 1996 EP
0770622 May 1997 EP
0780386 Jun 1997 EP
0787772 Aug 1997 EP
0818442 Jan 1998 EP
0837063 Apr 1998 EP
0931788 Jul 1999 EP
0970070 Jan 2000 EP
1004578 May 2000 EP
1181017 Feb 2002 EP
1786785 May 2007 EP
1866339 Dec 2007 EP
1947183 Jul 2008 EP
3401314 Nov 2019 EP
3055290 Dec 2019 EP
02233610 Sep 1990 JP
1990005719 May 1990 WO
1992005179 Feb 1992 WO
1992020642 Nov 1992 WO
1993011130 Jun 1993 WO
1994002136 Feb 1994 WO
1994002485 Feb 1994 WO
1994009010 Apr 1994 WO
1995009847 Apr 1995 WO
1995014023 May 1995 WO
1995016691 Jun 1995 WO
1995019774 Jul 1995 WO
1995019970 Jul 1995 WO
1996027583 Sep 1996 WO
1996030347 Oct 1996 WO
1996031510 Oct 1996 WO
1996033172 Oct 1996 WO
1996033980 Oct 1996 WO
1996041807 Dec 1996 WO
1997002266 Jan 1997 WO
1997013771 Apr 1997 WO
1997019065 May 1997 WO
1997027199 Jul 1997 WO
1997030034 Aug 1997 WO
1997030044 Aug 1997 WO
1997032880 Sep 1997 WO
1997032881 Sep 1997 WO
1997034895 Sep 1997 WO
1997038983 Oct 1997 WO
1997038994 Oct 1997 WO
1997049688 Dec 1997 WO
1998002434 Jan 1998 WO
1998002437 Jan 1998 WO
1998002438 Jan 1998 WO
1998002441 Jan 1998 WO
1998003516 Jan 1998 WO
1998007697 Feb 1998 WO
1998007726 Feb 1998 WO
1998014449 Apr 1998 WO
1998014450 Apr 1998 WO
1998014451 Apr 1998 WO
1998017662 Apr 1998 WO
1998030566 Jul 1998 WO
1998033768 Aug 1998 WO
1998033798 Aug 1998 WO
1998034915 Aug 1998 WO
1998034918 Aug 1998 WO
1999007675 Feb 1999 WO
1999007701 Feb 1999 WO
1999020758 Apr 1999 WO
1999029667 Jun 1999 WO
1999035132 Jul 1999 WO
1999035146 Jul 1999 WO
1999040196 Aug 1999 WO
1999045009 Sep 1999 WO
1999052889 Oct 1999 WO
1999052910 Oct 1999 WO
1999061422 Dec 1999 WO
2000002871 Jan 2000 WO
2000012089 Mar 2000 WO
2000059509 Oct 2000 WO
2001003720 Jan 2001 WO
2001014387 Mar 2001 WO
2001032651 May 2001 WO
2001037820 May 2001 WO
2002055501 Jul 2002 WO
2002059110 Aug 2002 WO
2002066470 Aug 2002 WO
2002068406 Sep 2002 WO
2004005279 Jan 2004 WO
2004007458 Jan 2004 WO
2004007481 Jan 2004 WO
2004009784 Jan 2004 WO
2005005434 Jan 2005 WO
2005007190 Jan 2005 WO
2005011700 Feb 2005 WO
2005016252 Feb 2005 WO
2005021546 Mar 2005 WO
2005055808 Jun 2005 WO
2005115451 Dec 2005 WO
2006044453 Apr 2006 WO
2006083289 Aug 2006 WO
2006121168 Nov 2006 WO
2006122806 Nov 2006 WO
2007133822 Nov 2007 WO
2008070740 Jun 2008 WO
2009036082 Mar 2009 WO
2009055730 Apr 2009 WO
2010003118 Jan 2010 WO
2011028683 Mar 2011 WO
2011051726 May 2011 WO
2011090754 Jul 2011 WO
2012142498 Oct 2012 WO
2013039954 Mar 2013 WO
2013155223 Oct 2013 WO
2014143659 Sep 2014 WO
2014152588 Sep 2014 WO
2015001076 Jan 2015 WO
2015054572 Apr 2015 WO
2015075483 May 2015 WO
2016044772 Mar 2016 WO
2016049524 Mar 2016 WO
2016049565 Mar 2016 WO
2016049568 Mar 2016 WO
2016164675 Oct 2016 WO
2016168540 Oct 2016 WO
2017015562 Jan 2017 WO
2017058728 Apr 2017 WO
2017058768 Apr 2017 WO
2017058792 Apr 2017 WO
2017058805 Apr 2017 WO
2017058807 Apr 2017 WO
2017058902 Apr 2017 WO
2017058915 Apr 2017 WO
2017087528 May 2017 WO
2017100546 Jun 2017 WO
2017172979 Oct 2017 WO
2017201161 Nov 2017 WO
2018064510 Apr 2018 WO
2018068017 Apr 2018 WO
2018119183 Jun 2018 WO
2018140598 Aug 2018 WO
2018217651 Nov 2018 WO
2018218069 Nov 2018 WO
2019051291 Mar 2019 WO
2019213516 Nov 2019 WO
2019213526 Nov 2019 WO
2019217691 Nov 2019 WO
2019232419 Dec 2019 WO
2019241157 Dec 2019 WO
2019243533 Dec 2019 WO
2019243535 Dec 2019 WO
2020050890 Mar 2020 WO
2020102730 May 2020 WO
2020106640 May 2020 WO
2020232130 Nov 2020 WO
2020236947 Nov 2020 WO
2020236948 Nov 2020 WO
Non-Patent Literature Citations (157)
Entry
AMG-510; CS-0081316; Source: AbaChemScene (CS-0081316); Deposit Date: May 13, 2019 Available Date: May 13, 2019; SID: 384060804[CID: 137278711] (available at https://pubchem.ncbi.nlm.nih.gov/substance/384060804/).
AMG-510; HY-114277; Source: MedChemexpress MCE (HY-114277); Deposit Date: May 13, 2019 Available Date: May 13, 2019; SID: 384060569[CID: 137278711] (available at https://pubchem.ncbi.nlm.nih.gov/substance/384060569).
Canon, et al., “The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity,” Nature, 575(7781): 217-223 (2019) (Supplementary Material, pp. 1-55).
Database Registry [Online], Chemical Abstracts Service, Columbus, Ohio, US; Jul. 31, 2017 (Jul. 31, 2017), XP002801805, retrieved from STN Database accession No. 2105944-09-8.
Meanwell, “Synopsis of Some Recent Tactical Application of Bioisosteres in Drug Design,” J. Med. Chem. 54:2529-2591 (2011).
Non-Final Office Action for U.S. Appl. No. 16/675,121, dated Feb. 2, 2021, 10 pages.
Notice of Allowance dated Jan. 14, 2021 for U.S. Appl. No. 16/402,589, 5 pages.
Notice of Allowance, dated Dec. 21, 2020, for U.S. Appl. No. 16/407,889, 5 pages.
Notice of Allowance, dated Jan. 26, 2021, for U.S. Appl. No. 16/438,349, 9 pages.
Notice of Allowance, dated Jan. 27, 2021, for U.S. Appl. No. 16/428,163, 9 pages.
Notice of Allowance, dated Nov. 16, 2020, for U.S. Appl. No. 16/402,538, 8 pages.
Shibata et al., “A Convenient Synthesis of 3-Cyano-2-methylpyridines under Ultrasonic Irradiation,” Bull. Chem. Soc. Jpn., 61:2199-2200 (1988).
Stanetty et al., “Synthesis of Aza Analogs of the Herbicide Sindone B,” Monatshefte Fuer Chemie, 130:441-450 (1999).
Third Party Observation filed for PCT/US2020/033831, submitted Jan. 15, 2021, 2 pages.
“A Phase 1, Study Evaluating the Safety, Tolerability, PK, and Efficacy of AMG 510 in Subjects With Solid Tumors With a S Mutation.” NCT03600883, comparison of version submitted Oct. 29, 2018 and Oct. 9, 2019 (update posted Oct. 10, 2019), for full history of change see https://clinicaltrials.gov/ct2/history/NCT03600883 (last accessed Nov. 11, 2020), pp. 1-22.
“Acute Leukemia,” The Merck Manual (Online Edition), pp. 1-6 (2013).
“KRASG12C Inhibitor,” Mirati Therapeutics, retrieved on Nov. 27, 2018, from https://www.mirati.com/mrtx849/, 5 pages.
Ahmadian, et al., “Guanosine triphosphatase stimulation of oncogenic Ras mutants,” PNAS, 96: 7065-7070, 1999.
Airoldi, et al., “Glucose-Derived Ras Pathway Inhibitors: Evidence of Ras—Ligand Binding and Ras—GEF (Cdc25) Interaction Inhibition,” ChemBioChem, 8: 1376-1379 (2007).
Attc “Organism: Mus musculus (B cell); Mus musculus (myeloma), mouse (B cell); mouse (myeloma),” Accession No. HB-8508, retrieved from https://www.atcc.org/˜/media/0DF7351153724BD6A3E7D78D5BA2F933.ashx, on Nov. 29, 2018.
Barnett, et al., “Identification and characterization of pleckstrin-holomogy-domain-dependent and isoenzyme specific Akt inhibitors,” Biochem. J.,385 (2): 399-408 (2005).
Bhatia, et al., “A Review on Bioisosterism: A Rational Approach for Drug Design and Molecular Modification,” Pharmacologyonline, 1:272-299 (2011).
Bull, et al., “Isoquino[2,1-c][1,3,2] Benzodiazaphosphorine Derivatives: New Potential Agents for Cancer Chemotherapy,” Phosphorus, Sulfur, and Silicon, 162:231-243 (2000).
Campillo, et al., “Novel Bronchodilators: Synthesis, Transamination Reactions, and Pharmacology of a Series of Pyrazino[2,3-c][1,2,6]thiadiazine 2,2-Dioxides,” J. Med. Chem., 43: 4219-4227 (2000).
Canon, et al., “The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity,” Nature, 575(7781): 217-223 (2019).
Cee, et al.,“Discovery of AMG 510, a first-in-humancovalent inhibitor of KRASG12C for the treatment of solid tumors,” Abstract and Presentation, ACS Spring Meeting, Orlando, FL, USA, Mar. 31-Apr. 4, 2019.
Cohen, “The development and therapeutic potential of protein kinase inhibitors,” Current Opinion in Chemical Biology, 3:459-465 (1999).
Cowen Slide deck—Warp Drive Bio, slides 1-32, “Corporate Overview Exploiting the Molecules and Mechanisms of Nature to Create Transformative Medicines” http://www.warpdrivebio.com/news/cowen%202016.pdf (last visited Apr. 2016).
Dasmahapatra, et al., “In vitro Combination Treatment with Perifosine and UCN-01 Demonstrates Synergism Against Prostate (PC-3) and Lung (A549) Epithelial Adenocarcinoma Cell Lines,” Clin. Cancer Res. 10(15): 5242-5252 (2004).
Dermer, et al., “Another Anniversary for the War on Cancer,” Bio/Technology, 12: 320 (1994).
Douelle, et al., “Highly Diastereoselective Synthesis of vicinal Quaternary and Tertiary Stereocenters Using the Iodo-aldol Cyclization,” Org. Lett., 9 (10): 1931-1934 (2007).
Erkkilä, et al., “Mild Organocatalytic α-Methylenation of Aldehydes,” J. Org. Chem.,71 (6), 2538-2541 (2006).
Extended European Search Report for European Patent Application No. 19208193.2, dated Jun. 3, 2020, pp. 1-8.
Fakih, et al., “Phase 1 study evaluating the safety, tolerability, pharmacokinetics (PK), and efficacy of AMG 510, a novel small molecule KRASG12C inhibitor, in advanced solid tumors,” Journal of Clinical Oncology, 37(15 suppl) (May 20, 2019) 3003, published online May 26, 2019.
Fakih, et al., “Phase 1 study evaluating the safety, tolerability, pharmacokinetics (PK), and efficacy of AMG 510, a novel small molecule KRASG12C inhibitor, in advanced solid tumors,” Presentation, ASCO, Chicago, IL, USA, May 31-Jun. 4, 2019.
Final Office Action for U.S. Appl. No. 15/984,855, dated Mar. 28, 2019, 7 pages.
Final Office Action for U.S. Appl. No. 16/661,907, dated Mar. 27, 2020, 29 pages.
Freshney, et al., Culture of Animal Cells, A Manual of Basic Technique, Alan R. Liss, Inc, New York, p. 4 (1983).
Gentile, et al., “Discovery and Structural Investigation of Novel Binders to the Ras Switch II Pocket,” NCI Initiative Symposium Poster (2015).
Gills and Dennis, “The development of phosphatidylinositol ether lipid analogues as inhibitors of the serine/threonine kinase, Akt,” Expert. Opin. Investig. Drugs, 13: 787-797 (2004).
Goldberg, et al., “Role of PD-1 and its ligand, B7-H1, in early fate decisions of CD8 T cells,” Blood,110(1): 186-192 (2007).
Goldstein, et al., “Biological efficacy of a chimeric antibody to the epidermal growth factor receptor in a human tumor xenograft model,” Clin. Cancer Res., 1: 1311-1318 (1995).
Golub, et al., “Molecular Classification of Cancer: Class Discovery and Class Prediction by Gene Expression Monitoring,” Science, 286: 531-537(1999).
Govindan, et al., “Safety, Efficacy, and Pharmacokinetics of AMG 510, a Novel KRASG12C Inhibitor, in Patients with Non-Small Cell Lung Cancer,” Abstract and Presentation, North American Conference on Lung Cancer (NACLC), Chicago, IL, USA, Oct. 10-12, 2019.
Govindan, et al., “Phase 1 Study of AMG 510, a Novel KRAS G12C Inhibitor, in Advanced Solid Tumors with KRAS p. G12C Mutation,” Abstract, ESMO Congress, Barcelona, Spain, Sep. 27-Oct. 1, 2019.
Govindan, et al., “Phase 1 Study of AMG 510, a Novel KRAS G12C Inhibitor, in Advanced Solid Tumors with KRAS p. G12C Mutation,” Poster, ESMO Congress, Barcelona, Spain, Sep. 27-Oct. 1, 2019.
Govindan, et al., “Phase 1 Study of Safety, Tolerability, Pharmacokinetics, and Efficacy of AMG510, a Novel KRASG12C Inhibitor, in Non-Small Cell Lung Cancer,” Abstract and Presentation, World Conference on Lung Cancer (WCLC), Barcelona, Spain, Sep. 7-10, 2019.
Gura, “Cancer Models: Systems for Identifying New Drugs Are Often Faulty,” Science, 278(5340):1041-1042 (1997).
Halford, “Amgen unveils its Kras covalent inhibitor AMG 510,” Chemical & Engineering News 97(14):4 (2019).
Hallin, et al., “The KRASG12C Inhibitor MRTX849 Provides Insight toward Therapeutic Susceptibility of KRAS-Mutant Cancers in Mouse Models and Patients,” Cancer Discov., 10: 54-71 (2020).
Hansen, et al., “Abstract 686: Drugging an undruggable pocket: the biochemical mechanism of covalent KRASG12C inhibitors,” Proceedings of the American Association for Cancer Research Annual Meeting 2018; Apr. 14-18, 2018; Chicago, IL; AACR; Cancer Res., 78(13 Suppl): Abstract 686 (2018).
Hichri, et al., “A Convenient Synthesis of 1,3,2-Benzodiazaphophorine-2-Oxide,” Phosphorus, Sulfur, and Silicon, 190: 29-35 (2015).
Hichri, et al., CAPLUS Abstract, 162:245378 (2015).
Hocker, et al., “Andrographolide derivatives inhibit guanine nucleotide exchange and abrogate oncogenic Ras function,” PNAS, 110(25): 10201-10206 (2013).
Huang, et al., “Epidermal Growth Factor Receptor Blockade with C225 Modulates Proliferation, Apoptosis, and Radiosensitivity in Squamous Cell Carcinomas of the Head and Neck,” Cancer Res., 59(8): 1935-1940 (1999).
International Search Report for PCT/US2017/067801, dated Jul. 25, 2018, 6 pages.
International Search Report for PCT/US2018/033714, dated Jul. 17, 2018, 3 pages.
International Search Report for PCT/US2018/050044, dated Oct. 30, 2018, 7 pages.
International Search Report for PCT/US2019/030593, dated Aug. 6, 2019, 4 pages.
International Search Report for PCT/US2019/030606, dated Jul. 23, 2019, 5 pages.
International Search Report for PCT/US2019/031535, dated Jul. 25, 2019, 7 pages.
International Search Report for PCT/US2019/034974, dated Aug. 9, 2019, 5 pages.
International Search Report for PCT/US2019/036397, dated Aug. 26, 2019, 5 pages.
International Search Report for PCT/US2019/036626, dated Jun. 2, 2020, 5 pages.
International Search Report for PCT/US2019/061815, dated Mar. 5, 2020, 6 pages.
International Search Report for PCT/US2019/062051, dated Mar. 2, 2020, 3 pages.
International Search Report for PCT/US2020/032686, dated Aug. 14, 2020, 4 pages.
International Search Report for PCT/US2020/033831, dated Jul. 9, 2020, 6 pages.
International Search Report for PCT/US2020/033832, dated Jul. 8, 2020, 4 pages.
Janes, et al., “Targeting KRAS Mutant Cancers with a Covalent G12C-Specific Inhibitor,” Cell, 172: 578-589 (2018).
Jarvis, “Notorious KRAS: Taking down cancer researchers' biggest foe,” Chemical & Engineering News, 97(37), 9 pages (2019).
Jin, et al., “Inhibition of AKT survival pathway by a small molecule inhibitor in human endometrial cancer cells,” Br. J. Cancer, 91: 1808-1812 (2004).
Johnson et al., “Relationships between drug activity in NCI preclinical in vitro and in vivo models and early clinical trials,” British Journal of Cancer, 84(10): 1424-1431 (2001).
Lanman, et al., “Abstract 4455: Discovery of AMG 510, a first-in-human covalent inhibitor of KRASG12C for the treatment of solid tumors,” Presentation, American Association for Cancer Research (AACR) Annual Meeting, Atlanta, GA, USA, Mar. 29-Apr. 3, 2019.
Lanman, et al., “Abstract 4455: Discovery of AMG 510, a first-in-human covalent inhibitor of KRASG12C for the treatment of solid tumors,” Proceedings of the American Association for Cancer Research Annual Meeting 2019; Mar. 29-Apr. 3, 2019; Atlanta, GA, USA, AACR, Cancer Res. 79(13 Suppl): Abstract nr 4455 (2019).
Lamian, et al., “Discovery of a Covalent Inhibitor of KRASG12C (AMG 510) for the Treatment of Solid Tumors,” J. Med Chem., 63: 52-65 (2020).
Li, et al., “Targeting Protein—Protein Interaction with Covalent Small-Molecule Inhibitors,” Current Topics in Medicinal Chemistry, 19(21): 1872-1876 (2019).
Lim, et al., “Therapeutic Targeting of Oncogenic K-Ras by a Covalent Catalytic Site Inhibitor,” Angew. Chem. Int. Ed, 53: 199-204 (2014).
Lipford, et al., “Pre-Clinical Development of AMG 510: The First Inhibitor of KRASG12C in Clinical Testing,” Presentation, American Association for Cancer Research (AACR) Annual Meeting, Atlanta, GA, USA, Mar. 29-Apr. 3, 2019.
Liu, Y., “Session SY28—Transformative Small Molecule Therapies—Targeting KRAS mutant cancers with a covalent G12C—specific inhibitor,” Presentation on Apr. 4, 2017, AACR Annual Meeting Presentation, Apr. 1-5, 2017, Washington, D.C. (2017).
Lopez, et al.,“Optimization of quinazolinone-based covalent inhibitors of KRASG12C in the discovery of AMG 510,” Abstract and Poster, ACS Fall Meeting, San Diego, CA, USA, Aug. 25-29, 2019.
Lu, et al., “KRAS G12C Drug Development: Discrimination between Switch II Pocket Configurations Using Hydrogen/Deuterium-Exchange Mass Spectrometry,” Structure, 25: 1-7 (2017).
Maurer, et al., “Small-molecule ligands bind to a distinct pocket in Rad and inhibit SOS-mediated nucleotide exchange activity,” PNAS, 109(14): 5299-5304 (2012).
McGregor, et al., “Expanding the Scope of Electrophiles Capable of Targeting K-Ras Oncogenes,” ACS Bio. Chem., 56: 3179-3183 (2017).
Mirati Therapeutics, “Corporate Presentation Nov. 2017,” Slides 1-41 (2017).
Modjtahedi, et al., “The human EGF receptor as a target for cancer therapy: six new rat mAbs AGainst the receptor on the breast carcinoma MDA-MB 468,” Br. J. Cancer, 67(2): 247-253 (1993).
Morrissey et al., “Immunotherapy and Novel Combinations in Oncology: Current Landscape, Challenges, and Opportunities,” Clin. Transl. Sci., 9(2):89-104 (2016).
National Cancer Institute identifier: NSC 154020, retrieved on Nov. 29, 2018, from https://cactus.nci.nih.gov/ncidb2.2/.
NCBI Reference Sequence, “GTPase KRas isoform a [Homo sapiens],” GenBank Accession No. NM_203524.1, Retrieved on Nov. 29, 2018 from https://www.ncbi.nlm.nih.gov/protein/15718763?sat=4&satkey=234448549, 4 pages.
Non-Final Office Action (Corrected) for U.S. Appl. No. 16/125,359, dated Apr. 8, 2019, 13 pages.
Non-Final Office Action for U.S. Appl. No. 15/849,905, dated Mar. 20, 2019, 18 pages.
Non-Final Office Action for U.S. Appl. No. 15/984,855, dated Sep. 27, 2018, 25 pages.
Non-Final Office Action for U.S. Appl. No. 16/125,359, dated Apr. 5, 2019, 13 pages.
Non-Final Office Action for U.S. Appl. No. 16/402,538, dated Oct. 30, 2019, 12 pages.
Non-Final Office Action for U.S. Appl. No. 16/402,589, dated Mar. 6, 2020, 17 pages.
Non-Final Office Action for U.S. Appl. No. 16/407,889, dated Jul. 1, 2020, 6 pages.
Non-Final Office Action for U.S. Appl. No. 16/428,163, dated Sep. 15, 2020, 6 pages.
Non-Final Office Action for U.S. Appl. No. 16/436,647, dated Aug. 7, 2020, 19 pages.
Non-Final Office Action for U.S. Appl. No. 16/438,349, dated Dec. 13, 2019, 15 pages.
Non-Final Office Action for U.S. Appl. No. 16/661,907, dated Nov. 18, 2019, 20 pages.
Notice of Allowance dated Jul. 24, 2020 for U.S. Appl. No. 16/402,538, 8 pages.
Notice of Allowance dated Sep. 16, 2020 for U.S. Appl. No. 16/402,589, 5 pages.
Notice of Allowance dated Sep. 9, 2020 for U.S. Appl. No. 16/438,349, 9 pages.
Ostrem, et al., “Development of mutant-specific small molecule inhibitors of K-Ras,” Poster, AACR 104th Annual Meeting 2013; Apr. 6-10, 2013; Washington, D.C. (2013).
Ostrem, et al., “K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions,” Nature, 503: 548-551 (2013).
Paez, et al., “EGFR Mutations in Lung Cancer Correlation with Clinical Response to Gefitinib Therapy,” Science, 304(5676): 1497-500 (2004).
Palmioli, et al., “First experimental identification of Ras-inhibitor binding interface using a water-soluble Ras ligand,” Bioorg. Med. Chem. Lett., 19: 4217-4222 (2009).
Patricelli, et al., “Selective Inhibition of Oncogenic KRAS Output with Small Molecules Targeting the Inactive State,” Cancer Discov, 6 (3): 316-329 (2016).
Pearce, et al., “Failure modes in anticancer drug discovery and development,” Cancer Drug Design and Discovery, Edited by Stephen Neidle, Chapter 18, pp. 424-435 (2008).
Peri, et al., “Design, Synthesis and Biological Evaluation of Sugar-Derived Ras Inhibitors,” ChemBioChem, 6: 1839-1848 (2005).
Peri, et al., “Sugar-Derived Ras Inhibitors: Group Epitope Mapping by NMR Spectroscopy and Biological Evaluation,” Eur. J. Org. Chem., 16: 3707-3720 (2006).
Peters, et al., “Selective inhibition of K-Ras G12C through allosteric control of GTP affinity and effector interactions,” EORTC Poster (2013).
Remington's Pharmaceutical Sciences, 1435-1712 (18th ed., Mack Publishing Co, Easton, Pennsylvania, 1990 (Table of Contents Only).
Rex et al., “KRAS—AACR 2018,” Amgen Collection of Information published at Proceedings of the American Association for Cancer Research Annual Meeting 2018; Apr. 14-18, 2018; Chicago, IL; AACR; slides 1-24 (2018).
Rex, et al., “Abstract 3090: In vivo characterization of AMG 510—a potent and selective KRASG12C covalent small molecule inhibitor in preclinical KRASG12C cancer models,” Poster, American Association for Cancer Research (AACR) Annual Meeting, Atlanta, GA, USA, Mar. 29-Apr. 3, 2019.
Rex, et al., “Abstract 3090: In vivo characterization of AMG 510—a potent and selective KRASG12C covalent small molecule inhibitor in preclinical KRASG12C cancer models,” Proceedings of the American Association for Cancer Research Annual Meeting 2019; Mar. 29-Apr. 3, 2019; Atlanta, GA, USA, AACR, Cancer Res. 79(13 Suppl): Abstract nr 3090 (2019).
Saiki, et al., “Abstract 4484: Discovery and in vitro characterization of AMG 510—a potent and selective covalent small-molecule inhibitor of KRASG12C,” Presentation, American Association for Cancer Research (AACR) Annual Meeting, Atlanta, GA, USA, Mar. 29-Apr. 3, 2019.
Saiki, et al., “Abstract 4484: Discovery and in vitro characterization of AMG 510—a potent and selective covalent small-molecule inhibitor of KRASG12C,” Proceedings of the American Association for Cancer Research Annual Meeting 2019; Mar. 29-Apr. 3, 2019; Atlanta, GA, USA, AACR, Cancer Res. 79(13 Suppl): Abstract nr 4484 (2019).
Sarkar, et al., “Indole-3-Carbinol and Prostate Cancer1,2,” J. Nutr., 134(12 Suppl): 3493S-3498S (2004).
Shima, et al., “In silico discovery of small-molecule Ras inhibitors that display antitumor activity by blocking the Ras—effector interaction,” PNAS, 110(20): 8182-8187 (2013).
Simone, “Part XIV Oncology: Introduction,” Cecil Textbook of Medicine, 20th Edition, 1:1004-1010 (1996).
Singh, et al., “Improving Prospects for Targeting RAS,” J. Clinc. Oncl, 33(31): 3650-3660 (2015).
Statsyuk, “Let K-Ras activate its own inhibitor,” Nature Structural & Molecular Biology, 25:435-439 (2018).
Sun, et al., “Discovery of Small Molecules that Bind to K-Ras and Inhibit Sos-Mediated Activation,” Angew. Chem. Int. Ed., 51: 6140-6143 (2012).
Taveras, et al., “Ras Oncoprotein Inhibitors: The Discovery of Potent, Ras Nucleotide Exchange Inhibitors and the Structural Determination of a Drug-Protein Complex,” Biorg. Med. Chem. Lett.,, 5(1): 125-133 (1997).
Teramoto, et al., 1996, Cancer 77 (4):639-645.
The ASCO Post Staff, “AACR-NCI-EORTC: Investigational KRAS G12C Inhibitor for KRAS-Mutant Solid Tumors,” The ASCO Post (2019).
Thompson, et al., “PD-1 Is Expressed by Tumor-Infiltrating Immune Cells and Is Associated with Poor Outcome for Patients with Renal Cell Carcinoma,” Clin. Cancer Res., 13(6): 1757-1761 (2007).
Traxler, “Tyrosine kinase inhibitors in cancer treatment (Part II),” Exp. Opin. Ther. Patents, 8(12): 1599-1625 (1998).
U.S. Appl. No. 60/528,340, filed Dec. 9, 2003.
Wang, et al., “Ras inhibition via direct Ras binding—is there a path forward?,” Bioorg. Med. Chem. Lett., 22: 5766-5776 (2012).
Written Opinion for PCT/US2017/067801, dated Jul. 25, 2018, 10 pages.
Written Opinion for PCT/US2018/033714, dated Jul. 17, 2018, 5 pages.
Written Opinion for PCT/US2018/050044, dated Oct. 30, 2018, 7 pages.
Written Opinion for PCT/US2019/030593, dated Aug. 6, 2019, 5 pages.
Written Opinion for PCT/US2019/030606, dated Jul. 23, 2019, 6 pages.
Written Opinion for PCT/US2019/031535, dated Jul. 25, 2019, 7 pages.
Written Opinion for PCT/US2019/034974, dated Aug. 9, 2019, 5 pages.
Written Opinion for PCT/US2019/036397, dated Aug. 26, 2019, 5 pages.
Written Opinion for PCT/US2019/036626, dated Jun. 2, 2020, 12 pages.
Written Opinion for PCT/US2019/061815, dated Mar. 5, 2020, 4 pages.
Written Opinion for PCT/US2019/062051, dated Mar. 2, 2020, 5 pages.
Written Opinion for PCT/US2020/032686, dated Aug. 14, 2020, 6 pages.
Written Opinion for PCT/US2020/033831, dated Jul. 9, 2020, 7 pages.
Written Opinion for PCT/US2020/033832, dated Jul. 8, 2020, 6 pages.
Xiong, et al., “Covalent Guanosine Mimetic Inhibitors of G12C KRAS,” ACS Med. Chem. Lett., 8: 61-66 (2017).
Yan, et al., “Pharmacogenetics and pharmacogenomics in oncology therapeutic antibody development,” BioTechniques, 29(4): 565-568 (2005).
Yang, et al., “Akt/Protein Kinase B Signaling Inhibitor-2, a Selective Small Molecule Inhibitor of Akt Signaling with Antitumor Activity in Cancer Cells Overexpressing Akt,” Cancer Res., 64, 4394-4399 (2004).
Yang, et al., “Eradication of Established Tumors by a Fully Human Monoclonal Antibody to the Epidermal Growth Factor Receptor without Concomitant Chemotherapy,” Cancer Res., 59: 1236-1243 (1999).
Zeng, et al., “Potent and Selective Covalent Quinazoline Inhibitors of KRAS G12C,” Cell Chemical Biology, 24: 1-12 (2017).
Zimmerman, et al., “Small molecule inhibition of the KRAS—PDEδ interaction impairs oncogenic KRAS signaling,” Nature, 1-5 (2017).
International Search Report for PCT/US2019/62064, dated Oct. 29, 2020, 9 pages.
Written Opinion for PCT/US2019/62064, dated Oct. 29, 2020, 13 pages.
Hong, et al.,“KRASG12C Inhibition with Sotorasib in Advanced Solid Tumors,” N. Engl. Med., 383:1207-1217 (2020).
Final Office Action for U.S. Appl. No. 16/436,647, dated Mar. 24, 2021, 7 pages.
Non-Final Office Action for U.S. Appl. No. 16/817,109, dated Mar. 3, 2021, 12 pages.
Notice of Allowance, dated Mar. 30, 2021, for U.S. Appl. No. 16/402,538, 8 pages.
Related Publications (1)
Number Date Country
20200165231 A1 May 2020 US
Provisional Applications (1)
Number Date Country
62769497 Nov 2018 US