SMALL MOLECULE MODULATORS OF FERROPTOSIS

Information

  • Patent Application
  • 20240315994
  • Publication Number
    20240315994
  • Date Filed
    February 14, 2024
    9 months ago
  • Date Published
    September 26, 2024
    a month ago
Abstract
Provided herein are inhibitors of GPX4, pharmaceutical compositions comprising the inhibitory compounds, and methods for using the GPX4 inhibitory compounds for the modulation of ferroptosis and treatment of disease.
Description
FIELD OF THE INVENTION

The invention relates to compounds, compositions, and methods for inducing ferroptosis in a cell.


BACKGROUND OF THE INVENTION

Efforts to inhibit GPX4 have focused on the use of electrophiles capable of forming covalent bonds with a selenocysteine residue in the GPX4 active site. Despite its clear biological role in protecting cancer cells from ferroptosis, to date, no targeted GPX4 inhibitors have been profiled in the clinic. There is a need in the art for novel modulators of GPX4.


INCORPORATION BY REFERENCE

All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. To the extent publications and patents or patent applications incorporated by reference contradict the disclosure contained in the specification, the specification is intended to supersede and/or take precedence over any such contradictory material.





BRIEF DESCRIPTION OF DRAWINGS


FIG. 1 is a plot showing the relative viability of HT-1080 fibrosarcoma cells exposed to each compound in the inhibitor screening panel.



FIG. 2A, FIG. 2B show the relative viability of HT-1080 fibrosarcoma cells exposed to inhibitor compounds in the presence and absence of 1 μM Liproxstatin-1.



FIG. 3 is a panel of plots showing the relative viability of HT-1080 fibrosarcoma cells exposed to inhibitor compounds in the presence and absence of 1.5 μM ferrostatin-1.



FIG. 4A is a panel of plots showing fluorescence images of Biodipy-C11 staining of cells treated with the five ferroptosis-inducing compounds identified in FIG. 3.



FIG. 4B is a panel of plots showing fluorescence images of Biodipy-C11 staining of cells treated with compound 8216 and the controls RSL3 and sodium arsenite.



FIG. 5A is a plot showing the mass spectrum for positive control ML162 binding to recombinant GPX4 in vitro.



FIG. 5B shows the mass spectrum for the negative control, 0568, binding to recombinant GPX4 in vitro.



FIG. 5C shows the mass spectrum for the negative control, 2604, binding to recombinant GPX4 in vitro.



FIG. 6A is a plot showing the mass spectrums for the ferroptosis-inducing compound, 1962, binding to recombinant GPX4 in vitro.



FIG. 6B is a plot showing the mass spectrums for the ferroptosis-inducing compound, 1816, binding to recombinant GPX4 in vitro.



FIG. 6C is a plot showing the mass spectrums for the ferroptosis-inducing compound, 3362, binding to recombinant GPX4 in vitro.



FIG. 6D is a plot showing the mass spectrums for the ferroptosis-inducing compound, 0973, binding to recombinant GPX4 in vitro.



FIG. 6E is a plot showing the mass spectrums for the ferroptosis-inducing compound, 8216, binding to recombinant GPX4 in vitro.



FIG. 7 is a plot showing relative GPX4 activity in vitro for the five ferroptosis-inducing compounds 1816, 8216, 3362, 1962, and 0973.



FIG. 8 is a plot showing the relative viability of HK-2 renal epithelial cells exposed to the ferroptosis-inducing compounds 8216 or 1962 over a range of concentrations.



FIG. 9 is a panel of plots showing relative viability of HK-2 kidney epithelial cells exposed to the inhibitor compounds 8216 or 1962 over a range of concentrations in the presence and absence of ferrostatin-1.



FIG. 10A is a plot showing transcriptional activity in IMR90 lung fibroblast cells exposed to RSL3.



FIG. 10B is a plot showing transcriptional activity in IMR90 lung fibroblast cells exposed to ML162.



FIG. 10C is a plot showing transcriptional activity in IMR90 lung fibroblast cells exposed to erastin.



FIG. 11 is a plot showing fold HMOX1 induction in HT-1080 fibrosarcoma cells exposed to compounds RSL3, 8216, 1962, 1816, or 3362.



FIG. 12 is a plot showing relative viability of HT-1080 fibrosarcoma cells exposed to different 8216 derivative compounds.



FIG. 13 is a plot showing relative viability of HT-1080 fibrosarcoma cells exposed to compounds with similar structures to compounds 6666 or 1816.



FIG. 14A, FIG. 14B and FIG. 14C illustrate a panel of plots 1400 showing relative viability of HT-1080 fibrosarcoma cells exposed to 15 inhibitor compounds with similar structures to 8216, 6666, or 1816 tested at 8 concentrations from 50 μM to 5 nM at half log dilutions in the presence of (red line) and absence of (black line) 1.5 UM ferrostatin-1. Ferrostatin-1 is a lipophilic antioxidant which inhibits the process of ferroptosis and rescues cells from undergoing this form of cell death. HT-1080 fibrosarcoma cells were plated on side-blackened, clear bottom plates 24 hours prior to treatment, with or without the addition of 2× ferrostatin-1. Each compound was diluted in DMSO and media and then added to the cells for a total of 24 hours, at which point cell viability was read out via Cell Titer Glo. Several compounds show a clear shift in EC50 with the addition of ferrostatin-1 (red line shifted to the right) which is evidence to suggest that those compounds induce cell death via ferroptosis.



FIG. 15 is a plot showing fold HMOX1 induction in HT-1080 fibrosarcoma cells exposed to compounds 8147, 6047, 3793, Rsl3, 6666 or 8216.



FIG. 16 is a panel of plots showing percent activity of GPX4 in response to exposure to compounds 8216, 6666, 8147, or 3793.



FIG. 17 shows a panel of representative images from fluorescent confocal microscopy of HT-1080 fibrosarcoma cells treated with compounds 8147, 8216, 3973, 6047, or 6666 in the presence or absence of 1.5 μM ferrostatin-1.



FIG. 18 is a panel of plots showing the intact protein mass spectrometry analyses of compounds 8216, 6666, and 8147.





DETAILED DESCRIPTION OF THE INVENTION
Terminology
Compound Terminology

“Isomer” means molecules with identical molecular formulas, that is the same number of atoms of each element, but distinct arrangements of atoms in space.


“Covalent inhibitor” means compounds that by design are intended to form a covalent bond with a specific molecular target.


Composition Terminology

“Pharmaceutically acceptable” means on balance, safe for use in humans or animals, without undue side effects.


“Pharmaceutically acceptable salt” means a salt of the compounds of the present invention which is pharmaceutically acceptable and which possess the desired pharmacological activity. Such salts include, for example, acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or with organic acids such as acetic acid, propionic acid, hexanoic acid, heptanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, o-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, p-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, p-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2.2.2]oct-2-ene-1-carboxylic acid, glucoheptonic acid, 4,4′-methylenebis(3-hydroxy-2-ene-1-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid and the like. Pharmaceutically acceptable salts also include base addition salts which may be formed when acidic protons present are capable of reacting with inorganic or organic bases. Acceptable inorganic bases include sodium hydroxide, sodium carbonate, potassium hydroxide, aluminum hydroxide and calcium hydroxide. Acceptable organic bases include ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine and the like.


Cell and Cell Death Terminology

“Ferroptosis” means regulated cell death that is iron-dependent. Ferroptosis is characterized by the iron-dependent accumulation of lethal lipid reactive oxygen species.


“GPX4” means the glutathione peroxidase 4, a glutathione metabolism enzyme.


“In vitro” means an artificial environment created outside a living multicellular organisms (e.g., a test tube or culture plate) used in experimental research to study a disease or process. As used herein, “in vitro” includes processes performed in intact cells growing in culture.


“In vivo” means that which takes place inside an organism and more specifically to a process performed in or on the living tissue of a whole, living multicellular organism (animal), such as a mammal, as opposed to a partial or dead one.


“Ex vivo” refers to a process performed in an artificial environment outside the organism on living cells or tissue which are removed from an organism and subsequently returned to an organism.


“Mesenchymal tumor” or “mesenchymal cancer” refers to tumors that arise from mesenchymal tissue or tumors that have undergone epithelial to mesenchymal transition. The epithelial-mesenchymal transition (EMT) is a process by which epithelial cells lose their cell polarity and cell-cell adhesion and gain migratory and invasive properties to become mesenchymal stem cells. EMT has also been shown to occur in the initiation of metastasis in cancer progression.


“Mesenchymal” refers to cells that develop into connective tissue, blood vessels, and lymphatic tissue.


Small Molecule Inducers of Ferroptosis

The invention provides small molecule inducers of ferroptosis. In various embodiments, the invention provides compounds that target the active site of the GPX4 enzyme, wherein binding of the compound to the active site of GPX4 effectively inhibits the activity of the enzyme.


In one embodiment, the invention provides a composition comprising a compound of the invention or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle.


In one embodiment, the invention provides a method for inducing ferroptosis in a cell, the method comprising contacting the cell with an effective amount of a compound of the invention or a pharmaceutically acceptable salt thereof.


In one embodiment, the invention provides a method for decreasing GPX4 activity in a cell, the method comprising contacting the cell with an effective amount of a compound of the invention or a pharmaceutically acceptable salt thereof.


The compounds of the invention are useful for inducing ferroptosis in a cell. In one embodiment, the compounds of the invention may be used in cancer therapy to induce ferroptosis in a cancer cell, such as a mesenchymal cancer cell.


Compounds

In various embodiments, the invention provides compounds that target the active site of the GPX4 enzyme, wherein binding of the compound to the active site of GPX4 effectively inhibits the activity of the enzyme.


In one embodiment, compounds of the invention, or pharmaceutically acceptable salts thereof, have the structure indicated by Formula 1:




embedded image




    • where n is 0 or 1; and

    • where B is selected from:







embedded image




    • where each ring individually may be aliphatic or aromatic;

    • where each ring individually may substituted or unsubstituted;

    • where when n is 0 and B is B1:
      • there are no ring substituents or ring heteroatoms; or there are ring heteroatoms comprising [A:1,5 and/or (B1:6 or B1:13 or B1:6,13) and/or (B2:8,9 or B2:10 or B2:11,12) is a N]; and/or B2:8 is a S; and/or (B2:8 or B2:10 or B2:13 is an O); and/or
      • if substituted, substituents on rings A and/or B1 and/or B2 are as follows:
        • one or more F, Br, Cl, CN, Me, OMe at any position on the ring;

    • where when n is 0 and B is B2:
      • there are no ring substituents or ring heteroatoms; or there are ring heteroatoms comprising (B1:6 or B1:6,13 or B1:6,14) and/or B2:8 is an N; and/or
      • if substituted, substituents on rings A and/or B1 and/or B2 are as follows:
        • one or more F, Br, Cl, CN, Me at any position on the ring;

    • where when n is 1 and B is B1:
      • there are no ring substituents or ring heteroatoms; or
      • there are ring heteroatoms comprising [(B1:6 or B1:13 or B1:6,13) and/or (B2:7,9 or B2:7,10 or B2:7,9,10 or B2:7,8,10 or B2:7,8,9,10 or B2:8 or B2:8,9 or B2:8,9,10 or B2:8,11 or B2:8,10,11 or B2:8,9,10,11 or B2:9,11 or B2:10 or B2:10,11) is an N]; and/or (B2:8 or B2:10 or B2:13 is an O); and/or B2:8 is an S; and/or
      • if substituted, substituents on rings A and/or B1 and/or B2 are as follows:
        • one or more F, Br, Cl, CN, Me at any position on the ring;

    • where when n is 1 and B is B2:
      • there are no ring substituents or ring heteroatoms; or
      • there are ring heteroatoms comprising [(B1:6 or B1:13 or B1:6,13 or B1:6,14) and/or (B2:8 or B2:9,11 or B2:11) is an N]; and/or
      • if substituted, substituents on rings A and/or B1 and/or B2 are as follows:
        • one or more F, Br, Cl, CN, Me at any position on the ring;

    • where when n is 1 and B is B3:
      • there are no ring substituents or ring heteroatoms; or
      • there are ring heteroatoms comprising (B1:6; or B1:6,8 or B1:6,10 or B1:7 or B1:8 is a N); and/or B1:8 is an O; and/or
      • if substituted, substituents on rings A and/or B1 include:
        • one or more F, Br, Cl, CN, Me, OMe, trihalomethane, OH at any position on the ring.





In one embodiment, compounds of the invention, or pharmaceutically acceptable salts thereof, have the structure of a compound provided in Table 1.












TABLE 1





Working


MS Data


Example
Structure
Name
(M + H)







A1


embedded image


2-chloro-N-(3,5-dimethylphenyl)-N- [(4-fluorophenyl)methyl]acetamide
306.1





A2


embedded image


2-chloro-N-(3,5-dimethylphenyl)-N- [(4-methoxyphenyl)methyl]acetamide
318.1





A3


embedded image


2-chloro-N-(3,5-dimethoxyphenyl)-N- [(4-fluorophenyl)methyl]acetamide
338.0





A4


embedded image


2-chloro-N-[(4-chlorophenyl)methyl]- N-(3,5-dimethylphenyl)acetamide
332.0





A5


embedded image


2-chloro-N-(3,5-dimethylphenyl)-N- [(3-methoxyphenyl)methyl]acetamide
318.0





A6


embedded image


2-chloro-N-[(2-chlorophenyl)methyl]- N-(3,5-dimethylphenyl)acetamide
322.0





A7


embedded image


2-chloro-N-(3,5-dimethylphenyl)-N- [(2-methoxyphenyl)methyl]acetamide
318.1





A8


embedded image


2-chloro-N-(3,5-dimethylphenyl)-N- (m-tolylmethyl)acetamide
302.1





A9


embedded image


2-chloro-N-[(3-chlorophenyl)methyl]- N-(3,5-dimethylphenyl)acetamide
332.0





A10


embedded image


2-chloro-N-(3,5-dimethylphenyl)-N-(p- tolylmethyl)acetamide
302.1





A11


embedded image


2-chloro-N-(3,5-dimethylphenyl)-N-(o- tolylmethyl)acetamide
302.1





A12


embedded image


2-chloro-N-cyclohexyl-N-[(4- fluorophenyl)methyl]acetamide
284.1





A13


embedded image


2-chloro-N-[(4-fluorophenyl)methyl]- N-(3-methoxyphenyl)acetamide
308.0





A14


embedded image


2-chloro-N-(3-chloro-5-methoxy- phenyl)-N-[(4- fluorophenyl)methyl]acetamide
342.0





A15


embedded image


2-chloro-N-(3,5-difluorophenyl)-N-[(4- fluorophenyl)methyl]acetamide
314.0





A16


embedded image


2-chloro-N-(3,5-dichlorophenyl)-N-[(4- fluorophenyl)methyl]acetamide
345.9





A17


embedded image


2-chloro-N-(3-chloro-4-methoxy- phenyl)-N-[(4- fluorophenyl)methyl]acetamide
342.0





A18


embedded image


2-chloro-N-(3-chlorophenyl)-N-[(4- fluorophenyl)methyl]acetamide
312.0





A19


embedded image


2-chloro-N-[(4-cyanophenyl)methyl]- N-(3,5-dimethoxyphenyl)acetamide
345.1





A20


embedded image


2-chloro-N-[(2,3- difluorophenyl)methyl]-N-(3,5- dimethoxyphenyl)acetamide
356.0





A21


embedded image


2-chloro-N-(3,5-dimethoxyphenyl)-N- [(4-methoxyphenyl)methyl]acetamide
350.1





A22


embedded image


2-chloro-N-[(4-fluorophenyl)methyl]- N-(6-quinolyl)acetamide
329.1





A23


embedded image


N-[(2-bromo-4-fluoro-phenyl)methyl]- 2-chloro-N-(3,5- dimethoxyphenyl)acetamide
416.0





A24


embedded image


2-chloro-N-(3,5-dimethoxyphenyl)-N- [(4-hydroxyphenyl)methyl]acetamide
336.1





A25


embedded image


2-chloro-N-[(4-fluorophenyl)methyl]- N-(3-methoxy-5-methyl- phenyl)acetamide
322.1





A26


embedded image


2-chloro-N-[(2,6-dichloro-4-fluoro- phenyl)methyl]-N-(3,5- dimethoxyphenyl)acetamide
406.0





A27


embedded image


2-chloro-N-(3,5-dimethoxyphenyl)-N- [[4-fluoro-2-(trifluoro- methyl)phenyl]methyl]acetamide
406.0





A28


embedded image


2-chloro-N-[(5-cyano-2-fluoro- phenyl)methyl]-N-(3,5- dimethoxyphenyl)acetamide
363.1





A29


embedded image


2-chloro-N-[(4-fluorophenyl)methyl]- N-(3-methoxy-4-methyl- phenyl)acetamide
322.1





A30


embedded image


2-chloro-N-(3,5-dimethoxyphenyl)-N- [(4-fluoro-2,6-dimethyl- phenyl)methyl]acetamide
366.1





A31


embedded image


2-chloro-N-(3,5-dimethoxyphenyl)-N- [[2-fluoro-3-(trifluoro- methyl)phenyl]methyl]acetamide
406.1





A32


embedded image


2-chloro-N-(2-fluoro-6-methoxy- phenyl)-N-[(4- fluorophenyl)methyl]acetamide
326.1





A33


embedded image


2-chloro-N-(3,5-dimethoxyphenyl)-N- [(4-fluoro-3-methoxy- phenyl)methyl]acetamide
368.1





A34


embedded image


2-chloro-N-(3-chloro-5-hydroxy- phenyl)-N-[(4- fluorophenyl)methyl]acetamide
328.1





A35


embedded image


2-chloro-N-(2-chloro-3-methoxy- phenyl)-N-[(4- fluorophenyl)methyl]acetamide
342.0





A36


embedded image


2-chloro-N-(2-cyano-4-methoxy- phenyl)-N-[(4- fluorophenyl)methyl]acetamide
333.1





A37


embedded image


2-chloro-N-(2-chloro-5-methoxy- phenyl)-N-[(4- fluorophenyl)methyl]acetamide
342.0





A38


embedded image


N-[(4-bromo-2,3-difluoro- phenyl)methyl]-2-chloro-N-(3,5- dimethoxyphenyl)acetamide
434.0





A39


embedded image


N-(3-bromo-1H-pyrazolo[3,4- b]pyridin-5-yl)-2-chloro-N-[(4- fluorophenyl)methyl]acetamide
397.0





A40


embedded image


2-chloro-N-[(4-fluorophenyl)methyl]- N-(4-methoxy-3,5-dimethyl- phenyl)acetamide
336.2





A41


embedded image


2-chloro-N-[(4-fluorophenyl)methyl]- N-(4-hydroxy-3,5-dimethyl- phenyl)acetamide
322.1





A42


embedded image


2-chloro-N-(2,4-dichloro-3-methyl- phenyl)-N-[(4- fluorophenyl)methyl]acetamide
360.0





A43


embedded image


2-chloro-N-[(2-chloro-3,4-difluoro- phenyl)methyl]-N-(3,5- dimethoxyphenyl)acetamide
390.1





A44


embedded image


2-chloro-N-(3,5-dimethoxyphenyl)-N- [(3-fluoro-5-methyl- phenyl) methyl]acetamide
352.1





A45


embedded image


N-(2,1,3-benzothiadiazol-4-ylmethyl)- 2-chloro-N-(3,5- dimethoxyphenyl)acetamide
378.1





A46


embedded image


2-chloro-N-[(4-fluorophenyl)methyl]- N-(2-methoxy-4-pyridyl)acetamide
309.1





A47


embedded image


2-chloro-N-[(4-fluorophenyl)methyl]- N-(6-methyl-3-pyridyl)acetamide
293.1









In one embodiment, compounds of the invention, or pharmaceutically acceptable salts thereof, have the structure indicated by Formula 2:




embedded image




    • where n is 0 or 1; and

    • where B is selected from:







embedded image




    • and where Y is selected from:







embedded image




    • where each ring individually may be aliphatic or aromatic;

    • where each ring each ring individually may substituted or unsubstituted;

    • where when n is 0 or 1; and

    • where when B is B1 or B2; and

    • where when Y is any one of Y1, Y2, Y3, Y4, Y5, Y6, Y7, Y8, Y9, Y10, Y11:
      • there are no additional ring substituents or ring heteroatoms; or
      • if substituted, substituents on A and/or B rings and/or ring Y1 are as follows:
        • one or more F, Br, Cl, CN, OMe, Me at any position on the ring;

    • where when n is 0 and Y is Y1:
      • there are no additional ring substituents or ring heteroatoms; or
      • B is a 2-propenyl substituent; or B is a hexyl substituent; or B is a hydrogen; and/or
      • if substituted, substituents on ring A1 are as follows:
        • one or more F, Br, Cl, CN, Me, OMe at any position on the ring;

    • where when n is 0, B is B1 and Y is Y1:
      • there are no additional ring substituents or ring heteroatoms; or
      • there are one or more isopropyl substituents on any position on the B ring; and/or
      • if substituted, substituents on rings A1 and/or B1 are as follows:
        • one or more F, Br, Cl, CN, Me, OMe at any position on the ring;

    • where when n is 0, B is B3 and Y is Y1:
      • there are no additional ring substituents or ring heteroatoms; or
      • if substituted, substituents on ring A1 are as follows:
        • one or more F, Br, Cl, CN, Me, OMe at any position on the ring.





In one embodiment, compounds of the invention, or pharmaceutically acceptable salts thereof, have the structure of a compound provided in Table 2.












TABLE 2





Working


MS Data


Example
Structure
Name
(M + H)







B1


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- 2-chloro-N-(3- methoxypropyl)acetamide
403.1





B2


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- N-(3-methoxypropyl)prop-2- ynamide
379.2





B3


embedded image


2-chloro-N-[4-(4- methoxyphenyl)thiazol-2- yl]-N-(3- methoxypropyl)acetamide
355.1





B4


embedded image


N-[4-(4- methoxyphenyl)thiazol-2- yl]-N-(3- methoxypropyl)prop-2- ynamide
331.2





B5


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- 2-chloro-N-(3,5- dimethylphenyl)acetamide
435.0





B6


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- 2-chloro-N-cyclohexyl- acetamide
413.0





B7


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- N-(3,5- dimethylphenyl)prop-2- enamide
413.0





B8


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- N-(3,5-dimethylphenyl)but- 2-ynamide
425.2





B9


embedded image


2-chloro-N-(3,5- dimethylphenyl)-N-[4-(4- methoxyphenyl)thiazol-2- yl]acetamide
387.2





B10


embedded image


2-chloro-N-(4- cyclohexylthiazol-2-yl)-N- (3,5- dimethylphenyl)acetamide
363.2





B11


embedded image


2-chloro-N-(3,5- dimethylphenyl)-N-(4- phenylthiazol-2- yl)acetamide
357.1





B12


embedded image


N-[4-(3- bromophenyl)thiazol-2-yl]- 2-chloro-N-(3,5- dimethylphenyl)acetamide
435.1





B13


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- N-(3,5- dimethylphenyl)prop-2- ynamide
411.0





B14


embedded image


N-benzyl-N-[4-(4- bromophenyl)thiazol-2-yl]- 2-chloro-acetamide
421.1





B15


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- 2-chloro-N-phenyl- acetamide
407.1





B16


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- 2-chloro-N-(m- tolyl)acetamide
421.1





B17


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- 2-chloro-N-(3- methoxyphenyl)acetamide
437.1





B18


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- 2-chloro-N-(3,5- dimethoxyphenyl)acetamide
467.1





B19


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- N-(3,5- dimethoxyphenyl)prop-2- ynamide
443.0





B20


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- 2-chloro-N-(3,5- dichlorophenyl)acetamide
476.7





B21


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- N-(3,5-dimethylphenyl)-2- iodo-acetamide
527.0





B22


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- N-(3,5-dichlorophenyl)prop- 2-ynamide
452.9





B23


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- N-(3,5-dimethylphenyl)-2- fluoro-prop-2-enamide
431.0





B24


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- 2-chloro-N-(3-chloro-5- methoxy-phenyl)acetamide
472.9





B25


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- 2-chloro-N-(3,5- difluorophenyl)acetamide
444.9





B26


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- N-(3-chloro-5-methoxy- phenyl)prop-2-ynamide
449.0





B27


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- N-(3,5-dimethylphenyl)-2- fluoro-acetamide
419.0





B28


embedded image


N-[4-(4- bromophenyl)thiazol-2-yl]- N-(3,5- dimethylphenyl)oxirane-2- carboxamide
429.0









In one embodiment, compounds of the invention have the structure indicated by Formula 3:




embedded image




    • where n is 0 or 1; and

    • where B is selected from:







embedded image




    • where each ring individually may be aliphatic or aromatic;

    • where each ring each ring individually may substituted or unsubstituted;

    • where when n is 0 and B is B1:
      • there are no ring substituents or ring heteroatoms; or
      • there are ring heteroatoms comprising [(B1:9 or B1:16 or B1:9,16) and/or (B2:11,12 or B2:13) is an N]; and/or (B2:11 or B2:13 is an O); and/or B2:11 is an S;
      • if substituted, substituents on rings A1 and/or A2 and/or B1 and/or B2 are as follows:
        • one or more F, Br, Cl, CN, Me at any position on the ring;

    • where when n is 1 and B is B1:
      • there are no ring substituents or ring heteroatoms; or
      • there are ring heteroatoms comprising [(A1:2,4 or A1:2,5 or A1:2,4,5 or A1:2,3,5 or A1:2,3,4,5 or A1:3,4 or A1:3,4,5 or A1:3,6 or A1:3,4,6 or A1:3,5,6 or A1:3,4,5,6 or A1:4,5 or A1:4,6 or A1:5 or A1:5,6) and/or (A2:1 or A2:1,8 or A2:8) is an N]; and/or A1:3 is an S; and/or (A1:3 or A1:5 is an O); and/or
      • if substituted, substituents on rings A1 and/or A2 and/or B1 and/or B2 are as follows:
        • one or more F, Br, Cl, CN, Me at any position on the ring;

    • where when n is 0 and B is B2:
      • there are no ring substituents or ring heteroatoms; or
      • there are ring heteroatoms comprising [(B1:9 or B1:17 or B1:9,16 or B1:9,17) and/or B2:11 is an N]; and/or
      • if substituted, substituents on rings A1 and/or A2 and/or B1 and/or B2 are as follows:
        • one or more F, Br, Cl, CN, Me at any position on the ring;

    • where when n is 1 and B is B2:
      • there are no ring substituents or ring heteroatoms; or
      • there are ring heteroatoms comprising [(A1:3 or A1:4 or A1:5 or A1:3,4 or A1:3,5 or A1:4,5 or A1:3,4,5) and/or (A2:2 or A2:6 or A2:8 or A2:6,8) and/or (B1:9 or B1:17 or B1:9,16 or B1:9,17) and/or (B2:11 or B2:12 or B2:14 or B2:12,14) is an N]; and/or A1:3 is an S; and/or
      • if substituted, substituents on rings A1 and/or A2 and/or B1 and/or B2 are as follows:
        • one or more F, Br, Cl, CN, Me at any position on the ring;

    • where when n is 1 and B is B3:
      • there are no ring substituents or ring heteroatoms; or
      • there are ring heteroatoms comprising A1:3,4,6 and/or A2:8 is an N; and/or
      • if substituted, substituents on rings A1 and/or A2 and/or B1 and/or B2 are as follows:
        • one or more F, Br, Cl, CN, Me at any position on the ring.





In one embodiment, compounds of the invention have the structure indicated by Formula 4:




embedded image




    • where n is 0 or 1; and

    • where B is selected from:







embedded image




    • where each ring individually may be aliphatic or aromatic;

    • where each ring individually may substituted or unsubstituted;

    • where when n is 1 and B is B1:
      • there are no ring substituents or ring heteroatoms; or
      • there are ring heteroatoms comprising [(A1:3 or A1:4 or A1:6) and/or (A2:1 or A2:1,8 or A2:1,9) is an N]; and/or
      • if substituted, substituents on rings A1 and/or A2 and/or B1 and/or B2 are as follows:
        • one or more F, Br, Cl, CN, Me at any position on the ring;

    • where when n is 1 and B is B2:
      • there are no ring substituents or ring heteroatoms; or
      • there are ring heteroatoms comprising [(A1:4 or A1:6 or A1:4,6) and/or A2:1,8 and/or (B2:11 or B2:12 or B2:14 or B2:12,14) is an N]; and/or (A1:3 is an S); and/or
      • if substituted, substituents on rings A1 and/or A2 and/or B1 and/or B2 are as follows:
        • one or more F, Br, Cl, CN, Me at any position on the ring.





In one embodiment, compounds of the invention have the structure indicated by Formula 5:




embedded image




    • where B is selected from:







embedded image




    • where each ring individually may be aliphatic or aromatic;

    • where each ring individually may substituted or unsubstituted;

    • and where Y is selected from:







embedded image




    • where when B is B2; and

    • where when Y is any of Y1, Y2, Y3, Y4, Y5, Y6, Y7, Y8, Y9, Y10:
      • there are no additional ring substituents or ring heteroatoms; or
      • if substituted, substituents on rings A and/or B and/or Y1 are as follows:
        • one or more F, Br, Cl, CN, Me, OMe at any position on the ring;

    • where when B is B1; and

    • where when Y is Y1:
      • there are no additional ring substituents or ring heteroatoms; or
      • there are ring heteroatoms comprising A:1,5 is a N; and/or
      • if substituted, substituents on rings A and/or B are as follows:
        • one or more F, Br, Cl, CN, Me, OMe at any position on the ring;

    • where when B is B3; and

    • where when Y is Y1:
      • there are no additional ring substituents or ring heteroatoms; or
      • there are ring heteroatoms comprising (B:6,7; and/or B:9 is a N); and/or B:7 is an O; and/or B:6 is an S; and/or
      • if substituted, substituents on rings A and/or B are as follows:
        • one or more F, Br, Cl, CN, Me, OMe at any position on the ring.





The compounds of the invention include the compounds of Formulas 1-5, and active derivatives and salts thereof.


In some embodiments, a compounds of the invention is a compound shown in Tables 1 and 2.


In some embodiments, a compounds of the invention is a compound shown in Table 3. The compounds of the invention include the compounds 1-643 and active derivatives and salts thereof.


Different versions of the compounds of the invention may be synthesized. For example, the base structure of Compound 90 (see Table 3) may be modified at one or more certain positions to include different “linker lengths” that may include different numbers of carbons at this position to result in Formula 91 of Table 3, where each n independently is 0, 1 or 2 and R is a 5, 6, 7 or 8 membered optionally substituted aromatic or non-aromatic carbon ring.


Synthesis of Compounds 1-643

The Compounds 1-643 are either commercially available or may be synthesized using standard synthetic techniques known to those of ordinary skill in the art.


Compositions

The invention provides a composition, the composition comprising a pharmaceutically acceptable carrier, adjuvant, or vehicle, and one or more compounds having any one of the structures of Compounds 1-643 or active derivatives or a pharmaceutically acceptable salt thereof.


A composition of the present invention may be administered in any desired and effective manner: for oral ingestion, or as an ointment or drop for local administration to the eyes, or for parenteral or other administration in any appropriate manner such as intraperitoneal, subcutaneous, topical, intradermal, inhalation, intrapulmonary, rectal, vaginal, sublingual, intramuscular, intravenous, intraarterial, intrathecal, or intralymphatic. Further, a composition of the present invention may be administered in conjunction with other treatments. A composition of the present invention may be encapsulated or otherwise protected against gastric or other secretions, if desired.


The compositions of the invention are pharmaceutically acceptable and may comprise one or more active ingredients in admixture with one or more pharmaceutically-acceptable carriers and, optionally, one or more other compounds, drugs, ingredients and/or materials. Regardless of the route of administration selected, the agents/compounds of the present invention are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art see, e.g., Remington, The Science and Practice of Pharmacy (21st Edition, Lippincott Williams and Wilkins, Philadelphia, Pa).


Pharmaceutically acceptable carriers are well known in the art (see, e.g., Remington, The Science and Practice of Pharmacy (21st Edition, Lippincott Williams and Wilkins, Philadelphia, Pa.) and The National Formulary (American Pharmaceutical Association, Washington, D.C.)) and include sugars {e.g., lactose, sucrose, mannitol, and sorbitol), starches, cellulose preparations, calcium phosphates (e.g., dicalcium phosphate, tricalcium phosphate and calcium hydrogen phosphate), sodium citrate, water, aqueous solutions (e.g., saline, sodium chloride injection, Ringer's injection, dextrose injection, dextrose and sodium chloride injection, lactated Ringer's injection), alcohols (e.g., ethyl alcohol, propyl alcohol, and benzyl alcohol), polyols (e.g., glycerol, propylene glycol, and polyethylene glycol), organic esters (e.g., ethyl oleate and triglycerides), biodegradable polymers (e.g., polylactide-polyglycolide, poly(orthoesters), and poly(anhydrides)), elastomeric matrices, liposomes, microspheres, oils (e.g., corn, germ, olive, castor, sesame, cottonseed, and groundnut), cocoa butter, waxes (e.g., suppository waxes), paraffins, silicones, talc, silicylate, etc. Each pharmaceutically acceptable carrier used in a composition of the invention must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the subject. Carriers suitable for a selected dosage form and intended route of administration are well known in the art, and acceptable carriers for a chosen dosage form and method of administration can be determined using ordinary skill in the art.


The compositions of the invention may, optionally, contain additional materials commonly used in such compositions. These ingredients and materials are well known in the art. Examples of pharmaceutically acceptable materials include:

    • fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and silicic acid;
    • binders, such as carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, hydroxypropylmethyl cellulose, sucrose, and acacia;
    • humectants, such as glycerol;
    • disintegrating agents, such as agar-agar, calcium carbonate, potato, or tapioca starch, alginic acid, certain silicates, sodium starch glycolate, cross-linked sodium carboxymethyl cellulose and sodium carbonate;
    • solution retarding agents, such as paraffin;
    • absorption accelerators, such as quaternary ammonium compounds;
    • wetting agents, such as cetyl alcohol and glycerol monostearate;
    • absorbents, such as kaolin and bentonite clay;
    • lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, and sodium lauryl sulfate;
    • suspending agents, such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth;
    • buffering agents;
    • excipients, such as lactose, milk sugars, polyethylene glycols, animal and vegetable fats, oils, waxes, paraffins, cocoa butter, starches, tragacanth, cellulose derivatives, polyethylene glycol, silicones, bentonites, silicic acid, talc, salicylate, zinc oxide, aluminum hydroxide, calcium silicates, and polyamide powder;
    • inert diluents, such as water or other solvents;
    • preservatives;
    • surface-active agents;
    • dispersing agents;
    • control-release or absorption-delaying agents, such as hydroxypropyl methyl cellulose, other polymer matrices, biodegradable polymers, liposomes, microspheres, aluminum monosterate, gelatin, and waxes;
    • opacifying agents;
    • adjuvants;
    • wetting agents;
    • emulsifying and suspending agents;
    • solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan;
    • propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane;
    • antioxidants;
    • agents which render the formulation isotonic with the blood of the intended recipient, such as sugars and sodium chloride;
    • thickening agents;
    • coating materials, such as lecithin; and
    • sweetening, flavoring, coloring, perfuming and preservative agents.


Ingredients and materials suitable for a selected dosage form and intended route of administration are well known in the art, and acceptable ingredients and materials for a chosen dosage form and method of administration may be determined using ordinary skill in the art.


Compositions suitable for oral administration may be in the form of capsules, cachets, pills, tablets, powders, granules, a solution, or a suspension in an aqueous or non-aqueous liquid, an oil-in-water or water-in-oil liquid emulsion, an elixir or syrup, a pastille, a bolus, an electuary or a paste. These formulations may be prepared by methods known in the art, e.g., by means of conventional pan-coating, mixing, granulation or lyophilization processes.


Solid dosage forms for oral administration (capsules, tablets, pills, dragées, powders, granules, and the like) may be prepared, e.g., by mixing the active ingredient(s) with one or more pharmaceutically-acceptable carriers and, optionally, one or more fillers, extenders, binders, humectants, disintegrating agents, solution retarding agents, absorption accelerators, wetting agents, absorbents, lubricants, and/or coloring agents. Solid compositions of a similar type may be employed as fillers in soft and hard-filled gelatin capsules using a suitable excipient. A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using a suitable binder, lubricant, inert diluent, preservative, disintegrant, surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine. The tablets, and other solid dosage forms, such as dragees, capsules, pills, and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein. They may be sterilized by, for example, filtration through a bacteria-retaining filter. These compositions may also optionally contain opacifying agents and may be of a composition such that they release the active ingredient only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. The active ingredient can also be in microencapsulated form.


Liquid dosage forms for oral administration include pharmaceutically-acceptable emulsions, microemulsions, solutions, suspensions, syrups, and elixirs. The liquid dosage forms may contain suitable inert diluents commonly used in the art. Besides inert diluents, the oral compositions may also include adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents. Suspensions may contain suspending agents.


Compositions for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more active ingredient(s) with one or more suitable nonirritating carriers which are solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound. Compositions which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams, or spray formulations containing such pharmaceutically-acceptable carriers as are known in the art to be appropriate.


Dosage forms for the topical or transdermal administration include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches, drops and inhalants. The active agent(s)/compound(s) may be mixed under sterile conditions with a suitable pharmaceutically-acceptable carrier. The ointments, pastes, creams and gels may contain excipients. Powders and sprays may contain excipients and propellants.


Compositions suitable for parenteral administrations comprise one or more agent(s)/compound(s) in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain suitable antioxidants, buffers, solutes which render the formulation isotonic with the blood of the intended recipient, or suspending or thickening agents. Proper fluidity can be maintained, for example, by the use of coating materials, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. These compositions may also contain suitable adjuvants, such as wetting agents, emulsifying agents, and dispersing agents. It may also be desirable to include isotonic agents. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption.


In some cases, in order to prolong the effect of a drug (e.g., pharmaceutical formulation), it is desirable to slow its absorption from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility.


The rate of absorption of the active agent/drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally-administered agent/drug may be accomplished by dissolving or suspending the active agent/drug in an oil vehicle. Injectable depot forms may be made by forming microencapsule matrices of the active ingredient in biodegradable polymers.


Depending on the ratio of the active ingredient to polymer, and the nature of the particular polymer employed, the rate of active ingredient release can be controlled. Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue. The injectable materials can be sterilized for example, by filtration through a bacterial-retaining filter.


The formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampules and vials, and may be stored in a lyophilized condition requiring only the addition of the sterile liquid carrier, for example water for injection, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the type described above.


Inducing Ferroptosis in Cells

The invention provides a method of inducing ferroptosis in a cell, the method comprising contacting the cell with an effective amount of one or more compounds according to the present invention.


In one aspect of this embodiment, the cell may be mammalian, preferably human. In other aspects of this embodiment, the cell may be from a laboratory animal. In addition to humans, categories of mammals within the scope of the present invention include, for example, agricultural animals, veterinary animals, laboratory animals, etc. Some examples of agricultural animals include cows, pigs, horses, goats, etc. Some examples of veterinary animals include dogs, cats, etc. Some examples of laboratory animals include rats, mice, rabbits, guinea pigs, etc.


In one aspect of this embodiment, the method is carried out in vitro. In other aspects of this embodiment, the method is carried out in vivo or ex vivo.


In one embodiment, the cell is a cancer cell, such as a mesenchymal cancer cell. Mesenchymal tumors (i.e., either sarcomas or tumors that have undergone epithelial to mesenchymal transition) are typically characterized by a relatively high content of polyunsaturated fatty acids and iron. Because of the relatively high polyunsaturated fatty acid and iron content, cellular lipids are subjected to relatively high levels of oxidation to produce lipid peroxides, which in the absence of GPX4 can be toxic to the cell.


Decreasing GPX4 Activity in Cells

The invention provides a method for decreasing GPX4 activity in a cell, the method comprising contacting the cell with an effective amount of one or more compounds of the present invention.


In one aspect of this embodiment, the cell may be mammalian, preferably human. In other aspects of this embodiment, the cell may be from a laboratory animal. In addition to humans, categories of mammals within the scope of the present invention include, for example, agricultural animals, veterinary animals, laboratory animals, etc. Some examples of agricultural animals include cows, pigs, horses, goats, etc. Some examples of veterinary animals include dogs, cats, etc. Some examples of laboratory animals include rats, mice, rabbits, guinea pigs, etc.


In one aspect of this embodiment, the method is carried out in vitro. In other aspects of this embodiment, the method is carried out in vivo or ex vivo.


Cancer Therapy

The compounds of the invention may be used in cancer therapy to induce ferroptosis in a cancer cell.


In one embodiment, the invention provides a method for treating a cancer in a subject in need thereof, the method comprising, administering to the subject a pharmaceutically effective amount of a pharmaceutical composition including one or more compounds, or pharmaceutically acceptable salts thereof, of the present invention.


In one aspect, the invention provides a method for treating a mesenchymal cancer, e.g., a sarcoma, in a subject in need thereof, the method comprising, administering to the subject a pharmaceutically effective amount of a pharmaceutical composition including one or more compounds, or pharmaceutically acceptable salts thereof, of the present invention.


In one aspect, the method reduces the growth rate of a tumor, reduces the size of a tumor, eliminates a tumor, or delays progression of a cancer stage.


EXAMPLES

The following examples are included for illustrative purposes only and are not intended to limit the scope of the inventive concepts herein.


Identification of Ferroptosis-Inducing Compounds

To identify potential covalent inhibitors of GPX4, we screened a panel of 294 diverse small molecules from the Enamine covalent inhibitor library (Catalog No. CSL-10480-0-Z-10, Enamine, Monmouth, NJ). Each compound contains either a chloroacetamide or acrylamide moiety theoretically capable of forming a covalent bond with a selenocysteine. The small molecule compounds were selected for a diversity of backbone structures. The screening protocol used the cell viability assay Cell Titer Glo® (available from Promega, Madison, WI). Cell Titer Glo® is a luminescent cell viability that determines the number of viable cells in culture by quantifying ATP, which indicates the presence of metabolically active cells. Briefly, HT-1080 fibrosarcoma cells were plated 24 hours prior to treatment with 10 UM of each compound in the screening panel. Cell Titer Glo® was used to read out cell viability after 48 hours of compound exposure. Data was collected in duplicate.



FIG. 1 is a plot 100 showing the relative viability of HT-1080 fibrosarcoma cells exposed to each compound in the inhibitor screening panel. We identified 36 out of the 294 (12%) small molecule compounds tested that resulted in greater than 50% reduction in cell viability at this dose. All but one of 36 small molecules identified in the screen contains a chloroacetamide moiety, which suggests that chloroacetamides are generally more toxic to HT-1080 fibrosarcoma cells than acrylamides.


To determine whether the reduction in cell viability by the compounds identified in FIG. 1 is driven by induction of ferroptosis, we performed dose response curves for each compound in the presence or absence of ferrostatin-1 or liproxstatin-1, which prevent ferroptosis by functioning as radical trapping antioxidants at cellular membranes. Briefly, HT-1080 fibrosarcoma cells were plated 24 hours prior to treatment with a range of concentrations of each inhibitor compound ranging from 100 μM to 100 nM in the presence or absence of 1 μM liproxstatin-1 or 1.5 UM ferrostatin-1. Cell Titer Glo® was used to read out cell viability after 48 hours compound exposure. Data was collected in triplicate.



FIG. 2A and FIG. 2 B are plots 200 showing the relative viability of HT-1080 fibrosarcoma cells exposed to inhibitor compounds in the presence and absence of 1 μM Liproxstatin-1. In this example, viability data are shown for HT-1080 cells exposed to the compounds 5001, 5758, 4480, 5814, 6988, 8684, 0418, 4458, and 3908 in the presence (red line) or absence (black line) of Liproxstatin-1. The data show that the majority of compounds tested displayed cell killing both in the presence and absence of 1 μM liproxstatin-1, which suggests a non-ferroptotic mode of action.



FIG. 3 is a panel of plots 300 showing the relative viability of HT-1080 fibrosarcoma cells exposed to inhibitor compounds in the presence (red line) and absence (black line) of 1.5 UM ferrostatin-1. In this example, viability data are shown for HT-1080 cells exposed to the compounds 8216, 1962, 0973, 3362, and 1816. The five compounds (8216, 1962, 0973, 3362, and 1816) resulted in cell death that was rescued by the addition of the antioxidant ferrostatin-1 to the media, as evidenced by the shift of the red line to the right. These results suggest that the cells are undergoing lipid peroxidation induced by these five inhibitor compounds (i.e., the five inhibitor compounds induce ferroptosis). The chemical structures of the five inhibitor compounds (8216, 1962, 0973, 3362, and 1816) are shown in Table 4.


To validate that the five ferroptosis-inducing compounds identified in FIG. 3 do cause lipid peroxidation, we used Bodipy-C11 staining in 786-O Renal Clear Cell Carcinoma cells to visualize using microscopy the presence (or absence) of oxidized lipids. Biodipy-C11 is a dye that traffics to lipid membranes and harbors a fluorophore that emits at different wavelengths depending on its oxidation status. Briefly, 786-O Renal Clear Cell Carcinoma cells were plated 24 hours prior to treatment with each of the five ferroptosis-inducing compounds (8216, 1816, 3362, 1962, and 0973) for one or two hours. DMSO was used as a control treatment. Following the treatment period, Hoechst stain (10 μM; a nuclear counterstain) and Bodipy-C11 (200 μM) were added to the cells for 30 minutes and then the cells were imaged using the Opera Phenix high content screening microscope.



FIG. 4A is a panel of plots 400 showing fluorescence images of Biodipy-C11 staining of cells treated with the five ferroptosis-inducing compounds identified in FIG. 3. In this example, images are shown for cells exposed to compound 8216 (6.25 μM) for 2 hours, compound 1816 (25 μM) for 1 hour, compound 3362 (12.5 μM) for 2 hours, compound 0973 (50 μM) for 2 hours, and compound 1962 (50 μM) for 2 hours. Cells treated with DMSO were used as a non-lethal control. Green: oxidized lipids; orange: reduced lipids; blue: nuclei (Hoechst nuclear counterstain). In the DMSO treated control cell cultures, all cells appear as orange, which indicates that all lipids are in the reduced form. In cell cultures treated with the ferroptosis-inducing compounds, some degree of lipid peroxidation was observed for all five compounds (as evidenced by the green color), but to varying degrees and at different dosages and time points. The data demonstrates that cellular lipids are becoming oxidized in response to exposure to these five compounds, which suggests the compounds induce lipid peroxidation and hence ferroptosis.



FIG. 4B is a panel of plots 410 showing fluorescence images of Biodipy-C11 staining of cells treated with compound 8216 and the controls RSL3 and sodium arsenite. Briefly, 786-O Renal Clear Cell Carcinoma cells were plated 24 hours prior to treatment with compound 8216 and the controls RSL3 and sodium arsenite for 1 hour with and without 1.5 μM of ferrostatin-1. Bodipy C-11 undergoes a shift in emission when lipids become oxidized, non-oxidized lipids are represented in orange and oxidized lipids are green; Hoescht was included as nuclear stain to represent cell bodies. A hallmark of ferroptosis is the oxidation of lipids, therefore with the shift in emission color from orange to green in these cells with 8216 treatment is suggestive of active ferroptosis.


Mechanism of Ferroptosis-Inducing Chloroacetamide Compounds

Mechanistically, ferroptosis can be induced by covalent inhibitors of GPX4 or by depletion of glutathione (GSH). Since GPX4 reduces lipid hydroperoxides using GSH as a co-substrate, both mechanisms ultimately result in loss of GPX4 activity, followed by elevated levels of reactive oxygen species which induces lipid peroxidation and subsequent cell death. In addition, changes in the levels of polyunsaturated fatty acids and/or iron in a cell in response to a potential covalent inhibitor of GPX4 activity may also induce or contribute to the ferroptotic phenotype.


To determine whether the five ferroptosis-inducing compounds identified in FIG. 3 and FIG. 4 could function through direct binding and inhibition of GPX4, we tested binding of each compound to recombinant GPX4 protein in vitro by intact protein mass spectrometry. Intact mass analysis is the assessment of a protein's total molecular weight by mass spectrometry without prior digestion or fragmentation of the molecule of interest. Covalent binding of a potential inhibitor to GPX4 can be detected by a shift in the mass of the intact protein. For the intact protein analysis, we used ML162, a well-characterized GPX4 inhibitor (available from Sigma Aldrich), as a positive control. As negative controls, we included two chloroacetamide-containing compounds (0568 and 2604) that did not kill HT-1080 fibrosarcoma cells in the initial 10 μM screen described with reference to FIG. 1. Briefly, compounds were incubated with recombinant GPX4 protein in vitro for 2 hours before quenching with trifluoroacetic acid (TFA) and analysis by mass spectroscopy.



FIG. 5A is a plot 500 showing the mass spectrum for positive control ML162 binding to recombinant GPX4 in vitro. FIG. 5A and FIG. 5B are plots 510 showing the mass spectrums for the negative controls 0528 and 2604, respectively, binding to recombinant GPX4 in vitro. Recombinant GPX4 has a mass of 21206 DA. Referring now to FIG. 5A, the data show that all of the GPX4 protein is bound by ML162 as evidenced by the shift in peak size from 21206 Da to 21648.1992 Da, which corresponds to the mass of 1 molecule of ML162 (441.95 Da) binding to GPX4. A secondary site in GPX4 was also bound by ML162 as evidenced by a second peak at 22136.4004, which corresponds to the mass of 2 molecules of ML162 binding to GPX4. Referring now to FIG. 5B, neither negative control compound (0568 or 2604) bound to GPX4 in vitro. These results confirm that intact protein mass spectrometry can be used to detect binding of chloroacetamide-containing small molecules to GPX4, and that GPX4 likely harbors multiple potential binding sites for such compounds in vitro.


We tested each of the five ferroptosis-inducing compounds identified in FIG. 3 and FIG. 4 for the ability to bind GPX4 in vitro. FIG. 6A-6E are plots 600 showing the mass spectrums for the five ferroptosis-inducing compounds 1816, 8216, 3362, 1962, and 0973 binding to recombinant GPX4 in vitro. Peaks corresponding to unbound GPX4 are outlined in blue boxes (Unbound GPX4) and peaks corresponding to bound GPX4 are outlined in red boxes (Bound GPX4). The data show that all fie compounds were capable of binding GPX4, but with varying degrees of specificity. For example, compounds 0973 and 1816 display a single mass shift, implying that they bind GPX4 at a single site, while compounds 8216 and 3362 bind either 1, 2, or 3 times, which may suggest that they are less selective binders than compounds 0973 or 1816 in vitro. Compound 1962 appears to be the least selective binder, however, as no singly-bound protein was detected. Instead, compound 1962 can bind GPX4 either 2, 3, or 4 times, suggesting poor specificity for the GPX4 active site for this molecule.


Inhibition of GPX4 Enzymatic Activity

To determine whether the five ferroptosis-inducing compounds (1816, 8216, 3362, 1962, and 0973) inhibit GPX4 enzymatic activity in vitro, we used a commercially available GPX4 inhibitor screening kit (Cayman Chemical cat #701880). This assay measures a compound's ability to prevent cumene hydroperoxide reduction by recombinant GPX4 protein. Briefly, the five ferroptosis-inducing compounds (25 μM) were incubated with recombinant GPX4 protein for one hour before being run in the Cayman Chemicals GPX4 inhibitor screening assay according to manufacturer's recommendations. The assay read out is a change in absorbance between NADPH and NADP. ML162 (25 μM) and RSL3 (25 μM) were used as positive control compounds. RSL3 (available from Apex Bio) is a GPX4 inhibitor that has been shown to require an adapter protein, 14-3-3, for efficient inhibitory activity in vitro.



FIG. 7 is a plot 700 showing relative GPX4 activity in vitro for the five ferroptosis-inducing compounds 1816, 3362, 0973, 1962, 8216 compared with the ML162 and RSL3 positive control compounds. As expected, ML162 resulted in a reduction in GPX4 enzymatic activity of about 94%, while RSL3 (without the 14-3-3 adapter protein) showed a reduction of about 55% in GPX4 enzymatic activity at the concentration tested. Two of the candidate ferroptosis-inducers, 8216 and 1962, exhibit potent GPX4 inhibitory capacity, while 0973, 3362, and 1816 do not.


Referring now to FIG. 6 and FIG. 7, the binding and GPX4 enzymatic activity results suggest that compounds 8216 and 1962 may be direct GPX4 inhibitors, with EC50s (half maximal effective concentration) of about 100 nM and 440 nM, respectively. Interestingly, compounds 0973, 3362, and 1816 do not inhibit GPX4 enzymatic activity in vitro, suggesting that they may induce ferroptosis via alternative mechanisms, or that they require the cellular context for GPX4 inhibition. Understanding the targets of these non-GPX4-targeting compounds may present new opportunities for induction of ferroptosis in cells.


Selectivity for Cancer Cells

To test whether the ferroptosis-inducing compounds 8216 or 1962 display selectivity for cancer cells we performed a dose response study in HK-2 kidney epithelial cells. HK-2 kidney epithelial cells are a non-cancerous cell line that is commonly used as a “healthy” control in ferroptosis experiments. Briefly, HK-2 kidney epithelial cells were plated 24 hours prior to treatment with concentrations of the ferroptosis-inducing compounds 8216 or 1962 ranging from 10 UM to 10 nM. The ferroptosis-inducing compound RSL3 was used as a positive control. As an additional control, we included a non-specific inducer of apoptosis, staurosporine. Cell Titer Glo® was used to read out cell viability after 48 hours of compound exposure. Data was collected in triplicate.



FIG. 8 is a plot 800 showing the relative viability of HK-2 renal epithelial cells exposed to the ferroptosis-inducing compounds 8216 or 1962 over a range of concentrations. The data show staurosporine killed HK-2 renal epithelial cells at all doses tested. The data also show that RSL3, which is used as a model compound for other inhibitors of GPX4 activity, also displays toxicity in HK-2 renal epithelial cells. In contrast, both 8216 and 1962 compounds were less toxic to the HK-2 renal epithelial cells than RSL3 or staurosporine.


To determine whether the reduction in viability of HK-2 renal epithelial cells exposed to compounds 8216 or 1962 is driven by induction of ferroptosis, we performed dose response curves for each compound in the presence or absence of ferrostatin-1. We also compared the EC50s for each compound between healthy cells (HK-2 renal epithelial cells) and cancer cells (HT-1080 fibrosarcoma cells) to determine a therapeutic window for healthy vs diseased cells. Briefly, HK-2 renal epithelial cells were plated 24 hours prior to treatment with concentrations of each inhibitor compound ranging from 10 UM to 10 nM in the presence or absence of 1.5 μM ferrostatin-1. The ferroptosis-inducing compound RSL3 was used as a positive control. Cell Titer Glo® was used to read out cell viability after 48 hours of compound exposure. Data was collected in triplicate.



FIG. 9 is a panel of plots 900 showing relative viability of HK-2 kidney epithelial cells exposed to the inhibitor compounds 8216 or 1962 over a range of concentrations in the presence and absence of ferrostatin-1. The black line is the relative HK-2 kidney epithelial cell viability in the absence of ferrostatin-1 and the red line is the relative HK-2 kidney epithelial cell viability in the presence of ferrostatin-1. HT-1080 fibrosarcoma cell EC50s are denoted by the vertical red dotted line for each compound. Taking the difference between the dotted red line for HT-1080 fibrosarcoma cells and the HK-2 kidney epithelial cells (black line) provides an estimate of a therapeutic index. The data show that the EC50 of the 8216 and 1962 compounds is significantly shifted in HK-2 renal epithelial cells relative to HT-1080 fibrosarcoma cells, suggesting that these compounds are selective for cancer cells. Furthermore, the therapeutic window for the 8216 compound appears to be comparable to that of RSL3, given the differences in EC50 between healthy (HK-2 kidney epithelial cells) and cancer cells (HT-1080 fibrosarcoma cells) for both compounds.


We next examined whether the toxicity observed in HK-2 kidney epithelial cells is due to on- or off-target effects of each compound by calculating the degree of rescue by ferrostatin-1 in these cells. Referring still to FIG. 9, RSL3 toxicity in HK-2 kidney epithelial cells was effectively rescued by addition of ferrostatin-1, suggesting that the toxicity imparted to HK-2 cells by RSL3 is likely due to partial induction of ferroptosis. Toxicity of compound 8216 is also modestly rescued by ferrostatin-1, whereas 1962 is not protected, suggesting that the toxicity imparted by these compounds is at least partly due to off-target effects.


Transcriptional Signature of GPX4 Inhibition

Experimental results described above suggest that five of our compounds induce ferroptosis, though likely through alternative mechanisms. This is expected as ferroptosis can be induced as a result of several mechanisms besides direct inhibition of GPX4. For example, inhibition of system Xc leads to the depletion of glutathione and subsequent inactivation of GPX4. Ferroptosis can also be induced by increasing the amount of polyunsaturated fatty acids or labile iron within a cell. Methods to distinguish these different modes (direct vs indirect) of ferroptosis-induction could be helpful in the development of ferroptosis-inducing small molecule compounds.


Expression of heme oxygenase 1 (HMOX1) mRNA may be used as a molecular response biomarker for induction of ferroptosis by direct inhibition of GPX4. To distinguish the transcriptional responses elicited by direct versus indirect inhibitors of GPX4, we profiled two direct GPX4 inhibitors, RSL3 and ML162, as well as one system Xc inhibitor, erastin, by Precision Run On followed by sequencing (PRO-seq) in IMR90 lung fibroblast cells. Briefly, IMR90 lung fibroblasts cells (a ferroptosis sensitive cell line) were treated with GPX inhibitors (RSL3 1 μM, ML162 1 μM) or system Xc inhibitor (erastin 10 UM) for one hour and samples were subjected to PRO-seq.



FIG. 10A, FIG. 10B, and FIG. 10C are plots 1000 showing transcriptional activity in IMR90 lung fibroblast cells exposed to RSL3, ML162, or erastin. Blue dots represent genes; HMOX1 is highlighted and shown in red in each graph. The Y-axis is log fold change; the X-axis is Base Mean. The data show that exposure of IMR90 lung fibroblast cells to the direct GPX4 inhibitors RSL3, or ML162 resulted in robust transcriptional activation of the HMOX1 gene at one hour. By contrast, no HMOX1 induction was observed in the erastin treated samples at the same time point, suggesting that rapid HMOX1 induction may be a biomarker of ferroptosis induced by direct GPX4 inhibition rather than through system Xc inhibition.


We tested several of our ferroptosis-inducing compounds for their ability to rapidly induce HMOX1 by RT-qPCR. Briefly, HT-1080 fibrosarcoma cells were treated with either 10 UM or 5 UM of compound RSL3, 8216, 1962, 1816, or 3362 for four hours. RNA was isolated and reverse transcribed and HMOX1 expression quantified by qPCR. Each sample was normalized to an ACTB housekeeping gene control and fold change was calculated relative to a DMSO control.



FIG. 11 is a plot 1100 showing fold HMOX1 induction in HT-1080 fibrosarcoma cells exposed to compounds RSL3, 8216, 1962, 1816, or 3362. The data show that, as expected, RSL3 induces HMOX1 dose-dependently at 10 μM (purple bars) and 5 μM (blue bars). Compound 8216 results in a remarkably similar dose-dependent HMOX1 induction, suggesting that RSL3 and 8216 may function to induce ferroptosis through similar mechanisms, namely GPX4 inhibition. Compound 1962, which binds and inhibits GPX4 in vitro, did not lead to dose responsive induction of HMOX1, suggesting a potentially non-linear relationship between target inhibition and induction of ferroptosis, potentially through interactions with off-target proteins. Compound 1816 did not induce HMOX1, consistent with the model that it does not directly inhibit GPX4, but likely induces ferroptosis through an alternate mechanism. Compound 3362 also results in induction of HMOX1, but our binding results suggest that 3362 does not bind GPX4 in vitro. Whether 3362-mediated HMOX1 activation is due to direct inhibition of GPX4 or by an alternative mechanism will be investigated in future experiments.


Evaluating Derivatives of Compounds 8216, 6666, and 1816

Experimental results described hereinabove suggest that compound 8216 is a potent and selective inhibitor of GPX4. To determine whether similar molecules could offer other starting points for a medicinal chemistry campaign, we screened 95 additional compounds that contain a similar base structure to that of 8216. Briefly, HT-1080 fibrosarcoma cells were treated with 10 UM of a compound for 48 hours and cell viability was measured by Cell Titer Glo®. Data was collected in duplicate. A list of the compounds tested, and a summary of the average relative cell viability data is shown in Table 5.



FIG. 12 is a plot 1200 showing relative viability of HT-1080 fibrosarcoma cells exposed to different 8216 derivative compounds. The data show that at 10 μM, 60 of the 74 compounds tested result in a greater than 50% reduction in HT-1080 fibrosarcoma cell viability. This result suggests that the 8216 is a good starting point for a medicinal chemistry campaign.


From the screen of 8216 derivative compounds, compound 6666 (see Table 5) was selected for further study.


An additional 34 compounds from the Enamine covalent inhibitor library with similar structures to compounds 6666 or 1816 were screened for inducing cell death. Briefly, HT-1080 fibrosarcoma cells were plated 24 hours prior to treatment on 96-well black-sided clear bottom plates. Each compound in the screening panel was tested at a 10 UM concentration for 24 hours prior to addition of Cell Titer Glo to evaluate relative cell viability.



FIG. 13 is a plot 1300 showing relative viability of HT-1080 fibrosarcoma cells exposed to compounds with similar structures to compounds 6666 or 1816. The data show that 41% of the compounds tested in this screen demonstrated a relative viability >50%, with a majority of the compounds representing structures more similar to compound 6666. The compounds identified in this figure will be further evaluated in subsequent figures.


Compounds from the Enamine covalent inhibitor library with similar structures to compounds 8216, 6666 or 1816 were further screened for inducing cell death via ferroptosis. Briefly, HT-1080 fibrosarcoma cells were plated on black-sided clear bottom plates 24 hours prior to treatment, with or without the addition of 2× ferrostatin-1, which inhibits the process of ferroptosis and rescues cells from undergoing this form of cell death. Compounds were diluted in DMSO and media then added to the cells for a total of 24 hours at which point cell viability was read out via Cell Titer Glo. Compounds were tested at 7 concentrations ranging from 50 μM to 5 nM at half log dilutions.



FIG. 14 is a panel of plots 1400 showing relative viability of HT-1080 fibrosarcoma cells exposed to 15 inhibitor compounds with similar structures to 8216, 6666, or 1816 tested over a range of concentrations from 50 μM to 5 nM at half log dilutions in the presence of (red line) and absence of (black line) 1.5 μM ferrostatin-1. Ferrostatin-1 is a lipophilic antioxidant which inhibits the process of ferroptosis and rescues cells from undergoing this form of cell death. The black line is the relative HT-1080 fibrosarcoma cell viability in the absence of ferrostatin-1 and the red line is the relative HT-1080 fibrosarcoma cell viability in the presence of 1.5 μM ferrostatin-1. HT-1080 fibrosarcoma cells were plated on side-blackened, clear bottom plates 24 hours prior to treatment, with or without the addition of 2× ferrostatin-1. Each compound was diluted in DMSO and media and then added to the cells for a total of 24 hours, at which point cell viability was read out via Cell Titer Glo. The data show that several compounds cause a clear shift in EC50 with the addition of ferrostatin-1 (red line shifted to the right) which is evidence to suggest that those compounds induce cell death via ferroptosis.


We tested several of the ferroptosis-inducing compounds for their ability to rapidly induce expression of HMOX1. Briefly, HT-1080 fibrosarcoma cells were plated in 6-well dishes 24 hours prior to treatment with 5 μM of 8147, 6047, 3793, Rsl3, 6666 or 8216 with and without the addition of ferrostatin-1 to the media. Cells were harvested from the plate after 4 hours of compound treatment by adding Trizol reagent directly to the plate to lyse the cells and preserve the RNA. RNA was isolated using the Direct-zol 96-well RNA kit (Zymo cat #R2054). Subsequently purity and quantity of RNA in the samples were measured using a Nanodrop (Thermo Fisher cat #ND-ONE-W). RNA samples were normalized to 2 μg and reverse transcribed to cDNA using the Multiscribe High-Capacity Reverse Transcription kit (Thermo Fisher cat #4368814) following manufacturer's instructions. Quantitative real time PCR was carried out in triplicate using SYBR Select Master Mix (Thermo Fisher cat #4472903) following manufacturer's instructions with primers designed against HMOX1 and ActB from IDT. HMOX1 expression levels were normalized to ActB for each sample then fold change was determined against the vehicle control, DMSO.



FIG. 15 is a plot 1500 showing fold HMOX1 induction in HT-1080 fibrosarcoma cells exposed to compounds 8147, 6047, 3793, Rsl3, 6666 or 8216. The data show that all of the compounds tested demonstrated an upregulation of HMOX1 which was rescued by the addition of ferrostatin-1, to varying degrees, suggesting that the compounds induce ferroptosis.


An enzymatic assay that measures the reduction of glutathione by GPX4 was used to test the ability of compounds 8216, 6666, 8147, and 3793 to inhibit the activity of GPX4. The enzymatic assay was performed using a commercially available kit (Cayman cat #701880). The compounds were tested at 8 concentrations in duplicate with the compound being diluted following manufacturer's instructions. The enzymatic reaction was allowed to continue for 1 hour prior to reading out the kinetic shift in absorbance that occurs as result of GPX4 reducing glutathione.



FIG. 16 is a panel of plots 1600 showing percent activity of GPX4 in response to exposure to compounds 8216, 6666, 8147, or 3793. The data show that compounds 8147 and 3793 display a concentration-dependent inhibition of GPX4 activity, suggesting that these compounds could induce ferroptosis by directly inhibiting GPX4 in vivo.


Compounds 6666, 6047, 3973, 8216, and 8147 were also evaluated for inducing lipid peroxidation in HT-1080 fibrosarcoma cells in the presence or absence of 1.5 UM ferrostatin-1. Briefly, HT-1080 fibrosarcoma cells were treated with 10 UM of compound with or without 1.5 μM ferrostatin-1 and incubated at 37° C. for 1 hour. Bodipy C-11 and Hoescht stain were then added to the media, the cells were allowed to incubate for an additional 30 minutes and then were imaged on the Opera Phenix confocal screening system.



FIG. 17 shows a panel of representative images 1700 from fluorescent confocal microscopy of HT-1080 fibrosarcoma cells treated with compounds 6666, 6047, 3973, 8216, or 8147 in the presence or absence of 1.5 UM ferrostatin-1. The data show that all compounds tested display a shift in emission reflective of lipid peroxidation, with ferrostatin-1 rescuing the cells from undergoing lipid peroxidation (indicated by orange i.e., no shift in emission). Blue is the nuclear stain Hoescht to show cell bodies.


To determine whether the ferroptosis-inducing compounds 6666 and 8147 could function through direct binding and inhibition of GPX4, we tested binding of each compound to recombinant GPX4 protein in vitro by intact protein mass spectrometry. Compound 8216 was used as a positive control (see FIG. 6). Briefly, 15 μL of GPX4 protein (Cayman Chemical cat #SO960203]) was incubated with 100 UM of each compound (4.5 L) and diluted with 25.5 μl of PBS. The incubation was performed at room temperature for 2 hours and then the reaction was quenched with 3 μL of trifluoroacetic acid. Samples were subjected to UPLC coupled to Synapt G2 mass spectrometer to evaluate if binding of the test compound to GPX4 occurs. Binding is evident by a mass shift equivalent to the mass size of the compound minus a chlorine atom plus the mass size of the GPX4 protein, as described by the manufacturer.



FIG. 18 is a panel of plots 1800 showing the intact protein mass spectrometry analyses of compounds 8216, 6666, and 8147. The data show that compounds 8216 and 8147 demonstrate a mass shift. However, compound 6666 did not show a mass shift which suggests that compound 6666 does not bind to GPX4 under these conditions, while compounds 8216 and 8147 do.


Computer-Aided Design of Compound 8216 Derivatives

As an orthogonal approach, we used the Autogrow4 algorithm to predict molecules that could bind to the GPX4 active site. Autogrow4 is a genetic algorithm that takes in a protein structure and through a series of generations (max of 30 generations) builds molecules using commonly used small molecule fragments. Throughout each generation small fragments that bind with high predicted affinity to the target will have a chance to move forward to the next level in the algorithm and interact with other small fragments to build a larger compound. At the end of each generation filters are applied to molecules so that toxic and mutant compounds are eliminated and don't proceed to the next level. In addition, Lipinski's rule was applied in order to keep the size of the molecules below 500 Da.


We ran the algorithm independently 15 times on the active site of GPX4 and recovered several compounds predicted to interact with the site with estimated binding energies ranging from −8 to −6 kcal/mol. In one of the runs, 68 compounds contained the 8216 structure as part of the molecule. We applied a clustering analysis to these compounds to divide them into similar groups. Once the clustering was done, the centroid of the clusters was selected (n=9) and among those, 4 compounds were synthesizable. These independently-derived molecules are currently being synthesized. The chemical structures of the nine compounds identified are shown in Table 4.


SYNTHESIS EXAMPLES
Synthesis reaction scheme of N-[4-(4-bromophenyl)thiazol-2-yl]-2-chloro-N-(3,5-dimethylphenyl)acetamide (Compound 90)



embedded image


Detailed Synthesis of N-[4-(4-bromophenyl)thiazol-2-yl]-2-chloro-N-(3,5-dimethylphenyl)acetamide (Compound 90)
Step (a): Synthesis of N-[(3,5-dimethylphenyl)carbamothioyl]benzamide



embedded image


22.2 g (136 mmol) of 3,5-dimethylaniline (1) was added in one portion to a solution of benzoyl isothiocyanate (2) at 20° C., prepared by dissolving 15 g (123 mmol) in 60 mL acetone. The mixture was heated to 60° C. and stirred for 1 h. LC-MS showed 3,5-dimethylaniline (1) was consumed completely and one main peak with desired MS (285.1, (M+H)+) was detected. The reaction mixture was then cooled to room temperature and poured into crushed ice, whereupon a white precipitate formed. The precipitate was collected by filtration, washed with 100 ml water and dried over air to give 28 g of crude white solid intermediate product N-[(3,5-dimethylphenyl)carbamothioyl]benzamide (3).


MS (ESI): calculated 285.1 [(M+H)+]; measured 285.1 [(M+H)+].


Step (b): Synthesis of 3,5-dimethylphenyl)thiourea



embedded image


28 g of N-[(3,5-dimethylphenyl)carbamothioyl]benzamide (3) was added in portions to a solution of NaOH in water, prepared by dissolving 5 g NaOH (140 mmol) in 200 ml of water. The mixture was heated at 80° C. and stirred for 2 hr. LC-MS showed N-[(3,5-dimethylphenyl)carbamothioyl]benzamide (3) was consumed completely and one main peak with desired MS (181.1, [M+H]+) was detected. The mixture was then cooled to room temperature and added dropwise to 200 ml of a 1N aqueous hydrochloric acid solution. The resulting mixture was diluted with 100 ml of water and extracted with EtOAc (300 mL×3). The combined organic layers were washed with saturated brine solution (300 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was triturated in 50 mL of methyl tert-butyl ether to yield 15 g (83.2 mmol) of the white solid intermediate product (3,5-dimethylphenyl)thiourea (4).


MS (ESI): calculated 181.1 [(M+H)+]; measured 181.1 [(M+H)+].



1H NMR (400 MHZ, DMSO-d6) δ 9.55 (br s, 1H), 7.94-7.08 (m, 2H), 6.96 (br s, 2H), 6.76 (br s, 1H), 2.24 (s, 6H).


Step (c): Synthesis of 4-(4-bromophenyl)-N-(3,5-dimethylphenyl)thiazol-2-amine



embedded image


15.4 g (55.5 mmol) of 2-bromo-1-(4-bromophenyl)ethanone (5) and 27.5 mL (20.0 g, 197 mmol) triethylamine was added to a 20° C. solution of (3,5-dimethylphenyl)thiourea (4), prepared by dissolving 10 g (55.5 mmol) in 100 mL ethanol. The mixture was heated to 80° C. and stirred for 2 h. LC-MS showed (3,5-dimethylphenyl)thiourea (4) was consumed completely and one main peak with desired MS (358.9 [M (79Br)+H]+, 360.9 [M (81Br)+H]+) was detected. The reaction mixture was then cooled to room temperature, quenched with 100 ml water at 20° C. and extracted with 3 aliquots of 100 mL EtOAc. The combined organic layers were washed with 3 aliquots of 100 mL saturated brine, dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel flash chromatography (300 g SepaFlash® silica column, ethyl acetate/petroleum, gradient 0%-30% ethyl acetate/petroleum ether @ 200 mL/min) to yield 19 g (52.9 mmol) of the yellow solid intermediate product 4-(4-bromophenyl)-N-(3,5-dimethylphenyl)thiazol-2-amine (6).


MS (ESI): calculated 359.0 [M (79Br)+H]+, 361.0 [M (81Br)+H]+; measured 358.9 [M (79Br)+H]+, 360.9 [M (81Br)+H]+.



1H NMR (400 MHZ, DMSO-d6) δ 10.14 (s, 1H), 7.86 (d, J=8.4 Hz, 2H), 7.62 (d, J=8.4 Hz, 2H), 7.38 (s, 1H), 7.30 (s, 1H), 7.31 (s, 1H), 6.62 (s, 1H), 2.27 (s, 6H).


Step (d): Synthesis of N-[4-(4-bromophenyl)thiazol-2-yl]-2-chloro-N-(3,5-dimethylphenyl)acetamide



embedded image


84.5 mg (0.835 mmol) triethylamine was added to a solution of 4-(4-bromophenyl)-N-(3,5-dimethylphenyl)thiazol-2-amine (6), which was prepared by dissolving 100 mg (0.278 mmol) in 2 mL dichloromethane. The mixture was cooled to 0° C. and 62.9 mg 2-chloroacetyl chloride (0.557 mmol) (7) was added dropwise. The mixture was then heated to 20° C. and stirred for 4 h. LC-MS showed 4-(4-bromophenyl)-N-(3,5-dimethylphenyl)thiazol-2-amine (6) was consumed completely and desired MS (435.0 [M (79Br)+H]+, 437.0 [M (81Br)+H]+) was detected. The reaction was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: Waters Xbridge, 5 μm, 150×25 mm; mobile phase A: water containing 0.1% trifluoroacetic acid, mobile phase B: acetonitrile; gradient elution of 60%-90% B:A over 9 min) and lyophilized to give 21 mg (0.048 mmol) of the white solid final product N-[4-(4-bromophenyl) thiazol-2-yl]-2-chloro-N-(3,5-dimethylphenyl)acetamide.


MS (ESI): calculated 435.0 [M (79Br)+H]+, 437.0 [M (81Br)+H]+; measured 435.0 [M (79Br)+H]+, 437.0 [M (81Br)+H]+.



1H NMR (400 MHZ, DMSO-d6) δ 7.85 (s, 1H), 7.61-7.56 (m, 2H), 7.56-7.52 (m, 2H), 7.20 (s, 1H), 7.17 (s, 2H), 4.29 (s, 2H), 2.35 (s, 6H).


Synthesis Reaction Scheme of Compound 637



embedded image


Detailed Synthesis of Compound 637

0.06 g (0.67 mmol, 1.2 eq) of compound (8) was dissolved in DCM (10 mL) under an argon atmosphere. The solution was cooled to 0° C. and the following were added sequentially 0.2 g (0.56 mmol, 1 eq) of compound (6) and 0.172 g (0.83 mmol, 1.5 eq) DCC. The reaction mixture was warmed to room temperature and stirred overnight. The resulting mixture was diluted with DCM, washed with water and brine, dried over anhydrous Na2SO4, and evaporated under reduced pressure. The residue was purified by HPLC to obtain 0.061 g (0.141 mmol, 25.5% yield) of compound 637.


Synthesis Reaction Scheme of Compound 635



embedded image


Detailed Synthesis of Compound 635

0.056 g (0.67 mmol, 1.2 eq) compound (9) was dissolved in DCM (10 mL) under an argon atmosphere. The solution was cooled to 0° C. and the following were added sequentially 0.2 g (0.56 mmol, 1 eq) compound (6) and DCC (0.172 g, 0.83 mmol, 1.5 eq). The reaction mixture was warmed to room temperature and stirred overnight. The resulting mixture was diluted with DCM, washed with water and brine, dried over anhydrous Na2SO4, and evaporated under reduced pressure. The residue was purified by HPLC to obtain compound 635 (0.012 g, 0.0282 mmol, 5% yield).


Synthesis Reaction Scheme of Compound 636



embedded image


Detailed Synthesis of Compound 636

0.2 g (0.56 mmol, 1 eq) of compound (6) was dissolved in DCM (10 mL) under an argon atmosphere. The solution was cooled to 0° C. and the following were added sequentially 0.36 g (2.78 mmol, 5 eq) DIPEA and 0.095 g (0.72 mmol, 1.3 eq) compound (10) dropwise. The reaction mixture was warmed to room temperature and stirred overnight. The resulting mixture was diluted with DCM, washed with water and brine, dried over anhydrous Na2SO4, and evaporated under reduced pressure. The residue was purified by HPLC to obtain compound 636 (0.094 g, 0.2 mmol, 37.2% yield).


Synthesis Reaction Scheme of Compound 634



embedded image


Detailed Synthesis of Compound 634

0.2 g (0.56 mmol, 1 eq) compound (6) was dissolved in DCM (10 mL) under an argon atmosphere. The solution was cooled to 0° C. and the following were added sequentially 0.36 g (2.78 mmol, 5 eq) DIPEA and 0.092 g (0.72 mmol, 1.3 eq) compound (11) dropwise. The reaction mixture was warmed to room temperature and stirred overnight. The resulting mixture was diluted with DCM, washed with water and brine, dried over anhydrous Na2SO4, and evaporated under reduced pressure. The residue was purified by HPLC to obtain compound 634 (0.099 g, 0.22 mmol, 39.5% yield).


Synthesis Reaction Scheme of Compound 641



embedded image


Detailed Synthesis of Compound 641

0.20 g (0.56 mmol, 1 eq) compound (6) was dissolved in DCM (10 mL) under an argon atmosphere. To the solution were added sequentially compound (12) 3-(trimethylsilyl)propiolic acid (0.095 g, 0.670 mmol, 1.2 eq) and DCC (0.172 g, 0.830 mmol, 1.5 eq) in a basic solution. The reaction mixture was stirred overnight at room temperature. The resulting mixture was diluted with 20 mL DCM. The DCU byproduct was filtered through a pad of celite and washed with minimal amount of DCM. The combined DCM extracts were washed with water (3×20 mL), dried over anhydrous Na2SO4, and evaporated under reduced pressure to obtain desired intermediate compound (13) (0.20 g, 0.41 mmol, 74.3% yield), which was used in the next step without further purification.


0.20 g (0.41 mmol, 1 eq) compound (13) was dissolved in THF (10 mL) under an argon atmosphere. The solution was cooled to 0° C. and TBAF (0.46 mL, 1 M in THF, 1.1 eq) was added. The reaction mixture was warmed to room temperature and stirred overnight. The resulting mixture was diluted with DCM (30 mL), washed with water (3×20 mL). The organic layer was dried over anhydrous Na2SO4 and evaporated under reduced pressure. The residue was purified by HPLC to obtain compound 634 (0.0103 g, 0.025 mmol, 6% yield).


Synthesis Reaction Scheme of Compound 639



embedded image


Detailed Synthesis of Compound 639

0.50 g (1.39 mmol, 1 eq) compound (6) was dissolved in DCM (20 mL) under an argon atmosphere. To the solution were added sequentially compound (14) 3-chlorocyclobutane-1-carboxylic acid (0.225 g, 1.670 mmol, 1.2 eq) and DCC (0.431 g, 2.09 mmol, 1.5 eq) in a basic solution. The reaction mixture was stirred overnight at room temperature. The resulting mixture was diluted with DCM (30 mL). The DCU byproduct was filtered through a pad of celite and washed with minimal amount of DCM. The combined DCM extracts was washed with water (3×30 mL), dried over anhydrous Na2SO4, and evaporated under reduced pressure to obtain desired intermediate compound (15) (0.40 g, 0.84 mmol, 60.4% yield), which was used in the next step without further purification.


0.4 g (0.84 mmol, 1 eq) compound (15) was dissolved in THF (15 mL) under an argon atmosphere. The solution was cooled to −40° C. and the following were added dropwise LiHMDS (0.92 mL, 1.1 M in THF, 1.1 eq) and DMAP. The reaction mixture was slowly warmed to 0° C. and stirred for 30 min. The resulting mixture was quenched with a saturated solution of NH4Cl and extracted with EtOAc (3×30 mL). The combined organic layer was dried over anhydrous Na2SO4 and evaporated under reduced pressure. The residue was purified by HPLC to obtain compound 639 (0.0092 g, 0.02 mmol, 2.44% yield).


Synthesis Reaction Scheme of Compound 640



embedded image


Detailed Synthesis of Compound 640

1 g (7.87 mmol, 1 eq) compound (16) was dissolved in 20 mL concentrated H2SO4 under an argon atmosphere. 1.19 g (11.8 mmol, 1.5 eq) KNO3 was added to the solution. The reaction mixture was stirred overnight at 50° C. The resulting mixture was poured in ice water and extracted with DCM (3×50 mL). The combined organic layer was dried over anhydrous Na2SO4 and evaporated under reduced pressure to obtain compound (17) (1.1 g, 6.3 mmol, 81.2% yield).


0.20 g (0.56 mmol, 1 eq) compound (6) was dissolved in DCM (10 mL) under an argon atmosphere. To the solution were added sequentially 0.115 g (0.670 mmol, 1.2 eq) compound (17) and 0.172 g (0.830 mmol, 1.5 eq) DCC in a basic solution. The reaction mixture was stirred overnight at room temperature. The resulting mixture was diluted with DCM (20 mL). The DCU byproduct was filtered through a pad of celite and washed with minimal amount of DCM. The combined DCM extracts was washed with water (3×20 mL), dried over anhydrous Na2SO4, and evaporated under reduced pressure. The residue was purified by HPLC to obtain compound 640 (0.0473 g, 0.092 mmol, 16.5% yield).


Compounds B1-B28 were prepared by an analogous synthetic route to compounds 90, 634, 635, 636, 637, 639, 640, and 641 as illustrated above.


Synthesis Reaction Scheme of Compound 593



embedded image


Detailed Synthesis of Compound 593

1 g, 5.3 mmol, 1 eq) compound (18) was dissolved in 50 mL dioxane under an argon atmosphere. To the solution were added sequentially compound (19) (0.706 g, 5.3 mmol, 1 eq), tris(dibenzylideneacetone)dipalladium(0) (0.485 g, 0.53, mol, 0.1 eq), [5-(diphenylphosphanyl)-9,9-dimethyl-9H-xanthen-4-yl]diphenylphosphane (0.46 g, 0.80 mmol, 0.15 eq), and Cs2CO3 (3.4 g, 10.4 mol, 3 eq). The reaction mixture was stirred overnight at 100° C. The resulting mixture was diluted with EtOAc (200 mL), washed with water and brine, dried over anhydrous Na2SO4, and evaporated under reduced pressure. The residue was purified by column chromatography to obtain compound (20) (0.8 g, 2.8 mmol, 52.8% yield).


0.1 g (0.35 mmol, 1 eq) compound (20) was dissolved in DCM (10 mL) under an argon atmosphere. The solution was cooled to 0° C. and the following were added sequentially and dropwise: DIPEA (0.045 g, 0.35 mmol, 1 eq) and 2-chloroacetyl chloride (0.04 g, 0.35 mmol, 1 eq). The reaction mixture was warmed to room temperature and stirred overnight. The resulting mixture was diluted with DCM, washed with water and brine, dried over anhydrous Na2SO4, and evaporated under reduced pressure. The residue was purified by HPLC to obtain compound 593 (0.008 g, 0.0221 mmol, 6.3% yield).


Synthesis Reaction Scheme of Compound 587



embedded image


Detailed Synthesis of Compound 587

1 g (5.6 mmol, 1 eq) compound (21) was dissolved in dioxane (50 mL) under an argon atmosphere. To the solution were added sequentially compound (22) (0.723 g, 5.6 mmol, 1 eq), tris(dibenzylideneacetone)dipalladium(0) (0.513 g, 0.56, mol, 0.1 eq), [5-(diphenylphosphanyl)-9,9-dimethyl-9H-xanthen-4-yl]diphenylphosphane (0.486 g, 0.84 mmol, 0.15 eq), and Cs2CO3 (4 g, 12.2 mol, 3 eq). The reaction mixture was stirred overnight at 100° C. The resulting mixture was diluted with EtOAc (200 mL), washed with water and brine, dried over anhydrous Na2SO4, and evaporated under reduced pressure. The residue was purified by column chromatography to obtain compound (23) (0.12 g, 0.442 mmol, 7.9% yield).


0.12 g (0.44 mmol, 1 eq) Compound (23) was dissolved in DCM (10 mL) under an argon atmosphere. The solution was cooled to 0° C. and the following were added sequentially DIPEA (0.057 g, 0.44 mmol, 1 eq) and 2-chloroacetyl chloride (0.05 g, 0.44 mmol, 1 eq) dropwise. The reaction mixture was warmed to room temperature and stirred overnight. The resulting mixture was diluted with DCM, washed with water and brine, dried over anhydrous Na2SO4, and evaporated under reduced pressure. The residue was purified by HPLC to obtain compound 587 (0.0114 g, 0.032 mmol, 7.4% yield).


Compounds A1, A2, and A4-A47 were prepared by an analogous synthetic route to compound A3 as illustrated below.


Example A3: 2-Chloro-N-(3,5-dimethoxyphenyl)-N-(4-fluorobenzyl)acetamide



embedded image


Step 1: N-(4-Fluorobenzyl)-3,5-dimethoxyaniline

To a solution of 4-fluorobenzaldehyde (2 g, 16.1 mmol, 1.6 mL) in DCM (30 mL) was added 3,5-dimethoxyaniline (2.2 g, 14.5 mmol), acetic acid (968 mg, 16.1 mmol, 0.922 mL) and NaBH(OAc)3 (4.6 g, 21.9 mmol). The mixture was stirred at 25° C. for 12 h. The reaction mixture was partitioned between aqueous 1M NaOH (20 mL) and DCM (20 mL). The aqueous phase was extracted with DCM (20 mL×3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue which was purified by flash silica gel chromatography (20 g silica) eluting with a gradient of 0-10% EtOAc/petroleum ether at a flow rate of 120 mL/min. N-(4-Fluorobenzyl)-3,5-dimethoxyaniline (3.1 g, 75% yield) was obtained as a yellow oil. M+H+=262.2 (LCMS); 1H NMR (400 MHZ, DMSO-d6) δ 7.36 (dd, J=5.8, 8.1 Hz, 2H), 7.13 (t, J=8.8 Hz, 2H), 6.26 (br t, J=5.9 Hz, 1H), 5.78-5.68 (m, 3H), 4.20 (br d, J=5.9 Hz, 2H), 3.61 (s, 6H).


Step 2: 2-Chloro-N-(3,5-dimethoxyphenyl)-N-(4-fluorobenzyl)acetamide

To a solution of N-(4-fluorobenzyl)-3,5-dimethoxyaniline (200 mg, 0.765 mmol) in DCM (4 mL) was added triethylamine (155 mg, 1.5 mmol, 0.213 mL) and 2-chloroacetyl chloride (173 mg, 1.5 mmol, 0.122 mL). The mixture was stirred at 25° C. for 3 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was partitioned between water (10 mL) and EtOAc (10 mL). The aqueous phase was extracted with EtOAc (10 mL×3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue which was purified by preparative TLC (silica) eluting with 20% EtOAc/petroleum ether. The title compound (131 mg, 49% yield) was obtained as a white solid. M+H+=338.0 (LCMS); 1H NMR (400 MHZ, CD3CN) δ=7.27-7.21 (m, 2H), 7.06-7.00 (m, 2H), 6.46 (t, J=2.3 Hz, 1H), 6.30 (d, J=2.3 Hz, 2H), 4.85 (s, 2H), 4.02 (s, 2H), 3.70 (s, 6H).









TABLE 3





Additional Compounds of the invention


















embedded image


(Compound 1)







embedded image


(Compound 2)







embedded image


(Compound 3)







embedded image


(Compound 4)







embedded image


(Compound 5)







embedded image


(Compound 6)







embedded image


(Compound 7)







embedded image


(Compound 8)







embedded image


(Compound 9)







embedded image


(Compound 10)







embedded image


(Compound 11)







embedded image


(Compound 12)







embedded image


(Compound 13)







embedded image


(Compound 14)







embedded image


(Compound 15)







embedded image


(Compound 16)







embedded image


(Compound 17)







embedded image


(Compound 18)







embedded image


(Compound 19)







embedded image


(Compound 20)







embedded image


(Compound 21)







embedded image


(Compound 22)







embedded image


(Compound 23)







embedded image


(Compound 24)







embedded image


(Compound 25)







embedded image


(Compound 27)







embedded image


(Compound 28)







embedded image


(Compound 29)







embedded image


(Compound 30)







embedded image


(Compound 33)







embedded image


(Compound 43)







embedded image


(Compound 44)







embedded image


(Compound 47)







embedded image


(Compound 51)







embedded image


(Compound 68)







embedded image


(Compound 70)







embedded image


(Compound 73)







embedded image


(Compound 75)







embedded image


(Compound 79)







embedded image


(Compound 84)







embedded image


(Compound 85)







embedded image


(Compound 90)







embedded image


(Compound 92)







embedded image


(Formula 91)







embedded image


(Compound 94)







embedded image


(Compound 93)







embedded image


(Compound 96)







embedded image


(Compound 95)







embedded image


(Compound 98)







embedded image


(Compound 97)







embedded image


(Compound 100)







embedded image


(Compound 99)







embedded image


(Compound 102)







embedded image


(Compound 101)







embedded image


(Compound 104)







embedded image


(Compound 103)







embedded image


(Compound 106)







embedded image


(Compound 105)







embedded image


(Compound 108)







embedded image


(Compound 107)







embedded image


(Compound 110)







embedded image


(Compound 109)







embedded image


(Compound 112)







embedded image


(Compound 111)







embedded image


(Compound 114)







embedded image


(Compound 113)







embedded image


(Compound 116)







embedded image


(Compound 115)







embedded image


(Compound 118)







embedded image


(Compound 117)







embedded image


(Compound 120)







embedded image


(Compound 119)







embedded image


(Compound 122)







embedded image


(Compound 121)







embedded image


(Compound 124)







embedded image


(Compound 123)







embedded image


(Compound 126)







embedded image


(Compound 125)







embedded image


(Compound 128)







embedded image


(Compound 127)







embedded image


(Compound 130)







embedded image


(Compound 129)







embedded image


(Compound 132)







embedded image


(Compound 131)







embedded image


(Compound 134)







embedded image


(Compound 133)







embedded image


(Compound 136)







embedded image


(Compound 135)







embedded image


(Compound 138)







embedded image


(Compound 137)







embedded image


(Compound 140)







embedded image


(Compound 139)







embedded image


(Compound 142)







embedded image


(Compound 141)







embedded image


(Compound 144)







embedded image


(Compound 143)







embedded image


(Compound 146)







embedded image


(Compound 145)







embedded image


(Compound 148)







embedded image


(Compound 147)







embedded image


(Compound 150)







embedded image


(Compound 149)







embedded image


(Compound 152)







embedded image


(Compound 151)







embedded image


(Compound 154)







embedded image


(Compound 153)







embedded image


(Compound 156)







embedded image


(Compound 155)







embedded image


(Compound 158)







embedded image


(Compound 157)







embedded image


(Compound 160)







embedded image


(Compound 159)







embedded image


(Compound 162)







embedded image


(Compound 161)







embedded image


(Compound 164)







embedded image


(Compound 163)







embedded image


(Compound 166)







embedded image


(Compound 165)







embedded image


(Compound 168)







embedded image


(Compound 167)







embedded image


(Compound 170)







embedded image


(Compound 169)







embedded image


(Compound 172)







embedded image


(Compound 171)







embedded image


(Compound 174)







embedded image


(Compound 173)







embedded image


(Compound 176)







embedded image


(Compound 175)







embedded image


(Compound 178)







embedded image


(Compound 177)







embedded image


(Compound 180)







embedded image


(Compound 179)







embedded image


(Compound 182)







embedded image


(Compound 181)







embedded image


(Compound 184)







embedded image


(Compound 183)







embedded image


(Compound 186)







embedded image


(Compound 185)







embedded image


(Compound 188)







embedded image


(Compound 187)







embedded image


(Compound 190)







embedded image


(Compound 189)







embedded image


(Compound 192)







embedded image


(Compound 191)







embedded image


(Compound 194)







embedded image


(Compound 193)







embedded image


(Compound 196)







embedded image


(Compound 195)







embedded image


(Compound 198)







embedded image


(Compound 197)







embedded image


(Compound 200)







embedded image


(Compound 199)







embedded image


(Compound 202)







embedded image


(Compound 201)







embedded image


(Compound 204)







embedded image


(Compound 203)







embedded image


(Compound 206)







embedded image


(Compound 205)







embedded image


(Compound 208)







embedded image


(Compound 207)







embedded image


(Compound 210)







embedded image


(Compound 209)







embedded image


(Compound 212)







embedded image


(Compound 211)







embedded image


(Compound 214)







embedded image


(Compound 213)







embedded image


(Compound 216)







embedded image


(Compound 215)







embedded image


(Compound 218)







embedded image


(Compound 217)







embedded image


(Compound 220)







embedded image


(Compound 219)







embedded image


(Compound 222)







embedded image


(Compound 221)







embedded image


(Compound 224)







embedded image


(Compound 223)







embedded image


(Compound 226)







embedded image


(Compound 225)







embedded image


(Compound 228)







embedded image


(Compound 227)







embedded image


(Compound 230)







embedded image


(Compound 229)







embedded image


(Compound 232)







embedded image


(Compound 231)







embedded image


(Compound 234)







embedded image


(Compound 233)







embedded image


(Compound 236)







embedded image


(Compound 235)







embedded image


(Compound 238)







embedded image


(Compound 237)







embedded image


(Compound 240)







embedded image


(Compound 239)







embedded image


(Compound 242)







embedded image


(Compound 241)







embedded image


(Compound 244)







embedded image


(Compound 243)







embedded image


(Compound 246)







embedded image


(Compound 245)







embedded image


(Compound 248)







embedded image


(Compound 247)







embedded image


(Compound 250)







embedded image


(Compound 249)







embedded image


(Compound 252)







embedded image


(Compound 251)







embedded image


(Compound 254)







embedded image


(Compound 253)







embedded image


(Compound 256)







embedded image


(Compound 255)







embedded image


(Compound 258)







embedded image


(Compound 257)







embedded image


(Compound 260)







embedded image


(Compound 259)







embedded image


(Compound 262)







embedded image


(Compound 261)







embedded image


(Compound 264)







embedded image


(Compound 263)







embedded image


(Compound 266)







embedded image


(Compound 265)







embedded image


(Compound 268)







embedded image


(Compound 267)







embedded image


(Compound 270)







embedded image


(Compound 269)







embedded image


(Compound 272)







embedded image


(Compound 271)







embedded image


(Compound 274)







embedded image


(Compound 273)







embedded image


(Compound 276)







embedded image


(Compound 275)







embedded image


(Compound 278)







embedded image


(Compound 277)







embedded image


(Compound 280)







embedded image


(Compound 279)







embedded image


(Compound 282)







embedded image


(Compound 281)







embedded image


(Compound 284)







embedded image


(Compound 283)







embedded image


(Compound 286)







embedded image


(Compound 285)







embedded image


(Compound 288)







embedded image


(Compound 287)







embedded image


(Compound 290)







embedded image


(Compound 289)







embedded image


(Compound 292)







embedded image


(Compound 291)







embedded image


(Compound 294)







embedded image


(Compound 293)







embedded image


(Compound 296)







embedded image


(Compound 295)







embedded image


(Compound 298)







embedded image


(Compound 297)







embedded image


(Compound 300)







embedded image


(Compound 299)







embedded image


(Compound 302)







embedded image


(Compound 301)







embedded image


(Compound 304)







embedded image


(Compound 303)







embedded image


(Compound 306)







embedded image


(Compound 305)







embedded image


(Compound 308)







embedded image


(Compound 307)







embedded image


(Compound 310)







embedded image


(Compound 309)







embedded image


(Compound 312)







embedded image


(Compound 311)







embedded image


(Compound 314)







embedded image


(Compound 313)







embedded image


(Compound 316)







embedded image


(Compound 315)







embedded image


(Compound 318)







embedded image


(Compound 317)







embedded image


(Compound 320)







embedded image


(Compound 319)







embedded image


(Compound 322)







embedded image


(Compound 321)







embedded image


(Compound 324)







embedded image


(Compound 323)







embedded image


(Compound 326)







embedded image


(Compound 325)







embedded image


(Compound 328)







embedded image


(Compound 327)







embedded image


(Compound 330)







embedded image


(Compound 329)







embedded image


(Compound 332)







embedded image


(Compound 331)







embedded image


(Compound 334)







embedded image


(Compound 333)







embedded image


(Compound 336)







embedded image


(Compound 335)







embedded image


(Compound 338)







embedded image


(Compound 337)







embedded image


(Compound 340)







embedded image


(Compound 339)







embedded image


(Compound 342)







embedded image


(Compound 341)







embedded image


(Compound 344)







embedded image


(Compound 343)







embedded image


(Compound 346)







embedded image


(Compound 345)







embedded image


(Compound 348)







embedded image


(Compound 347)







embedded image


(Compound 350)







embedded image


(Compound 349)







embedded image


(Compound 352)







embedded image


(Compound 351)







embedded image


(Compound 354)







embedded image


(Compound 353)







embedded image


(Compound 356)







embedded image


(Compound 355)







embedded image


(Compound 358)







embedded image


(Compound 357)







embedded image


(Compound 360)







embedded image


(Compound 359)







embedded image


(Compound 362)







embedded image


(Compound 361)







embedded image


(Compound 364)







embedded image


(Compound 363)







embedded image


(Compound 366)







embedded image


(Compound 365)







embedded image


(Compound 368)







embedded image


(Compound 367)







embedded image


(Compound 370)







embedded image


(Compound 369)







embedded image


(Compound 372)







embedded image


(Compound 371)







embedded image


(Compound 374)







embedded image


(Compound 373)







embedded image


(Compound 376)







embedded image


(Compound 375)







embedded image


(Compound 378)







embedded image


(Compound 377)







embedded image


(Compound 380)







embedded image


(Compound 379)







embedded image


(Compound 382)







embedded image


(Compound 381)







embedded image


(Compound 384)







embedded image


(Compound 383)







embedded image


(Compound 386)







embedded image


(Compound 385)







embedded image


(Compound 388)







embedded image


(Compound 387)







embedded image


(Compound 390)







embedded image


(Compound 389)







embedded image


(Compound 392)







embedded image


(Compound 391)







embedded image


(Compound 394)







embedded image


(Compound 393)







embedded image


(Compound 396)







embedded image


(Compound 395)







embedded image


(Compound 398)







embedded image


(Compound 397)







embedded image


(Compound 400)







embedded image


(Compound 399)







embedded image


(Compound 402)







embedded image


(Compound 401)







embedded image


(Compound 404)







embedded image


(Compound 403)







embedded image


(Compound 406)







embedded image


(Compound 405)







embedded image


(Compound 408)







embedded image


(Compound 407)







embedded image


(Compound 410)







embedded image


(Compound 409)







embedded image


(Compound 412)







embedded image


(Compound 411)







embedded image


(Compound 414)







embedded image


(Compound 413)







embedded image


(Compound 416)







embedded image


(Compound 415)







embedded image


(Compound 418)







embedded image


(Compound 417)







embedded image


(Compound 420)







embedded image


(Compound 419)







embedded image


(Compound 422)







embedded image


(Compound 421)







embedded image


(Compound 424)







embedded image


(Compound 423)







embedded image


(Compound 426)







embedded image


(Compound 425)







embedded image


(Compound 428)







embedded image


(Compound 427)







embedded image


(Compound 430)







embedded image


(Compound 429)







embedded image


(Compound 432)







embedded image


(Compound 431)







embedded image


(Compound 434)







embedded image


(Compound 433)







embedded image


(Compound 436)







embedded image


(Compound 435)







embedded image


(Compound 438)







embedded image


(Compound 437)







embedded image


(Compound 440)







embedded image


(Compound 439)







embedded image


(Compound 442)







embedded image


(Compound 441)







embedded image


(Compound 444)







embedded image


(Compound 443)







embedded image


(Compound 446)







embedded image


(Compound 445)







embedded image


(Compound 448)







embedded image


(Compound 447)







embedded image


(Compound 450)







embedded image


(Compound 449)







embedded image


(Compound 452)







embedded image


(Compound 451)







embedded image


(Compound 454)







embedded image


(Compound 453)







embedded image


(Compound 456)







embedded image


(Compound 455)







embedded image


(Compound 458)







embedded image


(Compound 457)







embedded image


(Compound 460)







embedded image


(Compound 459)







embedded image


(Compound 462)







embedded image


(Compound 461)







embedded image


(Compound 464)







embedded image


(Compound 463)







embedded image


(Compound 466)







embedded image


(Compound 465)







embedded image


(Compound 468)







embedded image


(Compound 467)







embedded image


(Compound 470)







embedded image


(Compound 469)







embedded image


(Compound 472)







embedded image


(Compound 471)







embedded image


(Compound 474)







embedded image


(Compound 473)







embedded image


(Compound 476)







embedded image


(Compound 475)







embedded image


(Compound 478)







embedded image


(Compound 477)







embedded image


(Compound 480)







embedded image


(Compound 479)







embedded image


(Compound 482)







embedded image


(Compound 481)







embedded image


(Compound 484)







embedded image


(Compound 483)







embedded image


(Compound 486)







embedded image


(Compound 485)







embedded image


(Compound 488)







embedded image


(Compound 487)







embedded image


(Compound 490)







embedded image


(Compound 489)







embedded image


(Compound 492)







embedded image


(Compound 491)







embedded image


(Compound 494)







embedded image


(Compound 493)







embedded image


(Compound 496)







embedded image


(Compound 495)







embedded image


(Compound 498)







embedded image


(Compound 497)







embedded image


(Compound 500)







embedded image


(Compound 499)







embedded image


(Compound 502)







embedded image


(Compound 501)







embedded image


(Compound 504)







embedded image


(Compound 503)







embedded image


(Compound 506)







embedded image


(Compound 505)







embedded image


(Compound 508)







embedded image


(Compound 507)







embedded image


(Compound 510)







embedded image


(Compound 509)







embedded image


(Compound 512)







embedded image


(Compound 511)







embedded image


(Compound 514)







embedded image


(Compound 513)







embedded image


(Compound 516)







embedded image


(Compound 515)







embedded image


(Compound 518)







embedded image


(Compound 517)







embedded image


(Compound 520)







embedded image


(Compound 519)







embedded image


(Compound 522)







embedded image


(Compound 521)







embedded image


(Compound 524)







embedded image


(Compound 523)







embedded image


(Compound 526)







embedded image


(Compound 525)







embedded image


(Compound 528)







embedded image


(Compound 527)







embedded image


(Compound 530)







embedded image


(Compound 529)







embedded image


(Compound 532)







embedded image


(Compound 531)







embedded image


(Compound 534)







embedded image


(Compound 533)







embedded image


(Compound 536)







embedded image


(Compound 535)







embedded image


(Compound 538)







embedded image


(Compound 537)







embedded image


(Compound 540)







embedded image


(Compound 539)







embedded image


(Compound 542)







embedded image


(Compound 541)







embedded image


(Compound 544)







embedded image


(Compound 543)







embedded image


(Compound 546)







embedded image


(Compound 545)







embedded image


(Compound 548)







embedded image


(Compound 547)







embedded image


(Compound 550)







embedded image


(Compound 549)







embedded image


(Compound 552)







embedded image


(Compound 551)







embedded image


(Compound 554)







embedded image


(Compound 553)







embedded image


(Forumla 556)







embedded image


(Compound 555)







embedded image


(Compound 558)







embedded image


(Compound 557)







embedded image


(Compound 560)







embedded image


(Compound 559)







embedded image


(Compound 562)







embedded image


(Compound 561)







embedded image


(Compound 564)







embedded image


(Compound 563)







embedded image


(Compound 565)







embedded image


(Compound 566)







embedded image


(Compound 567)







embedded image


(Compound 568)







embedded image


(Compound 569)







embedded image


(Compound 570)







embedded image


(Compound 571)







embedded image


(Compound 572)







embedded image


(Compound 573)







embedded image


(Compound 574)







embedded image


(Compound 575)







embedded image


(Compound 576)







embedded image


(Compound 577)







embedded image


(Compound 578)







embedded image


(Compound 579)







embedded image


(Compound 580)







embedded image


(Compound 581)







embedded image


(Compound 582)







embedded image


(Compound 583)







embedded image


(Compound 584)







embedded image


(Compound 585)







embedded image


(Compound 586)







embedded image


(Compound 587)







embedded image


(Compound 588)







embedded image


(Compound 589)







embedded image


(Compound 590)







embedded image


(Compound 591)







embedded image


(Compound 592)







embedded image


(Compound 593)







embedded image


(Compound 594)







embedded image


(Compound 595)







embedded image


(Compound 596)







embedded image


(Compound 597)







embedded image


(Compound 598)







embedded image


(Compound 599)







embedded image


(Compound 600)







embedded image


(Compound 601)







embedded image


(Compound 602)







embedded image


(Compound 603)







embedded image


(Compound 604)







embedded image


(Compound 605)







embedded image


(Compound 606)







embedded image


(Compound 607)







embedded image


(Compound 608)







embedded image


(Compound 609)







embedded image


(Compound 610)







embedded image


(Compound 611)







embedded image


(Compound 612)







embedded image


(Compound 613)







embedded image


(Compound 614)







embedded image


(Compound 615)







embedded image


(Compound 616)







embedded image


(Compound 617)







embedded image


(Compound 618)







embedded image


(Compound 619)







embedded image


(Compound 620)







embedded image


(Compound 621)







embedded image


(Compound 622)







embedded image


(Compound 623)







embedded image


(Compound 624)







embedded image


(Compound 625)







embedded image


(Compound 626)







embedded image


(Compound 627)







embedded image


(Compound 628)







embedded image


(Compound 629)







embedded image


(Compound 630)







embedded image


(Compound 631)







embedded image


(Compound 632)







embedded image


(Compound 633)







embedded image


(Compound 634)







embedded image


(Compound 635)







embedded image


(Compound 636)







embedded image


(Compound 637)







embedded image


(Compound 638)







embedded image


(Compound 639)







embedded image


(Compound 640)







embedded image


(Compound 641)







embedded image


(Compound 642)







text missing or illegible when filed















TABLE 4





Chemical structures of five ferroptosis-inducing compounds from


Enamine covalent inhibitors


















embedded image


Z56943362







embedded image


Z85921962







embedded image


Z104378216







embedded image


Z90660973







embedded image


Z56931816
















TABLE 5







Compound 8216 derivatives











Compound
Average
Stdev















Z1562139496
0.000174
5.9399E−05



Z199424090
0.00039001
0.00011031



Z56946047
0.00062402
6.7884E−05



Z147647938
0.00093603
0.00022062



Z4425210974
0.00128404
0.00015274



Z3837055210
0.00158404
0.00088249



Z4323362848
0.00162605
0.00019517



Z3629871044
0.00176405
0.00186681



Z381549666
0.00183605
0.00030548



Z56891378
0.00187205
0.00074673



Z4425210932
0.00192605
8.4855E−06



Z4425211258
0.00193805
0.00044973



Z56837054
0.00199206
0.00098432



Z4323349920
0.00202806
0.00071278



Z3837267295
0.00211206
3.3942E−05



Z3766260223
0.00223806
0.0008231



Z57052885
0.00224406
0.00015274



Z4425210755
0.00256207
0.00092492



Z1562136806
0.00264007
0.00074673



Z3766269910
0.00290408
0.00173105



Z4323320274
0.00292208
0.00021214



Z3766270794
0.0034741
0.00109463



Z4323385212
0.00376811
0.00049216



Z4425210756
0.00393011
0.0008231



Z4323486432
0.00442812
0.00210441



Z56837076
0.00469813
0.002639



Z4323314193
0.00483014
0.00104372



Z4425210609
0.00525615
0.00110312



Z108565162
0.00539415
0.00067036



Z3766271747
0.00561616
0.00218926



Z147647668
0.00583816
0.00206198



Z147647684
0.00657618
0.00113706



Z4425210448
0.00756021
0.00032245



Z3837152585
0.00768622
0.00092492



Z3837300268
0.00828023
0.00280022



Z56926666
0.00862224
0.00039882



RSL3
0.00933026
0.00107766



Z3837046637
0.00972027
0.00123889



Z89264994
0.01228234
0.00423427



Z3837197646
0.01316437
0.00268142



Z4323386801
0.01527043
0.00060247



Z4323373087
0.01670447
0.00079764



Z4425210708
0.0178325
0.00302084



Z4425210960
0.01924854
0.00156134



Z3766264621
0.0215166
0.00610957



Z3952175509
0.03046885
0.00453127



Z147647920
0.04392123
5.0913E−05



Z4323422705
0.04398123
0.00352998



Z4323310732
0.05331749
0.00432761



Z90122115
0.05781762
0.00098432



Z4425211012
0.07480409
0.03001328



Z4170009630
0.0891565
0.0032839



Z147647648
0.10065882
0.02391219



Z3837255186
0.13307773
0.01967792



Z1687602192
0.14450805
0.06494816



Z4425210882
0.1465421
0.00657628



Z3952174992
0.15167825
0.0843206



Z3837096793
0.17009876
0.01541819



Z3837506617
0.32674515
0.04773953



Z2596884436
0.40261127
0.01491754



Z4323324870
0.48795766
0.02530382



Z3837050450
0.51481441
0.0032245



Z3837366035
0.53099687
0.07527504



Z3766262414
0.54812735
0.03192252



Z4323355648
0.56906193
0.04350526



Z4323341765
0.70080162
0.10054491



Z3837644318
0.74069074
0.0241413



Z147652330
0.74589289
0.06537244



Z1562122996
0.75753921
0.11933185



Z3887631385
0.79383423
0.11932337



Z3952175125
0.88756285
0.05898284



Z4283095766
0.93423216
0.09513115



Z3337549802
1.03385295
0.08728205



Z2020497058
1.0891505
0.04266519



Z3217399415
1.25468913
0.0540273

















TABLE 6





Autogrow4 independently discovers nine 8216 derivatives


as potential GPX4 binders.









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image











Biological evaluation of working examples A1-A47 and B1-B28 was performed in HT1080 cells (ATCC, cat #CCL-121) according to the procedure provided below.


1. Cell Seeding





    • 1.1 Cells were harvested from flask into cell culture medium and then the cell number count was determined.

    • 1.2 The cells were suspended with 1.5 μM ferrostatin-1 and without ferrostatin-1.

    • 1.3 Cells were plated at a density of 1,500 cells per well in a 384-well black sided clear bottom plate, in 25 μL, using electronic multichannel pipette.

    • 1.4 Plates were covered with lid and spun for 1 min at 1000 rpm, then allowed to incubate overnight at 37° C. under 5% CO2.





2. Compound Treatment





    • 2.1 Compounds were dissolved at 10 mM stock solution and 10 μl of diluted solution was transferred to a 384 LDV-plate (LABCYTE, LP-0200). A 3 fold, 11-point dilution was performed.

    • 2.2 Cells were treated with compounds by plate reformat Echo software and spun for 1 min at 1000 rpm. Then incubated at 37° C. for 24 hrs in a CO2 incubator.





3. Readout





    • 3.1 The plates were removed from the incubator and allowed to equilibrate to room temperature, at least 30 minutes.

    • 3.2 CellTiter-Glo® 2.0 Reagent (Promega, G9242) was added (25 μL) to the assay plates and spun for 1 min at 1000 rpm. Then allowed to stand for about 20 minutes before reading luminescence signal on Envision.

    • 3.3 The IC50 calculations were determined with a nonlinear fit, variable slope (four parameters) log(inhibitor) vs. response model using GraphPad PRISM. Results for compounds of the invention are provided in Tables 7 and 8.














TABLE 7





Working

IC50


Example
Name
(nM)







A1
2-chloro-N-(3,5-dimethylphenyl)-N-[(4-
C



fluorophenyl)methyl]acetamide


A2
2-chloro-N-(3,5-dimethylphenyl)-N-[(4-
B



methoxyphenyl)methyl]acetamide


A3
2-chloro-N-(3,5-dimethoxyphenyl)-N-[(4-
B



fluorophenyl)methyl]acetamide


A4
2-chloro-N-[(4-chlorophenyl)methyl]-N-(3,5-
C



dimethylphenyl)acetamide


A5
2-chloro-N-(3,5-dimethylphenyl)-N-[(3-
B



methoxyphenyl)methyl]acetamide


A6
2-chloro-N-[(2-chlorophenyl)methyl]-N-(3,5-
C



dimethylphenyl)acetamide


A7
2-chloro-N-(3,5-dimethylphenyl)-N-[(2-
B



methoxyphenyl)methyl]acetamide


A8
2-chloro-N-(3,5-dimethylphenyl)-N-(m-
B



tolylmethyl)acetamide


A9
2-chloro-N-[(3-chlorophenyl)methyl]-N-(3,5-
B



dimethylphenyl)acetamide


A10
2-chloro-N-(3,5-dimethylphenyl)-N-(p-
C



tolylmethyl)acetamide


A11
2-chloro-N-(3,5-dimethylphenyl)-N-(o-
C



tolylmethyl)acetamide


A12
2-chloro-N-cyclohexyl-N-[(4-
C



fluorophenyl)methyl]acetamide


A13
2-chloro-N-[(4-fluorophenyl)methyl]-N-(3-
C



methoxyphenyl)acetamide


A14
2-chloro-N-(3-chloro-5-methoxy-phenyl)-N-[(4-
C



fluorophenyl)methyl]acetamide


A15
2-chloro-N-(3,5-difluorophenyl)-N-[(4-
D



fluorophenyl)methyl]acetamide


A16
2-chloro-N-(3,5-dichlorophenyl)-N-[(4-
C



fluorophenyl)methyl]acetamide


A17
2-chloro-N-(3-chloro-4-methoxy-phenyl)-N-[(4-
C



fluorophenyl)methyl]acetamide


A18
2-chloro-N-(3-chlorophenyl)-N-[(4-
C



fluorophenyl)methyl]acetamide


A19
2-chloro-N-[(4-cyanophenyl)methyl]-N-(3,5-
B



dimethoxyphenyl)acetamide


A20
2-chloro-N-[(2,3-difluorophenyl)methyl]-N-(3,5-
B



dimethoxyphenyl)acetamide


A21
2-chloro-N-(3,5-dimethoxyphenyl)-N-[(4-
B



methoxyphenyl)methyl]acetamide


A22
2-chloro-N-[(4-fluorophenyl) methyl]-N-(6-
C



quinolyl)acetamide


A23
N-[(2-bromo-4-fluoro-phenyl)methyl]-2-chloro-
B



N-(3,5-dimethoxyphenyl)acetamide


A24
2-chloro-N-(3,5-dimethoxyphenyl)-N-[(4-
C



hydroxyphenyl)methyl]acetamide


A25
2-chloro-N-[(4-fluorophenyl)methyl]-N-(3-
C



methoxy-5-methyl-phenyl)acetamide


A26
2-chloro-N-[(2,6-dichloro-4-fluoro-phenyl)methyl]-
B



N-(3,5-dimethoxyphenyl)acetamide


A27
2-chloro-N-(3,5-dimethoxyphenyl)-N-[[4-fluoro-2-
B



(trifluoromethyl)phenyl]methyl]acetamide


A28
2-chloro-N-[(5-cyano-2-fluoro-phenyl)methyl]-N-
B



(3,5-dimethoxyphenyl)acetamide


A29
2-chloro-N-[(4-fluorophenyl)methyl]-N-(3-methoxy-
B



4-methyl-phenyl)acetamide


A30
2-chloro-N-(3,5-dimethoxyphenyl)-N-[(4-fluoro-
A



2,6-dimethyl-phenyl)methyl]acetamide


A31
2-chloro-N-(3,5-dimethoxyphenyl)-N-[[2-fluoro-
B



3-(trifluoromethyl)phenyl]methyl]acetamide


A32
2-chloro-N-(2-fluoro-6-methoxy-phenyl)-N-[(4-
D



fluorophenyl)methyl]acetamide


A33
2-chloro-N-(3,5-dimethoxyphenyl)-N-[(4-fluoro-
A



3-methoxy-phenyl)methyl]acetamide


A34
2-chloro-N-(3-chloro-5-hydroxy-phenyl)-N-[(4-
C



fluorophenyl)methyl]acetamide


A35
2-chloro-N-(2-chloro-3-methoxy-phenyl)-N-[(4-
C



fluorophenyl)methyl]acetamide


A36
2-chloro-N-(2-cyano-4-methoxy-phenyl)-N-[(4-
D



fluorophenyl)methyl]acetamide


A37
2-chloro-N-(2-chloro-5-methoxy-phenyl)-N-[(4-
C



fluorophenyl)methyl]acetamide


A38
N-[(4-bromo-2,3-difluoro-phenyl)methyl]-2-
A



chloro-N-(3,5-dimethoxyphenyl)acetamide


A39
N-(3-bromo-1H-pyrazolo[3,4-b]pyridin-5-yl)-
D



2-chloro-N-[(4-fluorophenyl)methyl]acetamide


A40
2-chloro-N-[(4-fluorophenyl)methyl]-N-
B



(4-methoxy-3,5-dimethyl-phenyl)acetamide


A41
2-chloro-N-[(4-fluorophenyl)methyl]-N-
E



(4-hydroxy-3,5-dimethyl-phenyl)acetamide


A42
2-chloro-N-(2,4-dichloro-3-methyl-phenyl)-
C



N-[(4-fluorophenyl)methyl]acetamide


A43
2-chloro-N-[(2-chloro-3,4-difluoro-phenyl)methyl]-
A



N-(3,5-dimethoxyphenyl)acetamide


A44
2-chloro-N-(3,5-dimethoxyphenyl)-N-[(3-
B



fluoro-5-methyl-phenyl)methyl]acetamide


A45
N-(2,1,3-benzothiadiazol-4-ylmethyl)-2-
B



chloro-N-(3,5-dimethoxyphenyl)acetamide


A46
2-chloro-N-[(4-fluorophenyl)methyl]-N-
C



(2-methoxy-4-pyridyl)acetamide


A47
2-chloro-N-[(4-fluorophenyl)methyl]-N-
E



(6-methyl-3-pyridyl)acetamide





Note:


Assay IC50 data are designated within the following ranges:


A: ≤10 nM


B: >10 nM to ≤100 nM


C: >100 nM to ≤1,000 Nm


D: >1 μM to ≤10 μM


E: >10 μM















TABLE 8





Working

IC50


Example
Name
(nM)







B1
N-[4-(4-bromophenyl)thiazol-2-yl]-2-chloro-N-(3-




methoxypropyl)acetamide


B2
N-[4-(4-bromophenyl)thiazol-2-yl]-N-(3-
D



methoxypropyl)prop-2-ynamide


B3
2-chloro-N-[4-(4-methoxyphenyl)thiazol-2-yl]-
D



N-(3-methoxypropyl)acetamide


B4
N-[4-(4-methoxyphenyl)thiazol-2-yl]-N-(3-




methoxypropyl)prop-2-ynamide


B5
N-[4-(4-bromophenyl)thiazol-2-yl]-2-chloro-
C



N-(3,5-dimethylphenyl)acetamide


B6
N-[4-(4-bromophenyl)thiazol-2-yl]-2-chloro-




N-cyclohexyl-acetamide


B7
N-[4-(4-bromophenyl)thiazol-2-yl]-N-(3,5-
E



dimethylphenyl)prop-2-enamide


B8
N-[4-(4-bromophenyl)thiazol-2-yl]-N-(3,5-
E



dimethylphenyl)but-2-ynamide


B9
2-chloro-N-(3,5-dimethylphenyl)-N-[4-(4-
C



methoxyphenyl)thiazol-2-yl]acetamide


B10
2-chloro-N-(4-cyclohexylthiazol-2-yl)-N-(3,5-
C



dimethylphenyl)acetamide


B11
2-chloro-N-(3,5-dimethylphenyl)-N-(4-
C



phenylthiazol-2-yl)acetamide


B12
N-[4-(3-bromophenyl)thiazol-2-yl]-2-chloro-
C



N-(3,5-dimethylphenyl)acetamide


B13
N-[4-(4-bromophenyl)thiazol-2-yl]-N-(3,5-
B



dimethylphenyl)prop-2-ynamide


B14
N-benzyl-N-[4-(4-bromophenyl)thiazol-2-yl]-
D



2-chloro-acetamide


B15
N-[4-(4-bromophenyl)thiazol-2-yl]-2-chloro-
E



N-phenyl-acetamide


B16
N-[4-(4-bromophenyl)thiazol-2-yl]-2-chloro-
E



N-(m-tolyl)acetamide


B17
N-[4-(4-bromophenyl)thiazol-2-yl]-2-chloro-
C



N-(3-methoxyphenyl)acetamide


B18
N-[4-(4-bromophenyl)thiazol-2-yl]-2-chloro-




N-(3,5-dimethoxyphenyl)acetamide


B19
N-[4-(4-bromophenyl)thiazol-2-yl]-
B



N-(3,5-dimethoxyphenyl)prop-2-ynamide


B20
N-[4-(4-bromophenyl)thiazol-2-yl]-2-chloro-
D



N-(3,5-dichlorophenyl)acetamide


B21
N-[4-(4-bromophenyl)thiazol-2-yl]-N-(3,5-
B



dimethylphenyl)-2-iodo-acetamide


B22
N-[4-(4-bromophenyl)thiazol-2-yl]-N-(3,5-
B



dichlorophenyl)prop-2-ynamide


B23
N-[4-(4-bromophenyl)thiazol-2-yl]-N-(3,5-
E



dimethylphenyl)-2-fluoro-prop-2-enamide


B24
N-[4-(4-bromophenyl)thiazol-2-yl]-2-chloro-
C



N-(3-chloro-5-methoxy-phenyl)acetamide


B25
N-[4-(4-bromophenyl)thiazol-2-yl]-2-chloro-
E



N-(3,5-difluorophenyl)acetamide


B26
N-[4-(4-bromophenyl)thiazol-2-yl]-N-(3-chloro-
B



5-methoxy-phenyl)prop-2-ynamide


B27
N-[4-(4-bromophenyl)thiazol-2-yl]-N-(3,5-
E



dimethylphenyl)-2-fluoro-acetamide


B28
N-[4-(4-bromophenyl)thiazol-2-yl]-N-(3,5-
E



dimethylphenyl)oxirane-2-carboxamide





Note:


Assay IC50 data are designated within the following ranges:


A: ≤10 nM


B: >10 nM to ≤100 nM


C: >100 nM to ≤1,000 Nm


D: >1 μM to ≤10 μM


E: >10 μM





Claims
  • 1-42. (canceled)
  • 43. A method of treating cancer in a subject, comprising administering to the subject a pharmaceutically effective amount of a compound having any of the structures of Formulas 1-5 or 91, or a pharmaceutically acceptable salt thereof.
  • 44. The method of claim 43, wherein the method reduces the growth rate of a tumor in the subject, reduces the size of a tumor in the subject, eliminates a tumor in the subject, or delays progression of a cancer stage in the subject.
  • 45. The method of claim 43, wherein the cancer comprises a mesenchymal cancer.
  • 46. The method of claim 43, wherein the cancer is a sarcoma.
  • 47. The method of claim 43, wherein the compound inhibits GPX4 enzyme.
  • 48. The method of claim 43, wherein the compound induces ferroptosis in a cell.
  • 49. The method of claim 48, wherein the cell is a cancer cell.
  • 50. The method of claim 43, wherein the compound is selected from Compounds 1-643.
  • 51. The method of claim 43, wherein the compound is represented by a structure in Table 1.
  • 52. The method of claim 43, wherein the compound is represented by a structure in Table 2.
  • 53. The method of claim 43, wherein the compound is represented by a structure in Table 3.
  • 54. The method of claim 44, wherein the compound is selected from the group consisting of:
  • 55. A method for inhibiting GPX4 enzyme, the method comprising contacting the GPX4 enzyme with a compound selected from the group:
  • 56. A compound, or a pharmaceutically acceptable salt thereof, having a structure selected from the group:
  • 57. A pharmaceutical composition comprising a compound, or a pharmaceutically acceptable salt thereof of claim 56.
CROSS REFERENCE

This application is a continuation application of International Application No. PCT/US2022/040818, filed Aug. 18, 2022, which claims the benefit of U.S. Provisional Application No. 63/313,000, filed Feb. 23, 2022; U.S. Provisional Application No. 63/295,007, filed Dec. 30, 2021; U.S. Provisional Application No. 63/286,022, filed Dec. 4, 2021; and U.S. Provisional Application No. 63/234,829, filed Aug. 19, 2021; each of which is incorporated by reference in its entirety.

Provisional Applications (4)
Number Date Country
63234829 Aug 2021 US
63286022 Dec 2021 US
63295007 Dec 2021 US
63313000 Feb 2022 US
Continuations (1)
Number Date Country
Parent PCT/US2022/040818 Aug 2022 WO
Child 18441446 US