Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TARGETING PHOSPHOLIPASE A2 (PLA2) IN CANCER
Document Type and Number:
WIPO Patent Application WO/2024/077085
Kind Code:
A1
Abstract:
The present disclosure describes methods of treating a condition comprising administering a compound that inhibits PLA2G7. The present disclosure describes methods of treating a condition comprising administering a compound that inhibits PAFAH2. The disclosed methods can be used to treat cancer and reduce the progression of cancers that comprise one or more RAS mutations.

Inventors:
HELLER DANIEL (US)
JENA PRAKRIT (US)
HOROSZKO CHRISTOPHER (US)
TYLAWSKY DANIEL (US)
RUIZ STEPHEN (US)
Application Number:
PCT/US2023/075987
Publication Date:
April 11, 2024
Filing Date:
October 04, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MEMORIAL SLOAN KETTERING CANCER CENTER (US)
MEMORIAL HOSPITAL FOR CANCER AND ALLIED DISEASES (US)
SLOAN KETTERING INST CANCER RES (US)
International Classes:
C07D239/70; A61P35/00
Attorney, Agent or Firm:
MCPARLAND, James P. et al. (US)
Download PDF:
Claims:
CLAIMS

WHAT IS CLAIMED IS:

1. A method of treating a condition, the method comprising administering to a subject in need thereof a therapeutically-effective amount of a compound that inhibits PLA2G7 and a reduced amount of a second therapy, wherein the reduced amount of the second therapy is therapeutically effective for treating the condition in combination with the therapeutically-effective amount of the compound that inhibits PLA2G7, and wherein the reduced amount of the second therapy is less than an amount of the second therapy that is therapeutically effective for the condition in absence of the therapeutically-effective amount of the compound that inhibits PLA2G7.

2. The method of claim 1, wherein the compound that inhibits PLA2G7 is darapladib.

3. The method of claim 1, wherein the compound that inhibits PLA2G7 is rilapladib.

4. The method of claim 1, wherein the compound that inhibits PLA2G7 is AA39-2.

5. The method of claim 1, wherein the compound that inhibits PLA2G7 is ML256.

6. The method of claim 1, wherein the compound that inhibits PLA2G7 inhibits PAFAH2.

7. A method of treating a condition, the method comprising administering to a subject in need thereof a therapeutically-effective amount of a compound that inhibits PAFAH2 and a reduced amount of a second therapy, wherein the reduced amount of the second therapy is therapeutically effective for treating the condition in combination with the therapeutically-effective amount of the compound that inhibits PAFAH2, and wherein the reduced amount of the second therapy is less than an amount of the second therapy that is therapeutically effective for the condition in absence of the therapeutically-effective amount of the compound that inhibits PAFAH2.

8. The method of claim 7, wherein the compound that inhibits PAFAH2 is darapladib.

9. The method of claim 7, wherein the compound that inhibits PAFAH2 is rilapladib. The method of claim 7, wherein the compound that inhibits PAFAH2 is AA39-2 The method of claim 7, wherein the compound that inhibits PAFAH2 is ML256. The method of claim 7, wherein the compound that inhibits PAFAH2 inhibits

PLA2G7. The method of any one of claims 1-12, wherein the second therapy comprises a chemotherapeutic agent or radiation therapy. The method of any one of claim 13, further comprising administering to the subject a therapeutically-effective amount of a ferroptosis inducer. The method of any one of claim 14, wherein the ferroptosis inducer exhibits synergy with the compound that inhibits PLA2G7. The method of any one of claim 14, wherein the ferroptosis inducer exhibits synergy with the compound that inhibits PAFAH2. The method of any one of claim 14, wherein the ferroptosis inducer is a cystineglutamate system Xc- inhibitor. The method of any one of claim 14, wherein the ferroptosis inducer is a glutathione peroxidase (GPX) inhibitor. The method of any one of claim 1-18, further comprising administering to the subject a therapeutically-effective amount of a modulator of oxidative stress. The method of any one of claims 1-19, wherein the reduced amount of the second therapy comprises administering a reduced dosage of the second therapy to the subject. The method of any one of claims 1-19, wherein the reduced amount of the second therapy comprises administering the second therapy to the subject at a reduced frequency. The method of any one of claims 1-19, wherein the reduced amount of the second therapy comprises administering a reduced dosage of the second therapy to the subject at a reduced frequency. A method of treating a condition, the method comprising administering to a subject in need thereof a therapeutically-effective amount of a first compound that inhibits PLA2G7 and a therapeutically-effective amount of a second compound that exhibits synergy with the first compound that inhibits PLA2G7 for the condition. A method of treating a condition, the method comprising administering to a subject in need thereof a therapeutically-effective amount of a first compound that inhibits PAFAH2 and a therapeutically-effective amount of a second compound that exhibits synergy with the first compound that inhibits PAFAH2 for the condition. The method of claim 23 or 24, wherein the second compound comprises a ferroptosis inducer. The method of claim 25, wherein the ferroptosis inducer is a cystine-glutamate system Xc- inhibitor. The method of claim 25, wherein the ferroptosis inducer is a glutathione peroxidase (GPX) inhibitor. The method of claim 25, wherein the second compound comprises a modulator of oxidative stress. The method of any one of claims 1-28, wherein the condition is cancer. The method of claim 29, wherein the cancer harbors a RAS mutation. The method of claim 29, wherein the RAS mutation is KRAS. The method of claim 29, wherein the RAS mutation is NRAS. The method of claim 29, wherein the RAS mutation is HRAS. The method of any one of claims 30-33, wherein the RAS mutation is a constitutively active variant. The method of claim 34, wherein the constitutively active variant is a gain of function mutation. The method of claim 34, wherein the constitutively active variant is a duplication. The method of claim 34, wherein the constitutively active variant is a gene amplification. The method of claim 29, wherein the cancer harbors an STK11 mutation. The method of claim 29, wherein the cancer harbors an KEAP1 mutation. The method of claim 29, wherein the cancer harbors an STK11 mutation an KEAP1 mutation. The method of any one of claims 29-37, wherein the cancer further harbors an STK11 mutation. The method of any one of claims 29-37, wherein the cancer further harbors an KEAP1. The method of any one of claims 29-37, wherein the cancer further harbors an STK11 mutation an KE API mutation. The method of any one of claims 29-43, wherein the cancer cells from the subject exhibit elevated expression or activity of PLA2G7 compared to healthy control cells. The method of any one of claim 29-43, wherein the cancer cells from the subject exhibit elevated expression or activity of PAFAH2 compared to healthy control cells. The method of any one of claims 29-45, wherein the cancer is lung cancer. The method of any one of claims 29-45, wherein the cancer is lymphoma. The method of any one of claims 29-45, wherein the cancer is non-small cell lung. The method of any one of claims 29-45, wherein the cancer is pancreatic cancer. The method of any one of claims 29-45, wherein the cancer is clear-cell renal cell carcinoma. The method of any one of claims 29-45, wherein the cancer is diffuse large B-cell lymphoma.

52. The method of any one of claims 29-45, wherein the cancer is leukemia.

53. The method of any one of claims 29-45, wherein the cancer is astrocytoma.

54. The method of any one of claims 1-53, wherein the administering comprises oral administration.

55. The method of any one of claims 1-53, wherein the administering intravenous administration.

56. The method of any one of claims 1-53, wherein the administering intramuscular administration.

57. A method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a non -wild type RAS gene; and b) based on the determining that the subject possesses the non-wild type RAS gene, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

58. A method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses an elevated level of PLA2G7 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the elevated level of PLA2G7 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

59. A method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a decreased level of PLA2G7 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the decreased level of PLA2G7 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7. A method of treating a cancer in a subject in need thereof, the method comprising: a) determining that the cancer in the subject possesses a cell line in which PLA2G7 inhibits ferroptotic cell death; and b) based on the determining that the cancer in the subject possesses a cell line in which PLA2G7 inhibits ferroptotic cell death, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7. A method of treating a condition in a tissue in a subject in need thereof, the method comprising: a) determining that the tissue suffers from oxidative stress; and b) based on the determining that the tissue suffers from oxidative stress, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7. A method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject exhibits a marker associated with the condition, wherein the condition is associated with PLA2G7 activity; and b) based on the determining that the subject exhibits the marker associated with the condition, administering to the subject a therapeutically- effective amount of a compound that inhibits PLA2G7. A method of treating a condition in a subject in need thereof, the method comprising: a) determining based on an assay that the condition is amenable to treatment by a therapeutic agent that synergizes with a compound that inhibits PLA2G7 for the condition; and b) based on the determining that the condition is amenable to treatment by the therapeutic agent that synergizes with the compound that inhibits PLA2G7 for the condition, administering to the subject a therapeutically- effective amount of a compound that inhibits PLA2G7.

64. The method of any one of claims 57-63 wherein the compound that inhibits PLA2G7 is darapladib.

65. The method of any one of claims 57-63, wherein the compound that inhibits PLA2G7 is rilapladib.

66. The method of any one of claims 57-63, wherein the compound that inhibits PLA2G7 is AA39-2.

67. The method of any one of claims 57-63, wherein the compound that inhibits PLA2G7 is ML256.

68. The method of any one of claims 57-67, wherein the compound that inhibits PLA2G7 inhibits PAFAH2.

69. A method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a non -wild type RAS gene; and b) based on the determining that the subject possesses the non-wild type RAS gene, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2.

70. A method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses an elevated level of PAFAH2 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the elevated level of PAFAH2 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2.

71. A method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a decreased level of PAFAH2 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the decreased level of PAFAH2 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2. A method of treating a cancer in a subject in need thereof, the method comprising: a) determining that the cancer in the subject possesses a cell line in which PAFAH2 inhibits ferroptotic cell death; and b) based on the determining that the cancer in the subject possesses a cell line in which PAFAH2 inhibits ferroptotic cell death, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2. A method of treating a condition in a tissue in a subject in need thereof, the method comprising: a) determining that the tissue suffers from oxidative stress; and b) based on the determining that the tissue suffers from oxidative stress, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2. A method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject exhibits a marker associated with the condition, wherein the condition is associated with PAFAH2 activity; and b) based on the determining that the subject exhibits the marker associated with the condition, administering to the subject a therapeutically- effective amount of a compound that inhibits PAFAH2. A method of treating a condition in a subject in need thereof, the method comprising: a) determining based on an assay that the condition is amenable to treatment by a therapeutic agent that synergizes with a compound that inhibits PAFAH2 for the condition; and b) based on the determining that the condition is amenable to treatment by the therapeutic agent that synergizes with the compound that inhibits PAFAH2 for the condition, administering to the subject a therapeutically- effective amount of a compound that inhibits PAFAH2. The method of any one of claims 69-75, wherein the compound that inhibits PAFAH2 is darapladib. The method of any one of claims 69-75, wherein the compound that inhibits PAFAH2 is rilapladib. The method of any one of claims 69-75, wherein the compound that inhibits PAFAH2 is AA39-2. The method of any one of claims 69-75, wherein the compound that inhibits PAFAH2 is ML256. The method of any one of claims 69-75, wherein the compound that inhibits PAFAH2 inhibits PLA2G7. The method of any one of claims 57-80, further comprising administering to the subject a therapeutically-effective amount of a second therapy. The method of claim 81, wherein the second therapy exhibits synergy with the compound that inhibits PLA2G7. The method of claim 81, wherein the second therapy exhibits synergy with the compound that inhibits PAFAH2. The method of claim 81, wherein the second therapy is a therapeutically-effective amount of a ferroptosis inducer. The method of claim 84, wherein the ferroptosis inducer is a cystine-glutamate system Xc- inhibitor. The method of claim 84, wherein the ferroptosis inducer is a glutathione peroxidase (GPX) inhibitor. The method of claim 84, wherein the second therapy is a therapeutically-effective amount of a modulator of oxidative stress. A method of treating a cancer in a subject in need thereof, the method comprising: a) determining that the cancer in the subject possesses a cell line in which PLA2G7 inhibits ferroptotic cell death; and b) based on the determining that the cancer in the subject possesses a cell line in which PLA2G7 inhibits ferroptotic cell death, administering to the subject a therapeutically-effective amount of a compound that operates by a mechanism of action that synergizes with ferroptotic cell death. The method of claim 88, wherein the compound that synergizes with ferroptotic cell death is a compound that inhibits PLA2G7. A method of treating a cancer in a subject in need thereof, the method comprising: a) determining that the cancer in the subject possesses a cell line in which PAFAH2 inhibits ferroptotic cell death; and b) based on the determining that the cancer in the subject possesses a cell line in which PAFAH2 inhibits ferroptotic cell death, administering to the subject a therapeutically-effective amount of a compound that operates by a mechanism of action that synergizes with ferroptotic cell death. The method of claim 90, wherein the compound that synergizes with ferroptotic cell death is a compound that inhibits PAFAH2. The method of any one of claims 88-91, further comprising administering to the subject a therapeutically-effective amount of a second therapy. The method of claim 92, wherein the second therapy exhibits synergy with the compound that inhibits PLA2G7. The method of claim 92, wherein the second therapy exhibits synergy with the compound that inhibits PAFAH2. The method of claim 92, wherein the second therapy is a therapeutically-effective amount of a ferroptosis inducer. The method of claim 95, wherein the ferroptosis inducer is a cystine-glutamate system Xc- inhibitor. The method of claim 95, wherein the ferroptosis inducer is a glutathione peroxidase (GPX) inhibitor. The method of claim 92, wherein the second therapy is a therapeutically-effective amount of a modulator of oxidative stress. A method comprising: a) determining that a subject exhibits a level of a biomarker, wherein the subject is undergoing a therapeutic regimen of a compound that inhibits PLA2G7; and b) based on the level of the biomarker, determining whether to continue the therapeutic regimen of a compound that inhibits PLA2G7. The method of claim 99, wherein the biomarker is Lp-PLA2. A method compri sing : a) determining that a subject exhibits a level of a biomarker, wherein the subject is undergoing a therapeutic regimen of a compound that inhibits PAFAH2; and b) based on the level of the biomarker, determining whether to continue the therapeutic regimen of a compound that inhibits PAFAH2. A method comprising determining based on an assay of a subject that the subject has at least a threshold number of genes, wherein the threshold number of genes indicates that a compound that inhibits PLA2G7 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of genes is five. A method comprising determining based on an assay of a subject that the subject has at least a threshold number of genes, wherein the threshold number of genes indicates that a compound that inhibits PAFAH2 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of genes is five. A method comprising determining based on an assay of a subject that the subject expresses at least a threshold number of phospholipases, wherein the threshold number of phospholipases indicates that a compound that inhibits PLA2G7 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of phospholipases is five. A method comprising determining based on an assay of a subject that the subject expresses at least a threshold number of phospholipases, wherein the threshold number of phospholipases indicates that a compound that inhibits PAFAH2 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of phospholipases is five.

Description:
TARGETING PHOSPHOLIPASE A2 (PLA2) IN CANCER

CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application claims the benefit of U.S. Provisional Application No. 63/414,336 filed on October 7, 2022, which is incorporated herein by reference in its entirety.

U.S. GOVERNMENT SUPPORT

[0002] This invention was made with government support under 1752506 awarded by the National Science Foundation. The government has certain rights in the invention.

BACKGROUND

[0003] The phospholipase A2 (PLA2) superfamily comprises a group of lipolytic enzymes. Phospholipases belonging to Group VII are referred to as platelet-activating factor acetylhydrolases (PAF-AHs). There are 4 enzymes belonging to Group VII, which include (1) the gene PLA2G7 which encodes an enzyme known as plasma PAF-AH (plateletactivating factor acetylhydrolases), Lp-PLA2 (lipoprotein-associated PLA2) or GVIIA PLA2 (Group VIIA PLA2) and (2) the gene PAFAH2, which encodes an enzyme known as PAF- AH(II) (plasma-activating factor type II), PLA2G7B, or GVIIB PLA2 (Group VIIB PLA2). PLA2G7 and PA FA H 2 are associated with oncogenic processes through mechanisms associated with metabolic alterations.

INCORPORATION BY REFERENCE

[0004] 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. Definitions that are contained in text incorporated by reference are excluded to the extent that they contradict definitions in this disclosure.

SUMMARY

[0005] Disclosed herein is a method of treating a condition, the method comprising administering to a subject in need thereof a therapeutically-effective amount of a compound that inhibits PLA2G7 and a reduced amount of a second therapy, wherein the reduced amount of the second therapy is therapeutically effective for treating the condition in combination with the therapeutically-effective amount of the compound that inhibits PLA2G7, and wherein the reduced amount of the second therapy is less than an amount of the second therapy that is therapeutically effective for the condition in absence of the therapeutically-effective amount of the compound that inhibits PLA2G7.

[0006] Disclosed herein is a method of treating a condition, the method comprising administering to a subject in need thereof a therapeutically-effective amount of a compound that inhibits PAFAH2 and a reduced amount of a second therapy, wherein the reduced amount of the second therapy is therapeutically effective for treating the condition in combination with the therapeutically-effective amount of the compound that inhibits PAFAH2, and wherein the reduced amount of the second therapy is less than an amount of the second therapy that is therapeutically effective for the condition in absence of the therapeutically-effective amount of the compound that inhibits PAFAH2.

[0007] Disclosed herein is a method of treating a condition, the method comprising administering to a subject in need thereof a therapeutically-effective amount of a first compound that inhibits PLA2G7 and a therapeutically-effective amount of a second compound that exhibits synergy with the first compound that inhibits PLA2G7 for the condition.

[0008] Disclosed herein is a method of treating a condition, the method comprising administering to a subject in need thereof a therapeutically-effective amount of a first compound that inhibits PAFAH2 and a therapeutically-effective amount of a second compound that exhibits synergy with the first compound that inhibits PAFAH2 for the condition.

[0009] Disclosed herein is a method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a non-wild type RAS gene; and b) based on the determining that the subject possesses the non-wild type RAS gene, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2.

[0010] Disclosed herein is a method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a non-wild type RAS gene; and b) based on the determining that the subject possesses the non-wild type RAS gene, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0011] Disclosed herein is a method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a non-wild type RAS gene; and b) based on the determining that the subject possesses the non-wild type RAS gene, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2.

[0012] Disclosed herein is a method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a non-wild type STK11 gene; and b) based on the determining that the subject possesses the non-wild type STK11 gene, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0013] Disclosed herein is a method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a non-wild type STK11 gene; and b) based on the determining that the subject possesses the non-wild type STK11 gene, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2.

[0014] Disclosed herein is a method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a non-wild type KEAP1 gene; and b) based on the determining that the subject possesses the non-wild type KEAP1 gene, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0015] Disclosed herein is a method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a non-wild type KEAP1 gene; and b) based on the determining that the subject possesses the non-wild type KEAP1 gene, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2.

[0016] Disclosed herein is a method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses an elevated level of PLA2G7 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the elevated level of PLA2G7 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0017] Disclosed herein is a method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses an elevated level of PAFAH2 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the elevated level of PAFAH2 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2. [0018] Disclosed herein is a method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a decreased level of PLA2G7 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the lowered level of PLA2G7 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0019] Disclosed herein is a method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a decreased level of PAFAH2 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the lowered level of PAFAH2 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2.

[0020] Disclosed herein is a method of treating a cancer in a subject in need thereof, the method comprising: a) determining that the cancer in the subject possesses a cell line in which PLA2G7 inhibits ferroptotic cell death; and b) based on the determining that the cancer in the subject possesses a cell line in which PLA2G7 inhibits ferroptotic cell death, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0021] Disclosed herein is a method of treating a cancer in a subject in need thereof, the method comprising: a) determining that the cancer in the subject possesses a cell line in which PAFAH2 inhibits ferroptotic cell death; and b) based on the determining that the cancer in the subject possesses a cell line in which PAFAH2 inhibits ferroptotic cell death, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2.

[0022] Disclosed herein is a method of treating a condition in a tissue in a subject in need thereof, the method comprising: a) determining that the tissue suffers from oxidative stress; and b) based on the determining that the tissue suffers from oxidative stress, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0023] Disclosed herein is a method of treating a condition in a tissue in a subject in need thereof, the method comprising: a) determining that the tissue suffers from oxidative stress; and b) based on the determining that the tissue suffers from oxidative stress, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2.

[0024] Disclosed herein is a method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject exhibits a marker associated with the condition, wherein the condition is associated with PLA2G7 activity; and b) based on the determining that the subject exhibits the marker associated with the condition, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7. [0025] Disclosed herein is a method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject exhibits a marker associated with the condition, wherein the condition is associated with PAFAH2 activity; and b) based on the determining that the subject exhibits the marker associated with the condition, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2. [0026] Disclosed herein is a method of treating a condition in a subject in need thereof, the method comprising: a) determining based on an assay that the condition is amenable to treatment by a therapeutic agent that synergizes with a compound that inhibits PLA2G7 for the condition; and b) based on the determining that the condition is amenable to treatment by the therapeutic agent that synergizes with the compound that inhibits PLA2G7 for the condition, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0027] Disclosed herein is a method of treating a condition in a subject in need thereof, the method comprising: a) determining based on an assay that the condition is amenable to treatment by a therapeutic agent that synergizes with a compound that inhibits PAFAH2 for the condition; and b) based on the determining that the condition is amenable to treatment by the therapeutic agent that synergizes with the compound that inhibits PAFAH2 for the condition, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2.

[0028] Disclosed herein is a method of treating a cancer in a subject in need thereof, the method comprising: a) determining that the cancer in the subject possesses a cell type in which PLA2G7 inhibits ferroptotic cell death; and b) based on the determining that the cancer in the subject possesses a cell line in which PLA2G7 inhibits ferroptotic cell death, administering to the subject a therapeutically-effective amount of a compound that operates by a mechanism of action that synergizes with ferroptotic cell death.

[0029] Disclosed herein is a method of treating a cancer in a subject in need thereof, the method comprising: a) determining that the cancer in the subject possesses a cell line in which PAFAH2 inhibits ferroptotic cell death; and b) based on the determining that the cancer in the subject possesses a cell line in which PAFAH2 inhibits ferroptotic cell death, administering to the subject a therapeutically-effective amount of a compound that operates by a mechanism of action that synergizes with ferroptotic cell death. [0030] Disclosed herein is a method comprising: a) determining that a subject exhibits a level of a biomarker, wherein the subject is undergoing a therapeutic regimen of a compound that inhibits PLA2G7; and b) based on the level of the biomarker, determining whether to continue the therapeutic regimen of a compound that inhibits PLA2G7.

[0031] Disclosed herein is a method comprising: a) determining that a subject exhibits a level of a biomarker, wherein the subject is undergoing a therapeutic regimen of a compound that inhibits PAFAH2; and b) based on the level of the biomarker, determining whether to continue the therapeutic regimen of a compound that inhibits PAFAH2.

[0032] Disclosed herein is a method comprising determining based on an assay of a subject that the subject has at least a threshold number of genes, wherein the threshold number of genes indicates that a compound that inhibits PLA2G7 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of genes is five. [0033] Disclosed herein is a method comprising determining based on an assay of a subject that the subject has at least a threshold number of genes, wherein the threshold number of genes indicates that a compound that inhibits PAFAH2 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of genes is five. [0034] Disclosed herein is a method comprising determining based on an assay of a subject that the subject expresses at least a threshold number of phospholipases, wherein the threshold number of phospholipases indicates that a compound that inhibits PLA2G7 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of phospholipases is five.

[0035] Disclosed herein is a method comprising determining based on an assay of a subject that the subject expresses at least a threshold number of phospholipases, wherein the threshold number of phospholipases indicates that a compound that inhibits PAFAH2 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of phospholipases is five.

BRIEF DESCRIPTION OF THE DRAWINGS

[0036] FIG. 1 shows PLA2G7 overexpression in human cancer tissues (right) compared to normal samples (left) for tumor samples representing patients with Bladder Urothelial Carcinoma (BLCA), Breast invasive carcinoma (BRCA), Cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC), Cholangiocarcinoma (CHOL), Colon adenocarcinoma (COAD), Esophageal carcinoma (ESCA), Glioblastoma multiforme (GBM), Head and Neck squamous cell carcinoma (HNSC), Kidney Chromophobe (KICH), Kidney renal clear cell carcinoma (KIRC), Kidney renal papillary cell carcinoma (KIRP), Liver hepatocellular carcinoma (LIHC), Lung adenocarcinoma (LU AD), Lung squamous cell carcinoma (LUSC), Pancreatic adenocarcinoma (PAAD), Prostate adenocarcinoma (PRAD), Pheochromocytoma and Paraganglioma (PCPG), Rectum adenocarcinoma (READ), Sarcoma (SARC), Thyroid carcinoma (THCA), Thymoma (THYM), Stomach adenocarcinoma (STAD), and Uterine Corpus Endometrial Carcinoma (UCEC). Box represents upper and lower quartile; bar represents maximum and minimum value; horizonal line represents median.

[0037] FIG. 2 shows PAFAH2 overexpressed in human cancer tissues compared to normal samples. Box represents upper and lower quartile; bar represents maximum and minimum value; horizonal line represents median.

[0038] FIG. 3 shows that multiple genes correlate with PLA2G7 expression in various tumor samples.

[0039] FIG. 4 shows statistically significant correlations between PLA2G7 expression and in individual COAD tumor samples.

[0040] FIG. 5 shows statistically significant correlations between PLA2G7 expression and in individual BRCA tumor samples.

[0041] FIG. 6 shows statistically significant correlations between PLA2G7 expression and in individual DLBCL tumor samples.

[0042] FIG. 7 shows statistically significant correlations between PLA2G7 expression and in individual TGCT tumor samples.

[0043] FIG. 8 shows Western Blot analysis of expression of PLA2G7A and PLA2G7B in cell lines under normal conditions, and under oxidative stress induced via hydrogen peroxide.

[0044] FIG. 9 shows a plot of PLA2G7A and PLA2G7B expression levels in cells under normal conditions.

[0045] FIG. 10 shows a plot of PLA2G7A expression under normal conditions and under oxidative stress, with linear fit.

[0046] FIG. 11 shows a plot of PLA2G7B expression under normal conditions and under oxidative stress.

[0047] FIG. 12 shows a Volcano plot of RNA-seq data obtained from iKRas MEFS following KRAS expression, with Group 7 phospholipases.

[0048] FIG. 13 shows RNA expression of Group 7 phospholipases. Statistical analysis indicated overexpression of PLA2G4A, PNPLA2, PNPLA6, PLA2G6 and PLA2G7. [0049] FIG. 14 shows cell viability as a function of Darapladib concentration.

[0050] FIG. 15 shows a FACS histogram of Liperfluo intensity in iKRas cells in control media, after treatment with Darapladib and after siRNA silencing of PLA2G7.

[0051] FIG. 16 shows a statistically significant increase in Liperfluo intensity following treatment with Darapladib.

[0052] FIG. 17 shows a plot of Liperfluo intensity following Darapladib treatment against the baseline intensity for each cell line, with linear fit.

[0053] FIG. 18 shows viability of KP -Lung cells as a function of Darapladib concentration, in control media and media supplemented with H2O2.

[0054] FIG. 19 shows a heatmap of cell viability as a function of titrating both Darapladib and H2O2 concentration in KP-Lung cells.

[0055] FIG. 20 shows cytotoxicity of iKRas cells after PLA2G7 silencing under normal conditions and under oxidative stress induced by H2O2.

[0056] FIG. 21 shows a plot of cell sensitivity to peroxide as a function of PLA2G7 expression level across a panel of cell lines. Peroxide sensitivity is defined as total cell death, as measured via lactate dehydrogenase (LDH) release assay performed using the manufacturers instructions (ThermoFisher C20300). Expression levels of PLA2G7 were quantified from immunoblots.

[0057] FIG. 22 shows a plot of cell sensitivity to peroxide as a function of PAFAH2 expression level across a panel of cell lines. Peroxide sensitivity is defined as total cell death, as measured via lactate dehydrogenase (LDH) release assay. Expression levels of PAFAH2 were quantified from immunoblots.

[0058] FIG. 23 shows cytotoxicity in various cell lines following treatment by H2O2, Erastin, and H2O2 with Erastin.

[0059] FIG. 24 shows cytotoxicity on various cells lines following silencing of PLA2G7 by siRNA, treatment with Erastin, and following the combination of silencing of PLA2G7 by siRNA and treatment with Erastin.

[0060] FIG. 25 shows statistically significant dependence of sensitivity to Erastin on expression level of PLA2G7A.

[0061] FIG. 26 shows heatmap plots of PANC-1 viability when treated with a combination of Darapladib and Erastin.

[0062] FIG. 27 shows heatmap plots of PANC-1 viability when treated with a combination of Darapladib with FIN56. [0063] FIG. 28 shows heatmap plots of PANC-1 viability when treated with a combination of Darapladib with VLX600.

[0064] FIG. 29 shows heatmap plots of PANC-1 viability when treated with a combination of Darapladib with Sorafenib.

[0065] FIG. 30 shows heatmap plots of PANC-1 viability when treated with a combination of Darapladib with Omavexalon.

[0066] FIG. 31 shows heatmap plots of PANC-1 viability when treated with a combination of Darapladib with ML385.

[0067] FIG. 32 shows heatmap plots of PANC-1 viability when treated with a combination of Darapladib with Rucaparib.

[0068] FIG. 33 shows heatmap plots of PANC-1 viability when treated with a combination of Darapladib with H2O2.

[0069] FIG. 34 shows viability of various cells lines as a function of Darapladib concentration.

[0070] FIG. 35 shows viability of various cells lines as a function of RSL-3 concentration.

[0071] FIG. 36 shows viability of various cells lines as a function of Darapladib concentration in the presence of RSL-3.

[0072] FIG. 37 shows viability of PANC-1 cells as a function of Darapladib, RSL-3, Darapladib and RSL3, Darapladib and Ferrostatin, and Darapladib and RSL3 and Ferrostatin.

[0073] FIG. 38 shows viability of A549 cells as a function of Darapladib, RSL-3, Darapladib and RSL3, Darapladib and Ferrostatin, and Darapladib and RSL3 and Ferrostatin.

[0074] FIG. 39 shows upstream and downstream protein expression following treatment of A549 cells as a function of Darapladib, RSL-3, Darapladib and RSL3, Darapladib and Ferrostatin, and Darapladib and RSL3 and Ferrostatin.

[0075] FIG. 40 show heatmap plots for A549 cells, showing synergy between Darapladib and RSL-3, and rescue via the addition of 1 pM Ferrostatin.

[0076] FIG. 41 show heatmap plots for CAP AN- 1 cells, showing synergy between Darapladib and RSL-3, and rescue via the addition of 1 pM Ferrostatin.

[0077] FIG. 42 show heatmap plots for SUDHL-1 cells, showing synergy between Darapladib and RSL-3, and rescue via the addition of 1 pM Ferrostatin.

[0078] FIG. 43 show heatmap plots for PANC-1 cells, showing synergy between Darapladib and RSL-3, and rescue via the addition of 1 pM Ferrostatin. [0079] FIG. 44 show heatmap plots for CAP AN-2 cells, showing synergy between Darapladib and RSL-3, and rescue via the addition of 1 pM Ferrostatin.

[0080] FIG. 45 show heatmap plots for KARPAS-422 cells, showing synergy between Darapladib and RSL-3, and rescue via the addition of 1 pM Ferrostatin.

[0081] FIG. 46 show heatmap plots for KP Lung cells, showing synergy between Darapladib and RSL-3, and rescue via the addition of 1 pM Ferrostatin.

[0082] FIG. 47 show heatmap plots for MIA-PA-CA cells, showing synergy between Darapladib and RSL-3, and rescue via the addition of 1 pM Ferrostatin.

[0083] FIG. 48 show heatmap plots for U937 cells, showing synergy between Darapladib and RSL-3, and rescue via the addition of 1 pM Ferrostatin.

[0084] FIG. 49 shows cytotoxic synergy of a Darapladib with Gemcitabine combination on various cell lines.

[0085] FIG. 50 shows cytotoxic synergy of a Darapladib, Gemcitabine and the combination of Darapladib with Gemcitabine on A549 cells.

[0086] FIG. 51 shows cytotoxic synergy of a Darapladib, Gemcitabine and the combination of Darapladib with Gemcitabine on Panc-1 cells.

[0087] FIG. 52 shows cytotoxic synergy of Darapladib with chemotherapeutics on SF268 cells.

[0088] FIG. 53 shows cytotoxic synergy of Darapladib with chemotherapeutics on OV8 cells.

[0089] FIG. 54 shows cytotoxic synergy of Darapladib with chemotherapeutics on PC3 cells.

[0090] FIG. 55 shows immunoblots of SF268 and Panel cells. Cell lines were treated with lipofectamine and siRNA to deliver interfering RNA to knock down the targets listed across the top of the gel.

[0091] FIG. 56 shows immunoblots of SF268 and PANC-1 cells lines treated with additional Fenton substrates in media (-F) (top panel), or with added Fenton and polyunsaturated fatty acids F(PUFA); and immunoblots following the addition of PUFA substrates in the absence of a Fenton reagent (bottom panel).

[0092] FIG. 57 shows cell death following Rilapladib, RSL-3, combination of Rilapladib and RSL-3, and rescue via the addition of Ferrostatin in SF268 cells.

[0093] FIG. 58 shows cell death following Rilapladib, RSL-3, combination of Rilapladib and RSL-3, and rescue via the addition of Ferrostatin in A549 cells. [0094] FIG. 59 shows cell death following Rilapladib, RSL-3, combination of Rilapladib and RSL-3, and rescue via the addition of Ferrostatin in PANC-1 cells.

[0095] FIG. 60 shows immunoblots of PLA2G7 expression level by NRF2 and other antioxidant and lipid metabolic targets including fatty acid metabolism in iKRas cells. [0096] FIG. 61 shows immunoblots of PLA2G7 expression level by NRF2 and other antioxidant and lipid metabolic targets including fatty acid metabolism in A549 cells

DETAILED DESCRIPTION

[0097] The following terms are used throughout as defined below.

[0098] As used herein and in the appended claims, singular articles such as “a” and “an” and “the” and similar referents in the context of describing the elements (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the embodiments and does not pose a limitation on the scope of the claims unless otherwise stated. No language in the specification should be construed as indicating any non-claimed element as essential.

[0099] As used herein, “about” will be understood by persons of ordinary skill in the art and will vary to some extent depending upon the context in which it is used. If there are uses of the term which are not clear to persons of ordinary skill in the art, given the context in which it is used, “about” will mean up to plus or minus 10% of the particular term - for example, “about 10 wt.%” would mean “9 wt.% to 11 wt.%.” It is to be understood that when “about” precedes a term, the term is to be construed as disclosing “about” the term as well as the term without modification by “about” - for example, “about 10 wt.%” discloses “9 wt.% to 11 wt.%” as well as disclosing “10 wt.%.”

[0100] The phrase “and/or” as used in the present disclosure will be understood to mean any one of the recited members individually or a combination of any two or more thereof - for example, “A, B, and/or C” would mean “A, B, C, A and B, A and C, B and C, or the combination of A, B, and C.” [0101] As will be understood by one skilled in the art, for any and all purposes, particularly in terms of providing a written description, all ranges disclosed herein also encompass any and all possible subranges and combinations of subranges thereof. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as “up to,” “at least,” “greater than,” “less than,” and the like include the number recited and refer to ranges which can be subsequently broken down into subranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. Thus, for example, a group having 1-3 atoms refers to groups having 1, 2, or 3 atoms. Similarly, a group having 1-5 atoms refers to groups having 1, 2, 3, 4, or 5 atoms, and so forth.

[0102] Those of skill in the art will appreciate that compounds as disclosed in regard to the present technology may exhibit the phenomena of tautomerism, conformational isomerism, geometric isomerism and/or stereoisomerism. As the formula drawings within the specification and claims can represent only one of the possible tautomeric, conformational isomeric, stereochemical or geometric isomeric forms, it should be understood that the present technology encompasses any tautomeric, conformational isomeric, stereochemical and/or geometric isomeric forms of the compounds having one or more of the utilities described herein, as well as mixtures of these various different forms.

[0103] “ Tautomers” refers to isomeric forms of a compound that are in equilibrium with each other. The presence and concentrations of the isomeric forms will depend on the environment the compound is found in and may be different depending upon, for example, whether the compound is a solid or is in an organic or aqueous solution. For example, in aqueous solution, quinazolinones may exhibit the following isomeric forms, which are referred to as tautomers of each other:

As another example, guanidines may exhibit the following isomeric forms in protic organic solution, also referred to as tautomers of each other:

Because of the limits of representing compounds by structural formulas, it is to be understood that all chemical formulas of the compounds described herein represent all tautomeric forms of compounds and are within the scope of the present technology.

[0104] Stereoisomers of compounds include all chiral, diastereomeric, and racemic forms of a structure, unless the specific stereochemistry is expressly indicated. Thus, compounds used in the present technology include enriched or resolved optical isomers at any or all asymmetric atoms as are apparent from the depictions. Both racemic and diastereomeric mixtures, as well as the individual optical isomers can be isolated or synthesized so as to be substantially free of their enantiomeric or diastereomeric partners, and these stereoisomers are all within the scope of the present technology.

A. Introduction

[0105] Current methods of treating cancers are often hindered by the inevitable development of acquired resistance against systemic therapy. Accordingly, herein are described methods comprising administering a therapeutically-effective amount of a compound that inhibits PLA2G7. The present disclosure includes methods for treating a condition and methods of treating cancer.

Group 7 Phospholipases

[0106] Phospholipases are a group of enzymes that hydrolyze phospholipids into fatty acids and other lipophilic molecules. The phospholipase A2 (PLA2) family comprises a group of lipolytic enzymes that hydrolyze the sn-2 position of glycerophospholipids to fatty acids and lysophospholipids. The mammalian genome encodes more than 30 PLA2S or related enzymes. Several enzymes show PLA2 activity but are not named PLA2. The PLA2 reaction is important for signal transduction as polyunsaturated fatty acids (e.g., arachidonic acid) and lysophospholipids released by PLA2 can be converted into a wide variety of bioactive lipids referred to as lipid mediators, which exert multiple biological effects.

[0107] Phospholipase A2 Group VII (PLA2G7) gene is a member of the phospholipase A2 (PLA2) family that encodes an enzyme known as plasma PAF-AH (platelet-activating factor acetylhydrolases), Lp-PLA2 (lipoprotein-associated PLA2), PAF-AH VIIA, and GVIIA PLA 2 (Group VIIA PLA 2 ).

[0108] Platelet-activating factor acetylhydrolase 2 is an enzyme that is encoded by the PAFAH2 gene. The PAFAH2 gene is a member of the phospholipase A2 (PLA2) family that encodes PLA2G7B / PAF-AH(II), PAF-AH VIIB (type II), and Group VIIB PLA2.

[0109] As described herein, members of the mammalian phospholipase A2 family are associated with the ferroptosis defense pathway. Non-limiting examples of members of the mammalian phospholipase A2 family associated with the ferroptosis defense pathway include: platelet-activating factor acetylhydrolases (PAF-AHs, such as PLA2G7, PAFAH2, PAFAH1B2 and PAFAH1B3); Ca2+ independent PLA2s (iPLA2s, such as PNPLA1, PNPLA2, PNPLA3, PNPLA4, PNPLA5, PNPLA6, PNPLA7, PNPLA8 and PLA2G6); and cytosolic PLA2s (cPLA2s, such as PLA2G4A, PLA2G4B, PLA2G4C, PLA2G4D, PLA2G4E and PLA2G4F); ; secreted PLA2s (sPLA2s, such as PLA2G1B, PLA2G2A, PLA2G2C, PLA2G2D, PLA2G2E, PLA2G2F, PLA2G3, PLA2G5, PLA2G10, PLA2G12A and PLA2G12B), lysosomal PLA2 PLA2G15, PLA/acyltransferases (such as HRALSL1, HRASLS2, and PLATG16), and a/ hydrolases (such as ABHD3, ABHD4, ABHD5, ABHD6, ABHD12 and ABHD16A.)

Phospholipase A2 Group VII (PLA2G7)

[0110] PLA2G7 is a protein coding gene. The protein encoded by the PLA2G7 gene is a secreted enzyme that catalyzes the degradation of platelet-activating factor to biologically- inactive products. Two transcript variants encoding the same protein exist for the PLA2G7 gene. A paralogue of the PLA2G7 gene is PAFAH2.

[0111] Non-limiting examples of pathways associated with PLA2G7 include lipid metabolism in senescent cells, caloric restriction-induced immunometabolic effects in humans, phospholipid catabolism during inflammatory and oxidative stress response, and modulating the effect of IL1 on megakaryocytes in obesity.

[0112] Non-limiting examples of conditions associated with PLA2G7 or defects in the PLA2G7 gene include Platelet-Activating Factor Acetylhydrolase Deficiency and Ige Responsiveness, Atopic. Among the related pathways are Phospholipase-C Pathway and IL1 and megakaryocytes in obesity. Gene Ontology (GO) annotations related to this gene include phospholipid binding and calcium-independent phospholipase A2 activity. [0113] In some embodiments, a compound that inhibits PLA2G7 is a Lipoprotein- Associated Phospholipase A2 (Lp-PLA2) inhibitor. In some embodiments, a compound that inhibits PLA2G7 is a PAFAH2 inhibitor.

[0114] Non-limiting examples of compounds that inhibit PLA2G7 include: darapladib, rilapladib, AA39-2 (also referred to as ML225), ML256, and a pharmaceutically-acceptable salt of any of the foregoing. In some embodiments, a compound that inhibits PLA2G7 is darapladib. In some embodiments, a compound that inhibits PLA2G7 is rilapladib. In some embodiments, a compound that inhibits PLA2G7 is AA39-2. In some embodiments, a compound that inhibits PLA2G7 is ML256.

[0115] A therapeutically-effective amount of darapladib can be a dose based on the body mass of the subject, for example, about 0.5 mg/kg, about 1 mg/kg, about 1.25 mg/kg, about 1.5 mg/kg, about 1.75 mg/kg, about 2 mg/kg, about 2.25 mg/kg, about 2.5 mg/kg, about 2.75 mg/kg, about 3 mg/kg, about 3.25 mg/kg, about 3.5 mg/kg, about 3.75 mg/kg, about 4 mg/kg, about 4.25 mg/kg, about 4.5 mg/kg, about 4.75 mg/kg, or about 5 mg/kg. In some embodiments, darapladib is administered to the subject at a dose of about 0.813 mg/kg.

[0116] A therapeutically-effective amount of a compound described herein can be present in a composition described herein at a mass of, for example, about 1 mg, about 5 mg, about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, about 95 mg, about 100 mg, about 105 mg, about 110 mg, about 115 mg, about 120 mg, about 125 mg, about 130 mg, about 135 mg, about 140 mg, about 145 mg, about 150 mg, about 155 mg, about 160 mg, about 165 mg, about 170 mg, about 175 mg, or about 180 mg. In some embodiments, darapladib is administered to the subject at a dose of about 160 mg. In some embodiments, darapladib is administered to the subject at a dose of about 48.8 mg. In some embodiments, darapladib is administered to the subject once daily.

[0117] Non-limiting examples of structural formulas of compounds that inhibit PLA2G7 are provided below:

[0118] In some embodiments, darapladib inhibits intracellular PAFAH2. In some embodiments, darapladib inhibits plasma PLA2G7. In some embodiments, darapladib inhibits LDL-bound PLA2G7. In some embodiments, darapladib inhibits HDL-bound

PLA2G7. In some embodiments, darapladib inhibits both intracellular PAFAH2 and PLA2G7, present in plasma, or LDL-bound or HDL-bound.

[0119] In some embodiments, a compound that inhibit PLA2G7 is a PAF analogue. Nonlimiting examples of structural formulas of PAF analogs are provided below:

[0120] In some embodiments, R represents an acetamide or trifluoroacetamide, Z contains a high polar moiety including a quaternary ammonium group, and a XR1 is a long-chain fatty ether. [0121] In some embodiments, a compound that inhibits PLA2G7 is a pyrimidone derivative. Non-limiting examples of structural formulas of pyrimidone derivatives are provided below:

[0122] In some embodiments, a compound that inhibit PLA2G7 is a darapladib analogue.

Non-limiting examples of structural formulas of darapladib analogues are provided below:

[0123] In some embodiments, a compound that inhibits PLA2G7 is a pyrimidine Lp-PLA2 inhibitor. Non-limiting examples of structural formulas of pyrimidine Lp-PLA2 inhibitors are provided below:

[0124] In some embodiments, a compound that inhibits PLA2G7 is an imidazo[l,2- a]pyrimidine compound. Non-limiting examples of structural formulas of imidazo[l,2- a]pyrimidine compounds are provided below:

[0125] In some embodiments, a compound that inhibits PLA2G7 is a biaryl inhibitor. Nonlimiting examples of structural formulas of biaryl inhibitors are provided below: [0126] In some embodiments, a compound that inhibits PLA2G7 is a lactam inhibitor. Nonlimiting examples of structural formulas of lactam inhibitors are provided below:

[0127] In some embodiments, a compound that inhibits PLA2G7 is a sulfonamide inhibitor. Non-limiting examples of structural formulas of sulfonamide inhibitors are provided below:

[0128] In some embodiments, a compound that inhibits PLA2G7 is a covalent inhibitor. Non-limiting examples of covalent inhibitors include: organophosphorus compounds (e.g., nerve agents such as paraoxon, sarin, soman and tabun), SB-253514, linear carbamate inhibitors (e.g, HT-01), carbamate inhibitors (e.g, JMN4), and monocylic-P-lactams (e.g, SB-222657). Non-limiting examples of structural formulas of covalent inhibitors are provided below:

19

SUBSTITUTE SHEET (RULE 26)

[0129] Non-limiting examples of substrates hydrolyzed by PLA2G7/PAFAH2 include: Platelet activating factors PAFs (e.g., l-hexadecyl-2-acetyl-sn-glycero-3-phosphocholine); Phospholipids with short sn-2 acyl chains (e.g., propionyl (C3) and butyryl (C4) moieties); Phospholipids with oxidatively fragmented fatty acyl chains (e.g., succinoyl (C4 with cocarboxyl), glutaroyl (C5 with co-carboxyl), 5-oxovaleroyl (C4 with co-carbonyl), and azelaoyl (C8 with co-carboxyl) moieties (e.g., l-palmitoyl-2-azelaoyl-snglycero-3- phosphocholine, azelaoyl PC))); Short-chain diacylglycerols, triacylglycerols and acetylated alkanols; Non-fragmented oxidized phospholipids, including F2-iso-prostane-containing phospholipids (e.g., l-palmitoyl-2-(15-F2-isoprostanoyl)-sn-glycero-3-phosphochol ine (F2- isoprostane containing PC); Phospholipid hydroperoxides; and phospholipids with co-3 fatty acid epoxides.

[0130] A non-limiting example of a PLA2G7 serum biomarker is Lp-PLA2.

20

SUBSTITUTE SHEET (RULE 26) [0131] In some embodiments, a method disclosed herein comprises: a) determining that the subject possesses a elevated level of PLA2G7 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the elevated level of PLA2G7 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0132] In some embodiments, a method disclosed herein comprises: a) determining that the subject possesses a decreased level of PLA2G7 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the decreased level of PLA2G7 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0133] In some embodiments, a method disclosed herein comprises: a) determining that the subject possesses an elevated level of a substrate of PLA2G7 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the elevated level of a substrate of PLA2G7 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7. In some embodiments, the elevated level of a substrate of PLA2G7 is determined in a blood sample.

[0134] In some embodiments, a method disclosed herein comprises: a) determining that the subject possesses a decreased level of a substrate of PLA2G7 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the decreased level of a substrate of PLA2G7 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0135] In some embodiments, a method disclosed herein comprises: a) determining that the subject possesses an elevated level of a product of PLA2G7 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the elevated level of a product of PLA2G7 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0136] In some embodiments, a method disclosed herein comprises: a) determining that the subject possesses a decreased level of a product of PLA2G7 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the decreased level of a product of PLA2G7 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0137] In some embodiments, a method disclosed herein comprises detecting in a subject one or more markers. In some embodiments, a method disclosed herein comprises correlating the marker to a condition amenable to treatment by a compound that inhibits PLA2G7.

[0138] In some embodiments, a method disclosed herein comprises determining an aberrant expression pattern of one or more markers. In some embodiments, the aberrant expression comprises increased expression of a marker. In some embodiments, the aberrant expression comprises decreased expression of a marker.

[0139] In some embodiments, a method disclosed herein comprises determining that a subject possesses an elevated level of PLA2G7 expression. In some embodiments, a method disclosed herein comprises determining that a subject possesses an elevated level of PLA2G7 expression in comparison to a predetermined threshold value. In some embodiments, the predetermined threshold value is level of PLA2G7 expression determined from a normal control subject.

[0140] In some embodiments, the elevated level of PLA2G7 expression is at least about 0.5-fold, at least about 1-fold, at least about 1.5-fold, at least about 2 fold, at least about 2.5- fold, at least about 3-fold, at least about 3.5-fold, at least about 4-fold, at least about 4.5- fold, at least about 5-fold, at least about 5.5-fold, at least about 6-fold, at least about 6.5- fold, at least about 7 fold, at least about 7.5-fold, at least about 8-fold, at least about 8.5- fold, at least about 9-fold, at least about 9.5-fold, at least about 10-fold, at least about 10.5- fold, at least about 11-fold, at least about 11.5-fold, at least about 12 fold, at least about 12.5-fold, at least about 13-fold, at least about 13.5-fold, at least about 14-fold, at least about 14.5-fold, at least about 15-fold, or greater than about 15-fold higher in comparison to a predetermined threshold value. In some embodiments, the predetermined threshold value is a level of PLA2G7 expression determined from a normal control subject.

[0141] In some embodiments, the PLA2G7 expression is higher in a subject having bladder urothelial carcinoma (BLCA) compared to expression in a normal control subject. See, e.g., Table 1 below. In some embodiments, the PLA2G7 expression is higher in a subject having liver hepatocellular carcinoma (LH4C) compared to expression in a normal control subject. See, e.g., Table 1 below. In some embodiments, the PLA2G7 expression is higher in a subject having prostate adenocarcinoma (PRAD) compared to expression in a normal control subject. See, e.g., Table 1 below. In some embodiments, the elevated level of PLA2G7 expression is at least about 1.5-fold, at least about 1.6-fold, at least about 1.7-fold, at least about 1.8-fold, at least about 1.9-fold, or at least about 2-fold higher in the subject. [0142] In some embodiments, the PLA2G7 expression is higher in a subject having cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC) compared to expression in a normal control subject. See, e.g., Table 1 below. In some embodiments, the elevated level of PLA2G7 expression is at least about 1-fold, at least about 1.5-fold, at least about 2-fold, at least about 2.5-fold, at least about 3-fold, at least about 3.5-fold, at least about 4-fold, or at least about 4.5-fold higher in the subject.

[0143] In some embodiments, the PLA2G7 expression is higher in a subject having esophageal carcinoma (ESC A) compared to expression in a normal control subject. See, e.g., Table 1 below. In some embodiments, the elevated level of PLA2G7 expression is at least about 3-fold, at least about 3.5-fold, at least about 4-fold, at least about 4.5-fold, at least about 5-fold, at least about 5.5-fold, at least about 6-fold, or at least about 6.5-fold higher in the subject.

[0144] In some embodiments, the PLA2G7 expression is higher in a subject having head and neck squamous cell carcinoma (HNSC) compared to expression in a normal control subject. See, e.g., Table 1 below. In some embodiments, the elevated level of PLA2G7 expression is at least about 1-fold, at least about 2-fold, at least about 3-fold, or at least about 4-fold higher in the subject.

[0145] In some embodiments, the PLA2G7 expression is higher in a subject having kidney chromophobe (KICH) compared to expression in a normal control subject. See, e.g., Table 1 below. In some embodiments, the elevated level of PLA2G7 expression is at least about 3-fold, at least about 3.5-fold, at least about 4-fold, at least about 4.5-fold, at least about 5- fold, at least about 5.5-fold, at least about 6-fold, at least about 6.5-fold, at least about 7- fold, or at least about 7.5-fold higher in the subject.

[0146] In some embodiments, the PLA2G7 expression is higher in a subject having kidney renal clear cell carcinoma (KIRC) compared to expression in a normal control subject. See, e.g., Table 1 below. In some embodiments, the PLA2G7 expression is higher in a subject having kidney renal clear cell carcinoma (KIRC) is at least about 3-fold, at least about 4- fold, at least about 5-fold, at least about 6-fold, at least about 7-fold, at least about 8-fold, or at least about 9-fold higher in the subject.

[0147] In some embodiments, the PLA2G7 expression is higher in a subject having kidney renal papillary cell carcinoma (KIRP) compared to expression in a normal control subject. See, e.g., Table 1 below. In some embodiments, the PLA2G7 expression is higher in a subject having kidney renal papillary cell carcinoma (KIRP) is at least about 5-fold, at least about 10-fold, or at least about 15-fold higher in the subject.

[0148] In some embodiments, the PLA2G7 expression is higher in a subject having stomach adenocarcinoma (STAD) compared to expression in a normal control subject. See, e.g., Table 1 below. In some embodiments, the elevated level of PLA2G7 expression is at least about 1-fold, at least about 1.5-fold, at least about 2-fold, at least about 2.5-fold, or at least about 3 -fold higher in the subject.

Table 1. Exemplary expression values of PLA2G7 across cancers

Intracellular platelet-activating factor acetylhydrolase, type II (PAF-AH (II))

[0149] PAF-AH (II) is a monomeric 40 kDa enzyme that can hydrolyze the acetyl group of PAF (l-O-alkyl-2-acetyl-sn-glycero-3 -phosphocholine), a signaling phospholipid associated with diverse physiological and pathological events such as inflammation, anaphylaxis, reproduction, and fetal development. PAF-AH (II) can also hydrolyze phospholipids with short length or oxidatively modified sn-2 acyl chains.

[0150] The amino acid sequence of human PAF-AH (II) is 392 residues in length and contains a GXSXG motif. A unique feature of PAF-AH(II) is an N-myristoylation signal at the N-terminus (NH2-Met-Gly-X-X-X-Ser-). PAF-AH(II) is myristoylated and distributed in both the cytosol and the membrane fractions as observed with other myristoylated proteins. PAF-AH (II) is inactivated by sulfhydryl agents such as 5,5 '-dithiobis (2- nitrobenzoic acid). This tendency suggests that PAF-AH (II) has a free cysteine residue that is essential for catalysis. PAF-AH (II) is the only phospholipase A2 that is myristoylated at the N-terminus and is evolutionarily conserved from lower organisms to mammals. PAF- AH (II) can function as an antioxidant phospholipase.

[0151] Human PAF-AH (II), encoded by PAFAH2, shows about 43% identity with human plasma PAF-AH. In contrast, the amino acid sequence of PAF-AH (II) has no significant homology with any subunit of PAF-AH (I) or other intracellular PLA2s. PAF-AH (II) lacks the first 50 residues that target the plasma enzyme for secretion.

[0152] Using the crystal structure of plasma PAF-AH as a template, a homology model of PAF-AH (II) shows a classic lipase a/p hydrolase fold and contains a catalytic triad of Ser236, His314, and Asp259. Based on the homology model of PAF-AH (II), in addition to the myristoyl group, a hydrophobic patch consisting of Leu327, Ile328, and Phe331 is involved in the membrane binding of PAF-AH (II).

[0153] The PAFAH2 gene encodes intracellular platelet-activating factor acetylhydrolase, type II (PAF-AH (II)). The amino acid sequence of PAFAH2 comprises significant identity with plasma PAF-AH. A paralog of the PAFAH2 gene is PLA2G7.

[0154] A non-limiting example of a condition associated with the PAFAH2 gene includes Williams-Beuren Syndrome. [0155] In some embodiments, PAFAH2 is bound to a membrane during oxidative stress. [0156] In some embodiments, polyunsaturated fatty acids phospholipid hydroperoxides (PUFA-PL-OOH) accumulate in cell membranes at sufficiently high quantities to cause cell death following treatment with a compound that inhibits PAFAH2.

[0157] In some embodiments, lysosomal lipid content in cancer cells is decreased following treatment with a compound that inhibits PAFAH2. For example, the lysosomal lipid content of cancer cells is decreased following treatment with a compound that inhibits PAFAH2 by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, or at least about 75%.

[0158] In some embodiments, a method disclosed herein comprises detecting in a subject one or more markers. In some embodiments, a method disclosed herein comprises correlating the marker to a condition amenable to treatment by a compound that inhibits PAFAH2.

[0159] In some embodiments, a method disclosed herein comprises determining an aberrant expression pattern of one or more markers. In some embodiments, the aberrant expression is increased expression of a marker. In some embodiments, the aberrant expression is decreased expression of a marker.

[0160] In some embodiments, a method disclosed herein comprises determining that a subject possesses an elevated level of PAFAH2 expression. In some embodiments, a method disclosed herein comprises determining that a subject possesses an elevated level of PAFAH2 expression in comparison to a predetermined threshold value. In some embodiments, the predetermined threshold value is level of PAFAH2 expression determined from a normal control subject.

[0161] In some embodiments, the elevated level of PAFAH2 expression is at least about 0.5-fold, at least about 0.6-fold, at least about 0.7-fold, at least about 0.8-fold, at least about 0.9-fold, at least about 1-fold, at least about 1.1-fold, at least about 1.2-fold, at least about 1.2-fold, at least about 1.4-fold, at least about 1.5-fold, at least about 1.6 fold, at least about 1.7-fold, at least about 1.8-fold, at least about 1.9-fold, at least about 2-fold, or greater than about 2-fold higher in comparison to a predetermined threshold value. In some embodiments, the predetermined threshold value is a level of PAFAH2 expression determined from a normal control subject. [0162] In some embodiments, the PAFAH2 expression is higher in a subject having breast invasive carcinoma (BRCA) compared to expression in a normal control subject. See, e.g., Table 2 below. In some embodiments, the PAFAH2 expression is higher in a subject having cholangiocarcinoma primary tumor (CHOL) compared to expression in a normal control subject. See, e.g., Table 2 below. In some embodiments, the PAFAH2 expression is higher in a subject having Glioblastoma multiforme primary tumor (GBM) compared to expression in a normal control subject. See, e.g., Table 2 below. In some embodiments, the elevated level of PAFAH2 expression is at least about 0.5-fold, at least about 0.6-fold, at least about 0.7-fold, at least about 0.8-fold, at least about 0.9-fold, at least about 1-fold, at least about 1.1-fold, at least about 1.2-fold, at least about 1.3-fold, or at least about 1.4-fold higher in the subject.

Table 2. Exemplary expression values of PAFAH2 across cancers

[0163] In some embodiments, a method disclosed herein comprises determining that a subject possesses a decreased level of PAFAH2 expression. In some embodiments, a method disclosed herein comprises determining that a subject possesses a decreased level of PAFAH2 expression in comparison to a predetermined threshold value. In some embodiments, the predetermined threshold value is level of PAFAH2 expression determined from a normal control subject.

[0164] In some embodiments, the decreased level of PAFAH2 expression is at least about 0.5-fold, at least about 0.6-fold, at least about 0.7-fold, at least about 0.8-fold, at least about 0.9-fold, at least about 1-fold, at least about 1.1-fold, at least about 1.2-fold, at least about 1.2-fold, at least about 1.4-fold, at least about 1.5-fold, at least about 1.6 fold, at least about 1.7-fold, at least about 1.8-fold, at least about 1.9-fold, at least about 2-fold, or greater than about 2-fold lower in comparison to a predetermined threshold value. In some embodiments, the predetermined threshold value is level of PAFAH2 expression determined from a normal control subject.

[0165] In some embodiments, the PAFAH2 expression is decreased in a subject having colon adenocarcinoma (COAD) compared to expression in a normal control subject. See, e.g., Table 3 below. In some embodiments, the PAFAH2 expression is at least about 0.5- fold, at least about 0.6-fold, at least about 0.7-fold lower in the subject.

[0166] In some embodiments, the PAFAH2 expression is decreased in a subject having head and neck squamous cell carcinoma (HNSC) compared to expression in a normal control subject. See, e.g., Table 3 below. In some embodiments, the PAFAH2 expression is decreased in a subject having kidney chromophobe (KICH) compared to expression in a normal control subject. See, e.g., Table 3 below. In some embodiments, the PAFAH2 expression is decreased in a subject having liver hepatocellular carcinoma (LIHC) compared to expression in a normal control subject. See, e.g., Table 3 below. In some embodiments, the PAFAH2 expression is decreased in a subject having lung adenocarcinoma (LU AD) compared to expression in a normal control subject. See, e.g., Table 3 below. In some embodiments, the PAFAH2 expression is decreased in a subject having thyroid carcinoma (THCA) compared to expression in a normal control subject.

See, e.g., Table 3 below. In some embodiments, the decreased level of PAFAH2 expression is at least about 0.5-fold, at least about 0.6-fold, at least about 0.7-fold, at least about 0.8- fold, at least about 0.9-fold, at least about 1-fold, or at least about 1.1-fold lower in the subject.

[0167] In some embodiments, the PAFAH2 expression is decreased in a subject having kidney renal clear cell carcinoma (KIRC) compared to expression in a normal control subject. See, e.g., Table 3 below. In some embodiments, the PAFAH2 expression is decreased in a subject having lung squamous cell carcinoma (LUSC) compared to expression in a normal control subject. See, e.g., Table 3 below. In some embodiments, the PAFAH2 expression is decreased in a subject having rectum adenocarcinoma (READ) compared to expression in a normal control subject. See, e.g., Table 3 below. In some embodiments, the decreased level of PAFAH2 expression is at least about 0.5-fold, at least about 0.6-fold, at least about 0.7-fold, at least about 0.8-fold, at least about 0.9-fold, at least about 1-fold, at least about 1.1-fold, at least about 1.2-fold, or at least about 1.3-fold lower in the subject. Table 3. Exemplary expression values of PAFAH2 across cancers

[0168] Ferroptosis

[0169] Ferroptosis is an iron-dependent form of regulated cell death (RCD) that can be triggered by the toxic build-up of lipid peroxides in cell membranes. This phenomenon is mechanistically distinct from other forms of RCD. The execution of ferroptosis requires iron-catalyzed peroxidation of polyunsaturated fatty acid (PUFA)-containing phospholipids (PUFA-PLs) to generate toxic PUFA phospholipid hydroperoxides (PUFA-PL-OOH) at levels that exceed the buffering capacity of multiple cellular antioxidant mechanisms that form the ferroptosis defense system. This process is controlled by a complex network governing lipid metabolism, ROS biology and iron regulation.

[0170] At the plasma membrane, system x c " imports cystine, which is reduced in the cell to the amino acid cysteine. Cysteine and glutamate are used in the biosynthesis of reduced glutathione. Reduced glutathione is used by GPX4 to reduce lethal reactive polyunsaturated fatty acid phospholipid hydroperoxides (PUFA-PL-OH) to non-reactive and non-lethal PUFA phospholipid alcohols (PUFA-PL-OH). Alternatively, the oxidized PUFA-OOH tail can be cleaved from a phospholipid by the action of iPLA2p/PLA2G6. PUFA-PL-OOHs accumulate in the cellular membrane, the ER, and other organelles.

[0171] PUFA-PLs are oxidized by labile Fe(II) and Fe(II)-dependent enzymes such as ALOXs in conjunction with other proteins. Replacement of PUFA in cell membranes by MUFA (monounsaturated fatty acid) by ACSL3 can suppress ferroptosis by reducing the PUFA-PL substrate. Fe(III) is imported into cells by Tf/TFRl (transferrin/transferrin receptor), after which Fe(III) is reduced to Fe(II) by DMT. Iron is stored as Fe(III) in Ferritin, where Fe(III) is not available to promote ferroptosis. The export of iron in multivesicular bodies via prom2 suppresses ferroptosis. Acetyl-CoA is used to make free PUFAs, which are activated by ACSL4, LPCAT3, and ACSL1 to generate PUFA-PLs. ACSL4 can be phosphorylated by PKCbll for further activation.

[0172] Cancer cells can be vulnerable to ferroptosis due to the cancer cells’ altered lipid metabolism, genetic mutations in either oncogenes or tumor suppressors, or imbalanced ferroptosis defense pathways (e.g, Lei et al 2022 Targeting ferroptosis as a vulnerability in cancer. Nature Reviews Cancer 22:381-396).

[0173] Ferroptosis is a mechanism used by tumors to acquire drug resistance and to evade the immune system in multiple cancers.

[0174] As described herein, PLA2G7 is associated with the ferroptosis defense pathway. For example, inhibition of PLA2G7 induces the ferroptosis defense pathway.

[0175] In some embodiments, PLA2G7 recognizes and hydrolyzes oxidatively damaged and truncated phospholipids.

[0176] In some embodiments, PLA2G7 maintains the redox balance and lipid membrane stability in cells.

[0177] In some embodiments, PLA2G7 modulates the availability of undamaged lipid species that act as substrates of peroxidation. [0178] As described herein, PAFAH2 is associated with the ferroptosis defense pathway. For example, inhibition of PAFAH2 induces cell death via ferroptosis by compromising a ferroptosis defense pathway.

[0179] In some embodiments, PAFAH2 recognizes and hydrolyzes oxidatively damaged and truncated phospholipids. In some embodiments, PAFAH2 maintains the redox balance and lipid membrane stability in cells.

[0180] As described herein, activity of PLA2G7 removes lipid hydroperoxides from cell membranes.

[0181] As described herein, activity of PLA2G7 reduces oxidized phospholipids from cell membranes to prevent ferroptosis.

[0182] As described herein, activity of PLA2G7 removes polyunsaturated fatty acids from cell membranes.

[0183] As described herein, activity of PAFAH2 reduces oxidized phospholipids from cell membranes to prevent ferroptosis.

[0184] Ferroptosis can be induced by increasing the activity of proferroptotic pathways or decreasing the activity of antiferroptotic pathways. Of the four antioxidant defense systems, the most understood pathway is the GPX4-GSH system, in which the GPX4 enzyme with a reduced glutathione (GSH) cofactor convert toxic PUFA-PL-OOH into non-lethal PUFA- PL-OH. As GSH is derived from a reaction in which cysteine is the rate-limiting precursor, targeting cysteine uptake via the SLC7A11 transporter is another strategy for inducing ferroptosis. However, some cancer cell lines remain resistant to ferroptosis.

[0185] Non-limiting examples of compounds, agents, or methods that induce ferroptosis include: a cystine-glutamate system Xc- inhibitor, a glutathione peroxidase (GPX) inhibitor, a SLC7A11 inhibitor (e.g., sulfasalazine), a cysteine depletion, inhibition of glutathione synthesis (e.g. erastin, BSO), an FSP1 inhibitor, a DHODH inhibitor, a GCH1 inhibitor, a PTS inhibitor, an SPR inhibitor, a DHFR inhibitor, BH4 depletion, squalene depletion, iPLA2beta inhibition, TBH, chemerin monoclonal antibody, NFS1 inhibition, frataxin inhibition, CISD2 inhibitor (e.g., pioglitazone), inhibition of squalene synthesis (atorvastatin, zaragozic acid, SQS inhibitors), NEDD4 inhibition, DJI inhibition, PDK4 inhibitor, TYRO3 inhibitor, MT 1G inhibitor, arachidonic acid, oxidized LDL, and combinations thereof.

[0186] Additional non-limiting examples of compounds, agents, or methods that induce ferroptosis include: DNA stress inducers (e.g., Zalcitabine), GCL inhibitors (e.g., Buthionine sulfoximine), GPX4 inhibitors (e.g., Altretamine, Withaferin A, 1S,3R-RSL3, structural analogs of RSL3 ML162, ML210, ), GPX4 degraders (e.g. FIN56), GSH inhibitors (e.g. Cisplatin, BSO, and DPI2), iron activators (e.g., Neratinib, Salinomycin, Lapatinib), SLC7A11 inhibitors (e.g., Sorafenib, Sulfasalazine, Erastin, PR, IKE, SAS, Glutamate), and HMGCR inhibitors (e.g., Fluvastatin, Pravastatin, Lovastatin, and Simvastatin).

[0187] Non-limiting examples of targets of ferroptosis-inducing agents include: KRAS, GPX4, GSH, SLC7A11, USP14, and lipid peroxidation. Non-limiting examples of ferroptosis-inducing agents include: Adagrasib, Ammonium ferric citrate, BSO, Celastrol, Cisplatin, Cryptotanshinone, Curcumin, Dihydroartemisinin, Dihydroisotanshinone I, Erastin, Erianin, Ginkgetin, 6-Gingerol, Levobupivacaine, Orlistat, PACMA31, Parcetamol, PdPT, RSL3, Siramesine and Lapatinib, Sorafenib, Sulforaphane, Selenium depletion, Zinc, Deferasirox, and Dp44mT.

[0188] Non-limiting examples of pro-ferroptotic genes include: ACO1, ACSL1, ACSL4, ALOX12, ALOX15, ALOX5, ATM, BCNU, BID, BSO, CDO1, CHAC1, DMT, HM0X1, IFNy, IRB2, LPCAT3, LTF, MDR1, NCOA4, NOS1, NOS2, NOS3, PEBP1, PKCpiI, POR, PTGS2 (COX-2), SLC11A2, Tf/TFRl, and TP53.

[0189] Non-limiting examples of anti-ferroptosis genes include: ACC, ACLS3, AIFM2, AMPK, ATF3, ATF4, ATF6, CD44, , CISDI, CISD2, DHFR, DHODH, DNAJB1, DNAJB6, FASN, Ferritin, FPXL5, FSP1, FTH1, FTL, GCH1, GCL, GCLC, GPX1, GPX4, GPX7, GPX8, GSR, GSS, HMGCR, HSP90AA1, HSP90AA2, HSP90AB1, HSP90B1, HSPB6, HSPB8, HSPD1, IL4il, iPLA2p, ISCU, KEAP1, LPCAT1, NFE2L2, NFS1, P53, PEBP1, PRDX, PR0M2, SAT1, SESN2, SLC22A18, SLC22A4, SLC22A5, SLC22A7, SLC3A2, SLC7A11, SLCA15, SQLE, SQSTM1, TFRC, TXN, TXRD1 and XBP1.

[0190] Non-limiting examples of genes associated with the occurrence of ferroptosis include: ACSL4, CHAC1, PTGS1, PTGS2, RGS4, and SLC7A11.

[0191] Non-limiting examples of genes associated with lipid peroxidation include: ACSL3, ACSL4, ALOX15, and LPCAT3.

[0192] A non-limiting example of an inhibitor of ferroptosis is a lipid with monounsaturated fatty acids (MUFAs).

[0193] A non-limiting example of a promoter of ferroptosis is a polyunsaturated fatty acid (PUFA). PUFAs are long chain fatty acids with two or more carbon-carbon double bonds and all polyunsaturated fatty acid-containing phospholipids (PUFA-PLs) are susceptible to peroxidation. [0194] In some embodiments, PLs with a bisallylic group (-CH=CH-CH2-CH=CH-) are susceptible to oxidation. For example, phospholipids (PLs) with two bisallylic groups, such as arachidonic acid (AA,20:4) or PLs with adrenic acid (AdA,22:4) at the sn2 position. [0195] In some embodiments, PUFA-PLs are readily oxidized. For example, PLs with a single bisallylic, such as PLs containing linoleic acid (LA, 18:2) and PLs with three bisallylic groups, such as PLs containing docosahexaenoic acid (22:6).

[0196] Oxidative damage to PUFA-PLs can occur through a non-enzymatic free-radical chain reaction involving Fenton chemistry and enzymatically via a family of lipoxygenases (LOX) proteins. Non-enzymatic peroxidation of the arachidonoyl acyl chain in phospholipids can generate F2-iso-prostane-containing phospholipids. In some embodiments, depletion of MUFA-PLs results in an increase in PUFA-PLs. In some embodiments, an increase in PUFA-PLs is associated with an increased likelihood of cells undergoing lipid peroxidation in their cell membranes. In some embodiments, an increase in PUFA-PLs is associated with an increased likelihood of cells undergoing cell death via ferroptosis.

[0197] Arachidonic acid is hydrolyzed into 5-hydroperoxyeicosatetraenoic acid (5-HPETE) by ALOX5, 12-hydroperoxy eicosatetraenoic acid (12-HPETE) by ALOX12 and 15- hydroperoxyeicosatetraenoic acid (12-HPETE) by ALOX15.

[0198] A non-limiting example of a validated redox lipid death signal is 15 -hydroperoxy (Hp)-arachidonoyl-phosphatidylethanolamine (15-HpETE-PE).

[0199] In some embodiments, the assay to quantify PLA2G7 or PAFAH2 expression and activity is a serum biomarker assay (e.g., enzymatic assay for Lp-PLA2 activity performed on patient plasma, or in patient tissues extracted via biopsy, enzymatic assays) can be directly performed on cell lysates. In some embodiments, the assay to quantify PLA2G7 or PAFAH2 expression is a western blot. In some embodiments, the assay to quantify PLA2G7 or PAFAH2 expression is IHC/ICC, including for markers of oxidative stress such as 4HNE, MDA and COX-2. In some embodiments, Lipidomics/mass spectrometry (LC- MS) based assays of serum or tissue can provide lipidomic fingerprints to identify subjects. In some embodiments, the assay to quantify PLA2G7 or PAFAH2 expression is qPCR (e.g., using specific primers to PLA2G7 or PAFAH2). In some embodiments, the assay to quantify PLA2G7 or PAFAH2 expression is LC/MS proteomics of cells to quantify the number of protein fragments corresponding to the PLA2G7 or PAFAH2 protein sequence. [0200] In some embodiments, a method disclosed herein comprises determining markers of ferroptosis induced due to inhibition of PLA2G7. In some embodiments, a method disclosed herein comprises determining markers of ferroptosis induced due to inhibition of PAFAH2. [0201] In some embodiments, the marker correlates to lipid peroxidation. Non-limiting examples of determining lipid peroxidation includes: measurement of thiobarbituric acid reactive substances (TBARS), Cl l-BODIPY, Liperfluo, LC-MS/MS lipidomics, anti-MDA adduct antibody staining (1F83), or anti-HNE adduct antibody staining (HNEJ-1, FerAb). [0202] In some embodiments, the marker correlates to iron accumulation. Non-limiting examples of markers for determining iron accumulation includes: transferrin receptor marker (TfR), via an antibody such as 3F3-FMA, Ferritin (FTL, FTH), Hepcidin (HAMP), Ferroportin (SLC40A1), Iron-Regulated Proteins such as ALAS2, Hemoglobin (e.g, HBA1, HBA2, HBB), Transferrin (TF), Ferrochelatase (FECH), and Iron Regulatory Proteins (IRP I and IRP2).

Oxidative Stress

[0203] Oxidative stress refers to the relative excess of reactive oxygen species (ROS) when compared with antioxidants. Oxidative stress is a factor in pathophysiological conditions and cancer development. Cells possess complex biochemical and genetic mechanisms to maintain a balance between the relative abundance of ROS and antioxidants. Perturbations in the balance between the relative abundance of ROS and antioxidants can have pathophysiological consequences.

[0204] Healthy cells use mechanisms to maintain intracellular levels of reactive oxygen species (ROS) and overall redox homeostasis to avoid damage to DNA, proteins, and lipids. Cancer cells exhibit elevated ROS levels and upregulated protective antioxidant pathways. [0205] In some embodiments, the modulator of oxidative stress is an antioxidant enzyme mimics. Non-limiting examples of antioxidant mimics include: MAC, ALT-2073, Ebselen, GC4419, AEOL-10150, EUK-8, EUK-134, and EUK-189.

[0206] In some embodiments, the modulator of oxidative stress is a NRF2 activator. Nonlimiting examples of NRF2 activators include: Sulforaphane, Resveratrol, Quercetin, Curcumin, Bardoxolone-methyl(CDDO-Me, RTA402), Omaveloxolone (RTA-408), Dimethl fumarate, Oltipraz, CXA-10, Andrographolide, and Ursodiol, ALKS-8700), Sulfasalazine, and NOV-001.

[0207] In some embodiments, the modulator of oxidative stress is an anticancer drug. In some embodiments, the modulator of oxidative stress modulates intracellular ROS. Non- limiting examples of modulators of oxidative stress include: Sulfasalazine, NOV-002, PDGFR inhibitors (e.g., Imatinib), EGFR inhibitors (e.g., Erlotinib), BRAF inhibitors (e.g., Vemurafennib), anti-CD20 (e.g., Rituzimab), Procarbazine, Doxorubicin, Arsenic trioxide, 5-fluorouracil, 2-methoxyestradiol, N-(4-hydroxypenyl retinamide), platinum drugs, Gamitrinib, ARQ 501, STA-4783, Bortezomib, Celecoxib, Sanguinarine, and Rapamycin.

Ras

[0208] RAS proteins function as GDP-GTP -regulated binary on-off switches that regulate cytoplasmic signaling networks that control diver normal cellular processes. Three human RAS genes exist (KRAS, NRAS and HRAS) and encode four RAS proteins, with two KRAS isoforms that arise from alternative RNA splicing (KRAS4A and KRAS4B).

RAS protein binary switches cycle between ON and OFF states during signal transduction. These switches are normally tightly controlled, but in cancer, mutations in the RAS genes or their regulators can render RAS proteins persistently active. In the Ras signaling pathway, activated Ras further stimulates the phosphorylation of substrate proteins, mediating many signaling pathways involved in various vital cellular processes. The Ras signaling pathway is involved in many important cellular processes such as cell proliferation and survival, differentiation, apoptosis, cytoskeletal movement, protein transport & secretion.

[0209] Ras combines with GDP in an inactive state. When Ras protein is released from Ras/GDP and binds to GTP, Ras is activated. Guanylate exchange factors (GEFs) (e.g. Sos) are stimuli for the activation of Ras. GAP (GTPase activating proteins) catalyzing the hydrolysis of GTP to GDP in Ras/GTP, deactivating Ras.

[0210] GEFs promote the release of GDP from Ras/GDP. Sos cannot bind directly to the receptor due to the lack of the SH2 domain. The adaptor protein Grb2 binds to the phosphotyrosine residue of the receptor via SH2 and then binds to Sos through SH3. Sos subsequently contacts Ras on the membrane to activate Ras. Ras can be activated through the activation of RPTK by growth factors such as EGF, PDGF, and FGF.

[0211] Active Ras binds and activates Raf. Activated Raf phosphorylates and activates MEK1/2. MEK 1/2 phosphorylates ERK1/2 for activation. Activation of ERK1/2 into the nucleus activates the expression of many downstream genes such as Elk-1, elf-4E to promote cell proliferation and differentiation.

[0212] Activated Ras can directly bind and activate PI3K. Activation of PI3K converts PIP2 into PIP3. The second messenger PIP3 further stimulates PDK, which subsequently activates Akt, starting the PI3K-AKt signaling pathway that regulates cell proliferation. [0213] RalGDS stimulates GDP to dissociate from the Ras-related RalA and RalB GTPases. This dissociation allows GTP binding and activation of the GTPases. RalBP can inhibit RacGTP and Cdc42 enzymes, and then regulate actin cytoskeleton remodeling and activate transcription factor NF-KP through Rac/Cdc42. The process can promote production of anti-apoptotic proteins to inhibit apoptosis.

[0214] Missense gain-of-function mutations are found in multiple human cancers. RAS mutations can be defined by the isoform (KRAS, NRAS or HRAS) and codon (12, 13 or 61) and base change (e.g., KRAS G12D).

[0215] Ras gene mutations in tumors can include the substitution of the 12th glycine, the 13th glycine, or the 61st glutamine by other amino acid residues. GAP is unable to hydrolyze GTP in mutant RAS. Without the help of GAP, the hydrolysis of GTP is completely dependent on the GTPase of RAS, so the hydrolysis lasts longer. Thus, Ras/GTP cannot become Ras/GDP and is in a GTP -bound state for a long time. This state causes excessive activation of the Ras-Raf-MEK-ERK pathway, resulting in excessive cell proliferation and tumorigenesis.

Conditions

[0216] The methods herein can be used to detect, screen, diagnose, monitor, or treat a condition in a subject. The present disclosure describes methods and systems for treating conditions comprising administering an inhibitor of PLA2G7. In some embodiments, the condition is cancer.

[0217] Cancer is a collection of related diseases characterized by uncontrolled proliferation of cells with the potential to metastasize throughout the body. Cancer can be classified into five broad categories including, for example: carcinomas, which can arise from cells that cover internal and external parts of the body such as the lung, breast, and colon; sarcomas, which can arise from cells that are located in bone, cartilage, fat, connective tissue, muscle, and other supportive tissues; lymphomas, which can arise in the lymph nodes and immune system tissues; leukemia, which can arise in the bone marrow and accumulate in the bloodstream; and adenomas, which can arise in the thyroid, the pituitary gland, the adrenal gland, and other glandular tissues.

[0218] Although different cancers can develop in virtually any of the body's tissues, and contain unique features, the basic processes that cause cancer can be similar in all forms of the disease. Cancer begins when a cell breaks free from the normal restraints on cell division and begins to grow and divide out of control. Genetic mutations in the cell can preclude the ability of the cell to repair damaged DNA or initiate apoptosis, and can result in uncontrolled growth and division of cells.

[0219] In some embodiments, the cancer harbors one or more RAS mutations. In some embodiments, the one or more RAS mutation is KRAS, NRAS, HRAS, or a combination thereof. In some embodiments, the one or more RAS mutation is a constitutively active variant. In some embodiments, the e constitutively active variant is a gain of function mutation, a duplication, or a gene amplification.

[0220] In some embodiments, the cancer harboring one or more RAS mutations comprises a KRAS mutation. In some embodiments, the cancer harboring one or more RAS mutations comprises a NRAS mutation. In some embodiments, the cancer harboring one or more RAS mutations comprises a HRAS mutation.

[0221] In some embodiments, the cancer is associated with one or more HRAS mutations. Non-limiting examples of cancers associated with one or more HRAS mutations include: head and neck squamous cell carcinoma, anaplastic thyroid carcinoma, follicular thyroid carcinoma, and bladder urothelial carcinoma.

[0222] In some embodiments, the cancer is associated with one or more KRAS mutations. [0223] Non-limiting examples of cancers associated with one or more KRAS mutations include: anaplastic thyroid carcinoma, lung adenocarcinoma, small intestine adenocarcinoma, colon adenocarcinoma, rectal adenocarcinoma, cholangiocarcinoma, gallbladder carcinoma, pancreatic adenocarcinoma, bladder urothelial carcinoma, ovarian serous cystadenocarcinoma, uterine corpus endometrial carcinoma, cervical squamous carcinoma and endocervical adenocarcinoma, acute lymphoblastic leukemia, acute myeloid leukemia, chronic myeloid leukemia, and plasma cell myeloma.

[0224] In some embodiments, the cancer is associated with one or more NRAS mutations. [0225] Non-limiting examples of cancers associated with one or more NRAS mutations include: papillary thyroid carcinoma, anaplastic thyroid carcinoma, follicular thyroid carcinoma, skin cutaneous melanoma, acute lymphoblastic leukemia, acute myeloid leukemia, chronic myeloid leukemia, and plasma cell myeloma.

[0226] In some embodiments, the cancer is lymphoma.

[0227] Non-limiting examples of lymphoma include: subsets of lymphoma, Hodgkin lymphoma, Non-Hodgkin lymphoma, B cell lymphoma, Diffuse large B-cell lymphoma (DLBCL), Mantle cell lymphoma (MCL), Lymphoblastic lymphoma, Burkitt lymphoma (BL), Primary mediastinal (thymic) large B-cell lymphoma (PMBCL), Transformed follicular and transformed mucosa-associated lymphoid tissue (MALT) lymphomas, High- grade B-cell lymphoma with double or triple hits (HBL), Primary cutaneous DLBCL, leg type, Primary DLBCL of the central nervous system, Primary central nervous system (CNS) lymphoma, Acquired immunodeficiency syndrome (AIDS)-associated lymphoma, Follicular lymphoma (FL), Marginal zone lymphoma (MZL), Chronic lymphocytic leukemia/small-cell lymphocytic lymphoma (CLL/SLL), Gastric mucosa-associated lymphoid tissue (MALT) lymphoma, Lymphoplasmacytic lymphoma, Waldenstrom macroglobulinemia (WM), Nodal marginal zone lymphoma (NMZL), Splenic marginal zone lymphoma (SMZL), Mature T-cell and natural killer (NK)-cell lymphomas, Peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS), Systemic anaplastic large-cell lymphoma (ALCL), Lymphoblastic lymphoma, Hepatosplenic T-cell lymphoma, Enteropathy-associated intestinal T-cell lymphoma, Monomorphic epitheliotropic intestinal T-cell lymphoma, Angioimmunoblastic T-cell lymphoma (AITL), Adult T-cell leukemia/lymphoma, Extranodal natural killer (NK)/T-cell lymphoma (ENK/TCL), nasal type, Cutaneous T-cell lymphoma (CTCL), Mycosis fungoides (MF), Sezary syndrome (SS), Primary cutaneous anaplastic large-cell lymphoma (pcALCL), Subcutaneous panniculitis-like T-cell lymphoma (SPTCL), and Primary cutaneous gamma delta T-cell lymphoma.

[0228] In some embodiments, the cancer is lung cancer.

[0229] Non-limiting examples of lung cancer include: small-cell lung cancer (SCLC), limited-stage SCLC (LS-SCLC), extensive-stage SCLC (ES-SCLC), non-small cell lung cancer (NSCLC), lung adenocarcinoma (LU AD), lung squamous-cell carcinoma (LUSC), and NSCLC not otherwise specific (NOS).

[0230] In some embodiments, the small-cell lung cancer (SCLC) tumor does not have a gene mutation in TP53, RBI, NOTCH1, PTEN, CDKN2A, TP73, MYCL1, FGFR1, KIT, MYCC, MYCN, PIK3CA, STK11 or KEAPl. In some embodiments, the small-cell lung cancer (SCLC) tumor has a mutation in TP53, RBI, NOTCH1, PTEN, CDKN2A, TP73, MYCL1, FGFR1, KIT, MYCC, MYCN, PIK3CA, STK11 or KEAPl.

[0231] In some embodiments, the cancer is stomach cancer.

[0232] Non-limiting examples of stomach cancer include: adenocarcinoma of the stomach including gastric cardia cancer and non-cardia gastric cancer, gastroesophageal junction adenocarcinoma (GEJ), gastrointestinal neuroendocrine tumors, gastrointestinal stromal tumors (GIST), and primary gastric lymphoma

[0233] In some embodiments, the subject has limited-stage SCLC (LS-SCLC). In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of a combination of chemotherapy. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically- effective amount of cisplatin and etoposide. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of radiation therapy.

[0234] In some embodiments, the subject has extensive-stage SCLC (ES-SCLC). In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of a combination of chemotherapy. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically- effective amount of a platinum chemotherapeutic agent and etoposide. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of a platinum-based chemotherapy and irinotecan. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of immunotherapy. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of carboplatin, etoposide, and immune checkpoint blockade.

[0235] Non-limiting examples of platinum-based chemotherapy include carboplatin and cisplatin. Non-limiting examples of immune checkpoint blockade therapy include Atezolizumab and Durvalumab.

[0236] In some embodiments, the subject has early-stage NSCLC. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically- effective amount of a radiation therapy. Non-limiting examples of radiation therapy include targeted high intensity radiation, stereotactic body radiation, and radiofrequency ablation. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of a combination of ablation and adjuvant chemotherapy and radiation.

[0237] In some embodiments, the subject has advanced or metastatic LU AD. In some embodiments, the subject has NOS. In some embodiments, the subject has advanced or metastatic LUSC. In some embodiments, a tumor from the subject has an EGFR mutation, ALK mutation, KRAS mutation, ROS1 mutation, BRAF mutation, NTRK1/2/3 mutation, METexl4 skipping mutation, or RET mutation. In some embodiments, the subject has a positive PD-L1 test.

[0238] In some embodiments, a tumor from the subject has an EGFR Exon 19 Deletion or EGFR L858R mutation. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of targeted therapy or immunotherapy. Non-limiting examples of targeted therapy or immunotherapy include: Afatinib, Erlotinib, Dacomitinib, Gefitinib, Osimertinib, Erlotinib and ramucirumab, Erlotinib, and bevacizumab.

[0239] In some embodiments, a tumor from the subject has an EGFR S7681 mutation, EGFR L861Q mutation, or EGFR G 719X mutation. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of Afatinib, Erlotinib, Dacomitinib, Gefitinib, or Osimertinib.

[0240] In some embodiments, a tumor from the subject has a KRAS G12C mutation. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of Sotorasib and Adagrasib.

[0241] In some embodiments, a tumor from the subject has an ALK rearrangement. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of Alectinib, Brigatinib, Ceritinib, Crizotinib, or Lorlatinib.

[0242] In some embodiments, a tumor from the subject has ROS1 rearrangement. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of Ceritinib, Crizotinib, Entrectinib, Lorlatinib, or Entrectinib.

[0243] In some embodiments, a tumor from the subject has a BRAF V600E mutation. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of Dabrafenib/trametinib, Dabrafenib, or Vemurafenib.

[0244] In some embodiments, a tumor from the subject has a NTRK1/2/3 gene fusion. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of Larotrectinib or Entrectinib.

[0245] In some embodiments, a tumor from the subject has a MET Exon 14 skipping mutation. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of Capmatinib, Crizotinib, or Tepotinib. [0246] In some embodiments, a tumor from the subject has a RET rearrangement. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of Selpercatinib, Pralsetinib, or Cabozantibin.

[0247] In some embodiments, a tumor from the subject has a PD-L1>1%. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of Pembrolizumab, (carboplatin or cisplatin)/pemetrexed/pembrolizumab, carboplatin/paclitaxel/bevacizumab/atezolizumab, carboplatin/(paclitaxel or albumin-bound paclitaxel)/pembrolizumab, carboplatin/albumin- bound paclitaxel/atezolizumab, Nivolumab/ipilimumab, Nivolumab/ipilimumab/pemetrexed/(carboplatin or cisplatin), or Nivolumab/ipilimumab/paclitaxel/carboplatin.

[0248] In some embodiments, a tumor from the subject has a PL-Dl>50%. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of Atezolizumab, or Cemiplimab-rwlc.

[0249] In some embodiments, the lung adenocarcinoma (LU AD) tumor has a mutation in TP53, KRAS, EGFR, KEAP1, STK11, NF1, MET, PIK3CA, CDKN2A, RBI or RIT1. In some embodiments, the lung squamous-cell carcinoma (LUSC) tumor has a mutation in TP53, MLL3, AKT3, PIK3CA, NFE2L2, KEAP1, PTEN, NOTCH1.

[0250] In some embodiments, the cancer is pancreatic cancer.

[0251] Non-limiting examples of lung cancer include: pancreatic ductal adenocarcinoma (PDAC), acinar carcinoma, pancreaticoblastoma and neuroendocrine tumors.

[0252] In some embodiments, the subject has PDAC. In some embodiments, a tumor from the subject has a mutation in KRAS, CDKN2A, TP53, SMAD4, BRCA1, BRCA2, ATM, PALB2, MLH1, MSH2, MSH6, PMS2, SIKH.

[0253] In some embodiments, the subject has locally advanced or metastatic pancreatic cancer. In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of FOLFIRINOX (5-FU/leucovorin plus oxaliplatin and irinotecan) or modified FOLFIRINOX (initial dosing of bolus 5-FU and irinotecan each reduced by 25%). In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of gemcitabine monotherapy, GEMOX (gemcitabine and oxaliplatin) or GTX (gemcitabine, docetaxel and capecitabine).

[0254] In some embodiments, a biomarker of pancreatic cancer is human equilibrative nucleoside transporter 1 (hENTl).

[0255] In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of Gemcitabine in combinations with albumin-bound paclitaxel, cisplatin, Erlotinib, capecitabine, or fluoropyrimidine-based therapies. [0256] In some embodiments, the cancer is renal cell carcinoma (RCC). In some embodiments, the cancer is clear cell renal cell carcinoma (ccRCC). Non-limiting examples of kidney tumors include papillary, chromophobe, translocation and Bellini duct tumors.

[0257] In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of Axitinib and pembrolizumab, Cabozanitib and nivolumab, Lenvatinib and pembrolizumab, Axitinib and avelumab, Cabozantinib, Ipilimumab and nibolumab, Pazopanib, Sunitinib, or high-dose IL-2.

[0258] In some embodiments, a method disclosed herein further comprises administering to the subject a therapeutically-effective amount of Axitinib and pembrolizumab, Cabozanitib and nivolumab, Ipilimumab and nibolumab, Lenvatinib and pembrolizumab, Cabozantinib, Axitinib and avelumab, Pazopanib, Sunitinib, High-dose IL-2, or Temsirolimus.

[0259] In some embodiments, cancer cells from the subject exhibit elevated expression or activity of PLA2G7 or PAFAH2 compared to healthy control cells. In some embodiments, cancer cells from the subject exhibit elevated expression or activity of PLA2G7 or PAFAH2 compared to normal control cells. In some embodiments, cancer cells from the subject exhibit elevated expression or activity of PLA2G7 or PAFAH2 compared to primary tumor cells.

[0260] Non-limiting examples of cancers comprising cancer cells having elevated expression or activity of PLA2G7 compared to normal control cells include: Bladder urothelial carcinoma, Cervical squamous cell carcinoma, Esophageal carcinoma, Head and neck squamous cell carcinoma, Kidney chromophobe, Kidney renal clear cell carcinoma, Kidney renal papillary cell carcinoma, Liver hepatocellular carcinoma, Prostate adenocarcinoma, Metastatic prostate cancer, Metastatic skin cutaneous melanoma, Stomach adenocarcinoma, Testis germ cell, and Thyroid carcinoma.

[0261] Non-limiting examples of cancers comprising cancer cells having elevated expression or activity of PAFAH2 compared to normal control cells include: Adrenocortical carcinoma, Breast invasive carcinoma, Cholangiocarcinoma, Colon adenocarcinoma, Glioblastoma multiforme, Head and Neck squamous cell carcinoma, Kidney chromophobe, Kidney renal clear cell carcinoma, Kidney renal papillary cell carcinoma, Liver hepatocellular carcinoma, Lung adenocarcinoma, Lung squamous cell carcinoma, Rectum adenocarcinoma, Skin cutaneous melanoma metastasis, and Thyroid carcinoma.

Dosing and Administration [0262] In practicing the methods or use provided herein, therapeutically-effective amounts of the compounds described herein are administered to a subject having a disease or condition to be treated. A therapeutically-effective amount can vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compounds used, and other factors. Subjects can be, for example, humans, elderly adults, adults, adolescents, pre-adolescents, children, toddlers, infants, or neonates. A subject can be a patient.

[0263] The particular dosage of a compound required to treat the condition can depend on the severity of the condition, the route of administration, and related factors that can be decided by the attending physician. The particular dosage of a compound required to treat the cancer can depend on the severity of the cancer, the route of administration, and related factors that can be decided by the attending physician.

[0264] A therapeutically-effective amount of a compound of the present disclosure can be expressed as mg of the compound per kg of subject body mass. In some embodiments, a therapeutically-effective amount is 1-1,000 mg/kg, 1-500 mg/kg, 1-250 mg/kg, 1-100 mg/kg, 1-50 mg/kg, 1-25 mg/kg, or 1-10 mg/kg. In some embodiments, a therapeutically- effective amount is 5 mg/kg, 10 mg/kg, 25 mg/kg, 50 mg/kg, 75 mg/kg, 100 mg/kg, 150 mg/kg, 200 mg/kg, 250 mg/kg, 300 mg/kg, 400 mg/kg, 500 mg/kg, 600 mg/kg, 700 mg/kg, 800 mg/kg, 900 mg/kg, 1,000 mg/kg, about 5 mg/kg, about 10 mg/kg, about 25 mg/kg, about 50 mg/kg, about 75 mg/kg, about 100 mg/kg, about 150 mg/kg, about 200 mg/kg, about 250 mg/kg, about 300 mg/kg, about 400 mg/kg, about 500 mg/kg, about 600 mg/kg, about 700 mg/kg, about 800 mg/kg, about 900 mg/kg, or about 1,000 mg/kg.

[0265] A compound described herein can be present in a composition in a range of from about 1 mg to about 5 mg, from about 5 mg to about 10 mg, from about 10 mg to about 15 mg, from about 15 mg to about 20 mg, from about 20 mg to about 25 mg, from about 25 mg to about 30 mg, from about 30 mg to about 35 mg, from about 35 mg to about 40 mg, from about 40 mg to about 45 mg, from about 45 mg to about 50 mg, from about 50 mg to about 55 mg, from about 55 mg to about 60 mg, from about 60 mg to about 65 mg, from about 65 mg to about 70 mg, from about 70 mg to about 75 mg, from about 75 mg to about 80 mg, from about 80 mg to about 85 mg, from about 85 mg to about 90 mg, from about 90 mg to about 95 mg, from about 95 mg to about 100 mg, from about 100 mg to about 125 mg, from about 125 mg to about 150 mg, from about 150 mg to about 175 mg, from about 175 mg to about 200 mg, from about 200 mg to about 225 mg, from about 225 mg to about 250 mg, or from about 250 mg to about 300 mg. [0266] A compound described herein can be present in a composition in an amount of about 1 mg, about 5 mg, about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, about 95 mg, about 100 mg, about 125 mg, about 150 mg, about 175 mg, about 200 mg, about 225 mg, about 250 mg, or about 300 mg.

[0267] In some embodiments, a therapeutically-effective amount can be administered 1-35 times per week, 1-14 times per week, or 1-7 times per week. In some embodiments, a therapeutically-effective amount can be administered 1-10 times per day, 1-5 times per day, 1 time, 2 times, or 3 times per day.

[0268] In some embodiments, a compound disclosed herein can be administered in therapeutically-effective amounts by various forms and routes including, for example, by intravenous, intravitreal, subcutaneous, intramuscular, oral, rectal, aerosol, parenteral, ophthalmic, pulmonary, transdermal, vaginal, otic, nasal, intraocular, and topical administration. Non-limiting examples of parenteral or systemic administration includes subcutaneous, intravenous, intraperitoneal, and intramuscular injections.

[0269] Compounds of the present disclosure, whether administered alone, or in combination with anti-cancer agents and therapies such as radiotherapy, can be administered to a subject in need of such administration, for example a human or animal patient.

[0270] Compounds of the present disclosure, whether administered alone, or in combination with a ferroptosis inducer can be administered to a subject in need of such administration, for example a human or animal patient.

[0271] Compounds of the present disclosure, whether administered alone, or in combination with a modulator of oxidative stress can be administered to a subject in need of such administration, for example a human or animal patient.

Pharmaceutical Compositions

[0272] A pharmaceutical composition can be a combination of any compounds described herein with other chemical components, such as pharmaceutically acceptable carriers, stabilizers, binders, diluents, dispersing agents, suspending agents, thickening agents, solubilizing agents, or excipients. Such compositions can be in the form of, for example, granules, powders, tablets, capsules, syrup, suppositories, injections, emulsions, elixirs, suspensions or solutions. The pharmaceutical composition facilitates administration of the compound to an organism. [0273] Non-limiting examples of pharmaceutically-acceptable excipients can be found, for example, in Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington’s Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975; Liberman, H.A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins 1999), each of which is incorporated by reference in its entirety.

[0274] In some embodiments, the pharmaceutical composition provided herein comprises a buffer as an excipient. Non-limiting examples of buffers include potassium phosphate, sodium phosphate, phosphate buffer, citrate buffer, saline sodium citrate buffer (SSC), acetate, saline, physiological saline, phosphate buffer saline (PBS), 4-2-hydroxyethyl-l- piperazineethanesulfonic acid buffer (HEPES), 3-(N-morpholino)propanesulfonic acid buffer (MOPS), and piperazine-N,N'-bis(2-ethanesulfonic acid) buffer (PIPES), citric acid monohydrate, sodium dihydrogen phosphate, potassium dihydrogen phosphate, or any combination thereof.

[0275] In some embodiments, the pharmaceutical composition provided herein comprises an alcohol as an excipient. Non-limiting examples of alcohols include ethanol, propylene glycol, glycerol, polyethylene glycol, chlorobutanol, isopropanol, xylitol, sorbitol, maltitol, erythritol, threitol, arabitol, ribitol, mannitol, galactilol, fucitol, lactitol, or any combination thereof.

[0276] Pharmaceutical preparations can be formulated with polyethylene glycol (PEG). PEGs with molecular weights ranging from about 300 g/mol to about 10,000,000 g/mol can be used. Non-limiting examples of PEGs include PEG 200, PEG 300, PEG 400, PEG 540, PEG 550, PEG 600, PEG 1000, PEG 1450, PEG 1500, PEG 2000, PEG 3000, PEG 3350, PEG 4000, PEG 4600, PEG 6000, PEG 8000, PEG 10,000, and PEG 20,000.

[0277] Further excipients that can be used in a composition described herein include, for example, benzalkonium chloride, benzethonium chloride, benzyl alcohol, butylated hydroxyanisole, butylated hydroxytoluene, chlorobutanol, dehydroacetic acid, ethylenediamine, ethyl vanillin, glycerin, hypophosphorous acid, phenol, phenylethyl alcohol, phenylmercuric nitrate, potassium benzoate, potassium metabisulfite, potassium sorbate, sodium bisulfite, sodium metabisulfite, sorbic acid, thimerasol, acetic acid, aluminum monostearate, boric acid, calcium hydroxide, calcium stearate, calcium sulfate, calcium tetrachloride, cellulose acetate pthalate, microcrystalline celluose, chloroform, citric acid, edetic acid, and ethylcellulose. [0278] In some embodiments, the pharmaceutical composition provided herein comprises an aprotic solvent as an excipient. Non-limiting examples of aprotic solvents include perfluorohexane, a,a,a-trifluorotoluene, pentane, hexane, cyclohexane, methylcyclohexane, decalin, dioxane, carbon tetrachloride, freon-11, benzene, toluene, carbon disulfide, diisopropyl ether, diethyl ether, t-butyl methyl ether, ethyl acetate, 1,2-dimethoxy ethane, 2- methoxyethyl ether, tetrahydrofuran, methylene chloride, pyridine, 2-butanone, acetone, N- methylpyrrolidinone, nitromethane, dimethylformamide, acetonitrile, sulfolane, dimethyl sulfoxide, and propylene carbonate.

[0279] The amount of the excipient in a pharmaceutical composition described herein can be about 0.01%, about 0.02%, about 0.03%, about 0.04%, about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.5%, about 2%, about 2.5%, about 3%, about 3.5%, about 4%, about 4.5%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 60%, about 70%, about 80%, about 90%, about 100%, about 200%, about 300%, about 400%, about 500%, about 600%, about 700%, about 800%, about 900%, or about 1000% by mass of a compound in the pharmaceutical formulation.

[0280] The amount of the excipient in a pharmaceutical composition described herein can be about 0.01%, about 0.02%, about 0.03%, about 0.04%, about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.5%, about 2%, about 2.5%, about 3%, about 3.5%, about 4%, about 4.5%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55% about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 99%, or about 100% by mass or by volume of the unit dosage form.

[0281] In some embodiments, the addition of an excipient to a pharmaceutical composition described herein can increase or decrease the viscosity of the composition by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99%. In some embodiments, the addition of an excipient to a pharmaceutical composition described herein can increase or decrease the viscosity of the composition by no greater than 5%, no greater than 10%, no greater than 15%, no greater than 20%, no greater than 25%, no greater than 30%, no greater than 35%, no greater than 40%, no greater than 45%, no greater than 50%, no greater than 55%, no greater than 60%, no greater than 65%, no greater than 70%, no greater than 75%, no greater than 80%, no greater than 85%, no greater than 90%, no greater than 95%, or no greater than 99%.

[0282] In some embodiments, a therapeutically-effective amount can be an amount effective in treating a cancer, treating a tumor, shrinking a tumor, treating a neoplasm, shrinking a neoplasm, and/or increasing subject survival.

[0283] In some embodiments, a therapeutically-effective amount can be an amount effective in treating a cancer. In some embodiments, a therapeutically-effective amount can be an amount effective in treating a tumor. In some embodiments, a therapeutically- effective amount can be an amount effective in treating a neoplasm. In some embodiments, a therapeutically-effective amount can be an amount effective in increasing subject survival. [0001] In some embodiments, a therapeutically-effective amount can be an amount effective in shrinking a tumor. The tumor can be reduced in size by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 99%.

[0002] In some embodiments, a therapeutically-effective amount can be an amount effective in shrinking a neoplasm. The neoplasm can be reduced in size by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 99%.

[0003] In some embodiments, a therapeutically-effective amount is an amount effective to slow or reverse growth of the tumor. In some embodiments, a therapeutically-effective amount is an amount effective to slow or reverse growth of a neoplasm. In some embodiments, a therapeutically-effective amount is an amount effective to slow or reverse proliferation of a neoplasm.

Pharmaceutically-Acceptable Salts.

[0284] The present disclosure provides the use of pharmaceutically-acceptable salts of any compound described herein. Pharmaceutically-acceptable salts include, for example, acidaddition salts and base-addition salts. The acid that is added to the compound to form an acid-addition salt can be an organic acid or an inorganic acid. A base that is added to the compound to form a base-addition salt can be an organic base or an inorganic base. In some embodiments, a pharmaceutically-acceptable salt is a metal salt.

[0285] Metal salts can arise from the addition of an inorganic base to a compound described herein. The inorganic base consists of a metal cation paired with a basic counterion, such as, for example, hydroxide, carbonate, bicarbonate, or phosphate. The metal can be an alkali metal, alkaline earth metal, transition metal, or main group metal. In some embodiments, the metal is lithium, sodium, potassium, cesium, cerium, magnesium, manganese, iron, calcium, strontium, cobalt, titanium, aluminum, copper, cadmium, or zinc.

[0286] In some embodiments, a metal salt is a lithium salt, a sodium salt, a potassium salt, a cesium salt, a cerium salt, a magnesium salt, a manganese salt, an iron salt, a calcium salt, a strontium salt, a cobalt salt, a titanium salt, an aluminum salt, a copper salt, a cadmium salt, or a zinc salt.

[0287] Ammonium salts can arise from the addition of ammonia or an organic amine to a compound described herein. In some embodiments, the organic amine is triethyl amine, diisopropyl amine, ethanol amine, diethanol amine, triethanol amine, morpholine, N- methylmorpholine, piperidine, N-methylpiperidine, N-ethylpiperidine, dibenzylamine, piperazine, pyridine, pyrrazole, piprazole, imidazole, or pyrazine.

[0288] In some embodiments, an ammonium salt is a triethyl amine salt, a diisopropyl amine salt, an ethanol amine salt, a diethanol amine salt, a triethanol amine salt, a morpholine salt, an N-methylmorpholine salt, a piperidine salt, an N-methylpiperidine salt, an N-ethylpiperidine salt, a dibenzylamine salt, a piperazine salt, a pyridine salt, a pyrrazole salt, a piprazole salt, an imidazole salt, or a pyrazine salt.

[0289] Acid addition salts can arise from the addition of an acid to a compound described herein. In some embodiments, the acid is organic. In some embodiments, the acid is inorganic. In some embodiments, the acid is hydrochloric acid, hydrobromic acid, hydroiodic acid, nitric acid, nitrous acid, sulfuric acid, sulfurous acid, a phosphoric acid, isonicotinic acid, lactic acid, salicylic acid, tartaric acid, ascorbic acid, gentisinic acid, gluconic acid, glucaronic acid, saccaric acid, formic acid, benzoic acid, glutamic acid, pantothenic acid, acetic acid, propionic acid, butyric acid, fumaric acid, succinic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, oxalic acid, or maleic acid.

[0290] In some embodiments, the salt is a hydrochloride salt, a hydrobromide salt, a hydroiodide salt, a nitrate salt, a nitrite salt, a sulfate salt, a sulfite salt, a phosphate salt, isonicotinate salt, a lactate salt, a salicylate salt, a tartrate salt, an ascorbate salt, a gentisinate salt, a gluconate salt, a glucaronate salt, a saccarate salt, a formate salt, a benzoate salt, a glutamate salt, a pantothenate salt, an acetate salt, a propionate salt, a butyrate salt, a fumarate salt, a succinate salt, a methanesulfonate salt, an ethanesulfonate salt, a benzenesulfonate salt, a p-toluenesulfonate salt, a citrate salt, an oxalate salt, or a maleate salt.

[0001] The method disclosed herein can involve administering to a patient compounds of the present disclosure alone, or in combination with a second compound.

Combinations

[0291] In some embodiments, a method disclosed herein further comprises administering a therapeutically-effective amount of a second therapy. In some embodiments, the second therapy exhibits synergy with the compound that inhibits PLA2G7. In some embodiments, the second therapy exhibits an additive therapeutic effect to the compound that inhibits PLA2G7. In some embodiments, the compound that inhibits PLA2G7 exhibits an additive therapeutic effect to the second therapy.

[0292] In some embodiments, a method disclosed herein comprises administering to a subject in need thereof a therapeutically-effective amount of a compound that inhibits PLA2G7 and a reduced amount of a second therapy, wherein the reduced amount of the second therapy is therapeutically effective for treating the condition in combination with the therapeutically-effective amount of the compound that inhibits PLA2G7, and wherein the reduced amount of the second therapy is less than an amount of the second therapy that is therapeutically effective for the condition in absence of the therapeutically-effective amount of the compound that inhibits PLA2G7.

[0293] In some embodiments, the second therapy is a therapeutically-effective amount of a ferroptosis inducer. In some embodiments, the second therapy is a chemotherapeutic agent. In some embodiments, the second therapy is a therapeutic antibody.

[0294] Non-limiting examples of chemotherapeutic agents include: an alkylating agent, a nitrosourea, an antimetabolite, an anthracycline, a topoisomerase II inhibitor, a mitotic inhibitor, an anti estrogen, a progestin, an aromatase inhibitor, an anti -androgen, an LHRH agonist, a corticosteroid hormone, a DNA alkylating agent, a taxane, a vinca alkaloid, a microtubule poison, busulfan, cisplatin, carboplatin, oxaliplatin, an octahedral platinum (IV) compound, chlorambucil, cyclophosphamide, ifosfamide, dacarbazine (DTIC), mechlorethamine (nitrogen mustard), melphalan, temozolomide, carmustine (BCNU), lomustine (CCNU), 5-fluorouracil, capecitabine, 6-mercaptopurine, methotrexate, gemcitabine, cytarabine (ara-C), fludarabine, pemetrexed, daunorubicin, doxorubicin (Adriamycin), epirubicin, idarubicin, mitoxantrone, topotecan, irinotecan, etoposide (VP- 16), teniposide, paclitaxel, docetaxel, vinblastine, vincristine, vinorelbine, prednisone, dexamethasone, L-asparaginase, dactinomycin, thalidomide, tretinoin, imatinib (Gleevec), gefitinib (Iressa), erlotinib (Tarceva), rituximab (Rituxan), bevacizumab (Avastin), ipilimumab, nivolumab (Opdivo), pembrolizumab (Ketruda), tamoxifen, fulvestrant, anastrozole, exemestane, letrozole, megestrol acetate, bicalutamide, flutamide, leuprolide, goserelin, and a combination thereof.

[0295] Additional non-limiting examples of chemotherapeutic agents include: mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, anti -hormones, angiogenesis inhibitors, and anti -androgens. Non-limiting examples are chemotherapeutic agents, cytotoxic agents, and non-peptide small molecules such as Gleevec® (Imatinib Mesylate), Kyprolis® (carfilzomib). Velcade® (bortezomib), Casodex (bicalutamide), Iressa® (gefitinib), Venclexta™ (venetoclax) and Adriamycin™, (docorubicin) as well as a host of chemotherapeutic agents. Non-limiting examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (Cytoxan™); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamine; nitrogen mustards such as chlorambucil, chlornaphazine, chlorocyclophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, calicheamicin, carabicin, carminomycin, carzinophilin, Casodex™, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5 -fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6- mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine, androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane, folic acid replenisher such as frolinic acid, aceglatone; aldophosphamide glycoside, aminolevulinic acid, amsacrine, bestrabucil, bisantrene, edatraxate, defofamine, demecolcine, diaziquone, elformithine, elliptinium acetate, etoglucid, gallium nitrate, hydroxyurea: lentinan, lonidamine, mitoguazone, mitoxantrone, mopidamol, nitracrine: pentostatin, phenamet, pirarubicin, podophyllinic acid, 2-ethylhydrazide, procarbazine, PSK, razoxane, sizofiran, spirogermanium, tenuazonic acid, triaziquone, 2, 2', 2"- trichlorotriethylamine, urethan, vindesine, dacarbazine, mannomustine, mitobronitol, mitolactol, pipobroman, gacytosine, arabinoside (“Ara-C”), cyclophosphamide, thiotepa, taxanes, e.g. paclitaxel and docetaxel, retinoic acid, esperamicins, capecitabine, and a pharmaceutically acceptable salt, acid, or derivative of any of the above.

[0296] Additional non-limiting examples of chemotherapeutic agents include: Herceptin®, Avastin®, Erbitux®, Rituxan®, Taxol®, Arimidex®, Taxotere®, ABVD, AVICINE, Abagovomab, Acridine carboxamide, Adecatumumab, 17-N-Allylamino-17- demethoxygeldanamycin, Alpharadin, Alvocidib, 3-Aminopyridine-2-carboxaldehyde thiosemicarbazone. Amonafide, Anthracenedione, Anti-CD22 immunotoxins. Antineoplastic, Antitumorigenic herbs, Apaziquone, Atiprimod. Azathioprine, Belotecan, Bendamustine, BIBW 2992, Biricodar, Brostallicin, Bryostatin, Buthionine sulfoximine, CBV (chemotherapy), Calyculin, cell-cycle nonspecific antineoplastic agents, Dichloroacetic acid. Discodermolide, Elsamitrucin, Enocitabine, Epothilone, Eribulin, Everolimus, Exatecan, Exisulind, Ferruginol, Forodesine, Fosfestrol, ICE chemotherapy regimen, IT-101, Im exon, Imiquimod, Indolocarb azole, Irofulven, Laniquidar. Larotaxel, Lenalidomide, Lucanthone, Lurtotecan, Mafosfamide, Mitozolomide, Nafoxidine, Nedaplatin, Olaparib, Ortataxel, PAC-1, Pawpaw. Pixantrone, Proteasome inhibitor, Rebeccamycin, Resiquimod, Rubitecan, SN-38, Salinosporamide A, Sapacitabine, Stanford V, Swainsonine, Talaporfin, Tariquidar, Tegafur-uracil, Temodar, Tesetaxel, Triplatin tetranitrate, Tris(2-chloroethyl)amine, Troxacitabine, Uramustine, Vadimezan, Vinflunine, ZD6126, and Zosuquidar.

[0297] Radiation therapy for inhibiting abnormal cell growth or treating the hyperproliferative disorder in the mammal. The administration of the compound of the disclosure in this combination therapy can be determined as described herein. [0298] In some embodiments, the second therapy is radiation therapy. Non-limiting examples of methods for administering radiation therapy include: external-beam therapy, internal radiation therapy, implant radiation, stereotactic radiosurgery, systemic radiation therapy, radiotherapy and permanent or temporary interstitial brachytherapy.

[0299] To minimize damages to normal tissues, a high dose of IR can be delivered through multiple low doses, also referred to as dose fractionation. Fractionation includes conventionally fractionation (e.g., once daily, 5 times/week), hypofractionation (e.g., once daily, 5 times/week), and hyperfractionation (e.g., twice daily, 5 times/week). Different RT doses and fractionation schedules can result in differential levels of ferroptosis. Lipid peroxidation and ferroptosis can be augmented with increasing doses.

[0300] Any embodiments disclosed herein can be used in conjunction or individually. For example, any pharmaceutically-acceptable excipient, method, technique, solvent, or compound disclosed herein can be used together with any other pharmaceutically- acceptable excipient, method, technique, solvent, or compound disclosed herein to achieve any therapeutic result. Compounds, excipients, and other formulation components can be present at any amount, ratio, or percentage disclosed herein in any such formulation, and any such combination can be used therapeutically for any purpose described herein.

Subjects

[0301] Disclosed herein is a method of treating a condition in a subject in need thereof. In some embodiments, the subject possesses a non- wild type gene. In some embodiments, the subject possesses one, two, three, four, or five non -wild type genes disclosed herein. In some embodiments, the subject possesses a non- wild type RAS gene. In some embodiments, the subject possesses a non-wild type RAS gene and one other non-wild type gene disclosed herein, In some embodiments, the subject possesses a non-wild type RAS gene and a non-wild type STK11 gene. In some embodiment, the subject possesses a non- wild type RAS gene and a non-wild type KEAP1 gene. In some embodiments, the subject possesses a non-wild type STK11 gene and a non-wild type KEAP1 gene. In some embodiments, the subject possesses a non-wild type RAS gene and two other non-wild type gene disclosed herein. In some embodiments, the subject possesses a non-wild type RAS gene, a non-wild type STK11 gene, and a non-wild type KEAP1 gene.

[0302] Disclosed herein is a method comprising determining based on an assay of a subject that the subject has at least a threshold number of genes. In some embodiments, the threshold number of genes is 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 genes. [0303] In some embodiments, a method disclosed herein comprises determining based on an assay of a subject that the subject has at least a threshold number of genes, wherein the threshold number of genes indicates that a compound that inhibits PLA2G7 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of genes is five.

[0304] In some embodiments, a method disclosed herein comprises determining based on an assay of a subject that the subject has at least a threshold number of genes, wherein the threshold number of genes indicates that a compound that inhibits PLA2G7 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of genes is ten.

[0305] Disclosed herein is a method comprising determining based on an assay of a subject that the subject expresses at least a threshold number of phospholipases. In some embodiment, the threshold number of phospholipases is 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, or 45.

[0306] In some embodiments, a method disclosed herein comprises determining based on an assay of a subject that the subject expresses at least a threshold number of phospholipases, wherein the threshold number of phospholipases indicates that a compound that inhibits PLA2G7 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of phospholipases is five.

[0307] In some embodiments, a method disclosed herein comprises determining based on an assay of a subject that the subject expresses at least a threshold number of phospholipases, wherein the threshold number of phospholipases indicates that a compound that inhibits PLA2G7 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of phospholipases is twenty.

[0308] In some embodiments, a method disclosed herein comprises determining based on an assay of a subject that the subject expresses at least a threshold number of phospholipases, wherein the threshold number of phospholipases indicates that a compound that inhibits PLA2G7 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of phospholipases is forty.

[0309]

The examples herein are provided to illustrate advantages of the present technology and to further assist a person of ordinary skill in the art with preparing or using the present technology. The examples should in no way be construed as limiting the scope of the present technology, as defined by the appended claims. The examples can include or incorporate any of the variations, aspects, or embodiments of the present technology described above. The variations, aspects, or embodiments described above may also further each include or incorporate the variations of any or all other variations, aspects, or embodiments of the present technology.

EXAMPLES

EXAMPLE 1: Assays for detecting expression or activity of PLA2G7 and PAFAH2 in human tumors

[0310] Various assays to detect expression or activity of PLA2G7 and PAFAH2 were designed and tested as shown and described for FIGs. 1-2.

[0311] PLA2G7 and PAFAH2 gene expression data was accessed from cancer OMICS data. Gene expression data was compared for human cancer tissues from patients with various cancers and normal samples. FIG. 1 shows overexpression of PLA2G7 in human cancer tissues from patients with various cancers compared to normal samples. FIG. 2 shows overexpression of PAFAH2 in human cancer tissues from patients with various cancers compared to normal samples. In FIGs. 1 AND 2, the boxes represent upper and lower quartile, bars represent maximum and minimum values, and the horizonal lines represent medians. Statistical significance was estimated using Student’s t-test, considering unequal variance.

[0312] As shown in FIG. 1, PLA2G7 expression was higher compared to normal controls (with statistical significance) for human cancer tissues from patients with Bladder urothelial carcinoma (BLCA, p < 10' 4 ), Cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC, p < 10' 11 ), Esophageal carcinoma (ESC A, p < 10' 4 ), Head and Neck squamous cell carcinoma (HNSC, p < 10' 11 ), Kidney Chromophobe (KICH, p < 10' 2 ), Kidney renal clear cell carcinoma (KIRC, p < 10' 12 ), Kidney renal papillary cell carcinoma (KIRP, p < 10' 11 ), Liver hepatocellular carcinoma (LIHC, p < 10' 4 ), Prostate adenocarcinoma (PRAD, p < 10' 11 ), Stomach adenocarcinoma (STAD, p < 10' 11 ), and Thyroid carcinoma (THCA, p < 10' 11 ). Additionally, PLA2G7 expression was higher for human cancer tissues from patients with metastatic prostate cancer tumors with ERG fusion compared to tumors without ERG fusion, and metastatic skin cutaneous melanoma compared to primary tumor. PLA2G7 expression was higher for testis germ cell tumors seminoma compared to non-seminoma.

[0313] As shown in FIG. 2, PAFAH2 expression was higher compared to normal controls (with statistical significance) for human cancer tissues from patients with Breast invasive carcinoma (BRCA, p < 10' 7 ), Cholangiocarcinoma primary tumor (CHOL, p < 10' 2 ), and Glioblastoma multiforme primary tumor (GBM, p < 10' 2 ). Also shown in FIG. 2, PAFAH2 expression was higher in normal controls (with statistical significance) compared to primary tumor for human cancer tissues from patients with Colon adenocarcinoma (COAD, p < 10" n ), Head and Neck squamous cell carcinoma (HNSC, p < 10' 2 ), Kidney chromophobe (p < 10' 7 ), Kidney renal clear cell carcinoma (p < 10' 11 ), Kidney renal papillary cell carcinoma (p

< 10' 13 ), Liver hepatocellular carcinoma (p < 1.5 x 10' 2 ), Lung adenocarcinoma (LU AD, p < 10' 4 ), Lung squamous cell carcinoma (LUSC, p < 10' 11 ), Rectum adenocarcinoma (READ, p

< IO' 10 ), Skin cutaneous melanoma metastasis (SKCM, p < 10' 3 ), and Thyroid carcinoma normal (p < 5 x 10' 3 ). Additionally, PAFAH2 expression was also higher for human cancer tissues from patients with Adrenocortical carcinoma stage 2 compared to stage 3.

[0314] EXAMPLE 2: Assays for detecting expression or activity of PLA2G7 and PAFAH2 in human tumors

[0315] Enzyme activity assay - PED6 assay for PLA2G7

[0316] PED6 (N-((6-(2,4-Dinitrophenyl)amino)hexanoyl)-2-(4,4-Difluoro-5, 7-Dimethyl-4- Bora-3a,4a-Diaza-s-Indacene-3-Pentanoyl)-l-Hexadecanoyl-sn-G lycero-3- Phosphoethanolamine, Triethylammonium Salt) is a dye-labeled phospholipids used to monitor phospholipase A (PLA) activity. Lipoprotein-associated phospholipase A2 (Lp- PLA2) is a calcium-independent serine lipase of 50-kDa that hydrolyzes the acetyl group at the sn-2 position of platelet-activating factor (PAF). Upon cleavage of the fluorogenic sn-2 acyl chain by Lp-PLA2, the intramolecular quenching effect of a dinitrophenyl quencher on a lipid head group is relieved, resulting in a fluorescence increase.

[0317] Enzyme activity assay - Thio-PAF assay for PLA2G7 and PAFAH2

[0318] (2-thio-PAF) is a substrate for PAF -hydrolases (PAF-AH). Upon cleavage with PAF-AH, the free thiol is released at the sn-2 position, which can then react with a thiolreactive coumarin 7-diethylamino-3-(4'-maleimidylphenyl)-4-methylcoumarin (CPM). This reaction results in an increase in fluorescence. In the presence of inhibitors of Lp-PLA2, cleavage is prevented and no increase in fluorescent is observed.

[0319] Radioactive labeling

[0320] Radioactive labeling of the lipid substrate ([3H] acetyl-PAF or free fatty acids etc.) directly evaluates hydrolysis rate of different substrates.

[0321] ELISA assays [0322] Immunoassays are used, for example, enzyme linked immunosorbant assays (ELISA). This technique is based upon the special properties of antigen-antibody interactions with simple phase separations to produce assays for detecting biological molecules. A sandwich enzyme immunoassay is used for in vitro quantitative measurement of Lp-PLA2, PLA2G7, and PAF-AH in human serum, plasma, tissue homogenates, cell lysates, cell culture supernates (e.g., Invitrogen Human Lp-PLA2/PLA2G7/PAF-AH ELISA Kit quantitates human Lp-PLA2/PLA2G7/PAF-AH in serum, plasma, supernatant). The assay recognizes both natural and recombinant human Lp-PLA2, PLA2G7, and PAF-AH. [0323] Transcriptomics, RNA-seq, proteomics are also performed to detect activity.

[0324] Additional activity assays

[0325] Using a lipid nanosensor, changes in lysosomal lipid content of live cells are detected following inhibition of PLA2G7/PAFAH2 by darapladib. In some cell lines, inhibiting GPX4 is synthetic lethal with inhibiting PLA2G7. These cell lines are dependent on the intracellular activity of PLA2G7/PAFAH2. Other cell lines are not dependent and do not show synthetic lethality.

EXAMPLE 3: PLA2G7 Expression Correlation with Genes in Tumor Samples [0326] Assays to correlate PLA2G7 expression with genes in tumor samples were designed and tested as shown and described for FIGs. 3-7 and TABLE 4.

[0327] Gene correlations were obtained from the GEPIA webserver for analyzing RNA sequencing data from the TCGA and GTEx projects. RNA expression data from tumors of human cancer patients with various cancers were analyzed for genes associated with ferroptosis. A Pearson correlation coefficient was calculated against PLA2G7.

[0328] FIG. 3 shows a plot of correlation coefficients with PLA2G7 expression determined for genes in various tumor samples. TABLE 4 shows multiple genes strongly correlated (R 2 > 0.8) with PLA2G7 expression in tumor samples representing patients with CESC, COAD, HNSC, KIRC, KIRP, LUSC, DLBC, PDAC, TGCT and UVM. TABLE 4

[0329] FIGs. 4-7 show that statistically significant correlations are observed between PLA2G7 expression and genes in individual tumor samples from patients with Colon Adenocarcinoma (COAD), Breast Invasive Carcinoma (BRCA), Lymphoid Neoplasm Diffuse Large B-cell Lymphoma (DLBCL), and Testicular Germ Cell Tumors (TGCT), respectively.

[0330] EXAMPLE 4: Assays of PLA2G7 Expression

[0331] Assays to determine regulation of PLA2G7 expression in response to stimuli, were designed and tested as shown and described for FIGs. 8-25 and TABLE 5. a. Oxidative Stress

[0332] Cell lines were incubated in control media for baseline conditions. To simulate oxidative stress conditions, 30 pM H2O2 was added to the cell media. Immunoblotting quantification and statistical analysis were performed to characterize the expression of PLA2G7A (PLA2G7) and PLA2G7B (PAFAH2) expression under baseline and oxidative stress conditions. FIGs. 8-11 show expression of PLA2G7A and PLA2G7B in various cell lines increased in cells under oxidative stress induced via hydrogen peroxide.

[0333] Immunoblotting. All blot panel quantification was performed after density normalization to loading control or, in the case of phospho-proteins, normalization by totakphospho ratio. Protease and phosphatase inhibitor cocktail (ThermoFisher 78446) supplemented with 100 pM Pepstatin A (Sigma 11359053001) was diluted 1 : 100 into RIPA Buffer (Pierce 89901) or denaturing immunoprecipitation (IP) buffer without ethylmaleimide (20 mM HEPES, 50 mM NaCl, 0.5% NP-40, 1 mM EDTA, 0.5% SDS, 0.5% SDC) to make working cell lysis buffer for soluble targets (RIP A) or insoluble/membrane associated targets (IP buffer). Cells were washed twice with cold lx HBSS. Ice cold lysis buffer was added, and cells were scrapped with a rubber policeman. Lysate was pooled into an Eppendorf on wet ice and set to mix for 30 minutes at 4 °C. Lysate was homogenized through a 17-gauge needle with 5 full strokes, and then spun at 16,000 ref for 20 minutes in a 4 °C centrifuge. RT Bradford reagent (BioRad 5000205) was mixed 1 : 1 with deionized water diluted samples containing BSA standard or the sample lysate. Absorbance was measured at 595 nm in a Tecan Infinite M1000 Pro plate reader. Loading buffer consisted of lx Laemmli Buffer/2-Mercaptoethanol (BioRad 1610747/1610710) and deionized water if necessary. Final loading samples were aliquoted and stored at -80 °C until use. Frozen samples were immediately heated for 8 minutes at 75 °C (phospho-protein targets) or for 6 minutes at 90 °C (all other targets). TGX precast gels (BioRad 4568094, 4-20%) were loaded into a Tetra Cell electrophoresis apparatus (BioRad 1658004) filled with chilled lx Tris/Glycine/SDS buffer (BioRad 1610732). 1.5 pL running ladder (LLCOR 92698000), 10-30 pL sample, or 10-30 pL blank sample buffer were injected into each lane. Electrophoresis was run at 110 Volts, CV, for 80 minutes or until appropriate ladder separation. Blot sandwiches comprised a 0.2 pm PVDF membrane (BioRad 1620174) pre-incubated in methanol followed by transfer buffer (BioRad 10026938) held between buffer wetted-stack paper (BioRad) and the gel. A Trans-Blot Turbo transfer system (BioRad 1704150) was used on the 7-minute Midi-Turbo setting. Blot membranes were cut and immediately rinsed in lx TBS, then moved into a blocking solution (3% w/v BSA in lx TBS-T) (BioRad 1706435; Sigma P1379) and agitated at RT for 1.5 hours. Primary antibody was diluted into 1 mL of blocking buffer, which was sandwiched with the membrane between two parafilm sheets before sealing. Membrane sandwiches were incubated overnight at 4 °C with rotation. Membranes were peeled from the parafilm into lx TBS-T and washed three times for 10 minutes at RT with agitation. HRP conjugated secondary antibody was diluted in enough blocking solution to cover the membrane for 1 hour at RT with agitation. After three more 10-minute washes, membranes were rinsed in RT lx TBS then incubated for 1 minute in HRP substrate (Millipore WBLUR0500). To expose film, moist membranes were placed inside plastic inserts in an X-ray cassette. The following primary antibodies were used: Antibodies were as follows: anti-Coxl (1 :500, Cell Signaling 9896), anti-Cox2 (1 : 1000, Cell Signaling 12282), anti-p38 MAPK (1 : 1300, Cell Signaling 8690), anti-phospho-p38 MAPK T180/Y182 (1 :500, Cell Signaling 4511), anti-ATF-2 (1 : 1000, Cell Signaling 9226), anti-phospho-ATF-2 T69/T71 (1 :1000, Cell Signaling 5112), anti-TBP (1 : 1500, Cell Signaling 44059), anti-cPLA 2 (1 :1000, Cell Signaling 5249), anti-phospho-cPLA2 S505 (1 :750, Cell Signaling 53044), anti-phospho-lKBa S32 (1 :500, Cell Signaling 2859), anti-lKBa (1 : 1000, Cell Signaling 4812) anti-NF-KB p65 (1 : 1000, Selleckchem.com A5075), anti-phospho-NF-xB p65 S468 (1 :1000, Cell Signaling 3039), anti-phospho-NF-KB p65 S536 (1 : 1000, Cell Signaling 3033), anti-TNF-a (1 : 1000, Cell Signaling 11948), anti-IL-6 (1 :500, Cell Signaling 12912), anti-prostaglandin E synthase (1 : 1000, Abeam abl80589), anti-p44/42 MAPK (1 : 1000, Cell Signaling 4695), anti-phospho-p44/42 MAPK (1 : 1000, Cell Signaling 9101), anti-pl6 (1 pg, Abeam 189034), anti- p 21 wafl/ci P 1 (1.6 pg, Abeam 109199), anti-SAPK/JNK (1 : 1000; Cell Signaling 9252), anti-phospho-SAPK/JNK T183/Y185 (1 :500; Cell Signaling 4668), anti-pl9ARF (1 : 1000, Abeam ab80), anti-PLA2g7 (1 :400, Proteintech 15526-1-AP), anti- PAFAH2 (1 :400, Proteintech 10085-1-AP); secondary antibody was an HRP-conjugated anti-rabbit (1 :2000-1 :5000, Cell Signaling 7074).

[0334] Immunoblot Analysis. Pearson correlation analysis of quantified was performed using OriginPro 2021. b. KRAS Induction

[0335] In an induced iKRAS cell line model, RNA-seq was performed in control vector cell lines and iKRas cell lines following KRAS expression to identify differentially expressed genes. FIGs. 12-13 show overexpression of PLA2G4A, PNPLA2, PNPLA6, PLA2G6 and PLA2G7 in response to KRAS induction.

[0336] RNA Sequencing (RNAseq). To ensure adequate starting material, two biological replicates each of vector and -ras G12V were grown in 6-well plates so that three wells were dedicated for each replicate. On the day of extraction, media was aspirated and 500 pL of Trizol LS reagent (Thermo 10296010) was pipetted into each well. Plates were placed on wet ice, and the contents of each well were agitated and scraped with a rubber policeman to ensure all material was homogenous before transfer into a pre-cooled 2 mL Eppendorf (Fisher 05-402-24C). Samples were flash frozen in isopropanol/dry ice slurry and stored at - 80 °C until processing by the MSKCC Integrated Genomics Operation (IGO): extraction, poly-A enrichment, quality control, library preparation, and sequencing (30-40 million reads, HiSeq-PE50). Downstream bioinformatics was performed by the MSKCC Bioinformatics Core (BIC) using a standard delivery pipeline for alignment, clustering, Htseq counts, and differential expression. c. Inhibition of PLA2G7 via Darapladib [0337] To assess the response of oncogenic RAS-harboring cancer cells to a pharmacologic inhibitor of group 7 sPLA2, several tumor-derived cell lines were evaluated with darapladib. The cell lines derived from lung, pancreas, and colorectal tumors were incubated with darapladib and assayed for viability. FIG. 14 shows all cell lines were RAS harboring lines and died at low micromolar concentrations darapladib.

[0338] Cell Viability. 5,000 cells/well of doxycycline induced iKRas or target mouse/human cells were seeded in 96-well plates 1 day prior to the start of treatment. On the day of the experiment, increasing concentrations of darapladib were added in triplicate to the conditioned media. After incubation for 48 hours, the cell viability was assessed using the CellTiter-Glo® luminescent assay (Promega G9681). Prior to use, reconstituted kit reagent was equilibrated to room temperature. To initiate cell lysis, 50 pL of reagent was added to each well. Plates were incubated for 10 minutes at room temperature with gentle mixing on an orbital shaker. Luminescence was measured using a Tecan Infinite Ml 000 Pro plate reader (Tecan Group Ltd.). Results from the assays were calculated as a percentage of the non-treated control. The reported EC50 of darapladib ranges from 10 - 1000 nM, depending on the cell type. The enzyme IC50 ranges from 1 - 10 nM, as reported by the literature.

[0339] Inhibition of PLA2G7 via darapladib was found to increase lipid peroxides. FIG. 15 shows the FACS analysis of Liperfluo intensity in iKRas cells in control media, after treatment with Darapladib and after siRNA silencing of PLA2G7. FIGs. 16-17 show Liperfluo intensity increased following treatment with Darapladib compared to vehicle control.

[0340] Fluorescence Analysis. Liperfluo lyophilized dye (Dojindo Laboratories) was stored at 4 °C and reconstituted in DMSO per manufacturer instructions, protected from light. Cells were washed once in lx HBSS and treated with serum-free media containing 2 pM of dye for 30 minutes at 37 °C. After labelling, cells were rinsed once and resuspended in cold FACS buffer. Cells were stored on ice and taken for flow cytometry using Cytek Aurora Spectral Flow Cytometer gated to detect dye fluorescence in the green/FITC channel (Excitation: 488nm, Emission: 500-550 nm). Liperfluo analysis was performed in GraphPad Prism 9 and plots generated using OriginPro 2021 d. Combination of Darapladib and Oxidative Stress for viability. FIG. 25 shows statistically-significant dependence of sensitivity to Erastin on expression level of PLA2G7A.

[0346] EXAMPLE 5: Assays of Synergy with PLA2G7 Inhibitors

[0347] Assays to determine synergy of inhibitors of PLA2G7 (e.g., by darapladib) in combination with modulators of oxidative stress, modulators of ferroptosis, and chemotherapeutic agents were designed and tested as shown and described for FIGs. 26-56 and TABLE 6. a. Combination of Darapladib and Modulators of Oxidative Stress

[0348] FIGs. 26-33 show heatmap plots of the tumor-derived PANC-1 cell viability assessed as a function of inhibition of PLA2G7 (e.g., by darapladib) in combination with various modulators. Table 6 shows synergy with darapladib was observed with Erastin (FIG. 26), FIN56 (FIG. 27) and H2O2, (FIG. 33).

TABLE 6

[0349] Synergy Analysis. Drug synergy was calculated using SynergyFinder 3.0, as described in: lanevski, A., Giri, K. A., Aittokallio, T., 2022. SynergyFinder 3.0: an interactive analysis and consensus interpretation of multi-drug synergies across multiple samples. Nucleic Acids Research. gkac382.

[0350] Titrations of darapladib, RSL3 and combinations were provided as input data to calculate percentage viability and synergy. A four-parameter logistic regression curvefitting algorithm was used to fit single-dose response curves. Synergy scores were

61

SUBSTITUTE SHEET (RULE 26) for viability. FIG. 25 shows statistically-significant dependence of sensitivity to Erastin on expression level of PLA2G7A.

[0346] EXAMPLE 5: Assays of Synergy with PLA2G7 Inhibitors

[0347] Assays to determine synergy of inhibitors of PLA2G7 (e.g., by darapladib) in combination with modulators of oxidative stress, modulators of ferroptosis, and chemotherapeutic agents were designed and tested as shown and described for FIGs. 26-56 and TABLE 6. a. Combination of Darapladib and Modulators of Oxidative Stress

[0348] FIGs. 26-33 show heatmap plots of the tumor-derived PANC-1 cell viability assessed as a function of inhibition of PLA2G7 (e.g., by darapladib) in combination with various modulators. Table 6 shows synergy with darapladib was observed with Erastin (FIG. 26), FIN56 (FIG. 27) and H 2 O 2 , (FIG. 33).

TABLE 6

[0349] Synergy Analysis. Drug synergy was calculated using SynergyFinder 3.0, as described in: lanevski, A., Giri, K. A., Aittokallio, T., 2022. SynergyFinder 3.0: an interactive analysis and consensus interpretation of multi-drug synergies across multiple samples. Nucleic Acids Research. gkac382.

[0350] Titrations of darapladib, RSL3 and combinations were provided as input data to calculate percentage viability and synergy. A four-parameter logistic regression curvefitting algorithm was used to fit single-dose response curves. Synergy scores were calculated using the Bliss reference model, which assumed a stochastic process in which two drugs elicit effects independently, and the expected combination effect was be calculated based on the probability of independent events. b. Combination of Darapladib and Modulators of Ferroptosis

[0351] The tumor-derived cell lines were assessed as a function of inhibition of PLA2G7 (e.g., by darapladib) in combination with various modulators of ferroptosis.

[0352] Cell viability was assessed according to methods described in Example 4(c), where 1 pM ferrostatinl was used. In assays combining darapladib and RSL3, cells were treated with the same concentration of both RSL3 and darapladib. Immunoblots were performed and assessed according to methods described in Example 4(a). FIG. 34 shows increased cytotoxicity resulting in decreased in cell viability of various cells with increasing concentrations of darapladib. FIG. 35 shows increased cytotoxicity resulting in decreased in cell viability of cells with increasing concentrations of ferroptosis inducer, RSL-3. FIG. 36 shows increased cytotoxicity resulting in decreased in cell viability of various cells with increasing concentrations of darapladib in the presence of RSL-3.

[0353] FIG. 37 shows cell viability of PANC-1 cells as a function of Darapladib, RSL-3, Darapladib and RSL3, Darapladib and ferroptosis inhibitor Ferrostatin, and Darapladib with RSL3 and Ferrostatin. FIG. 38 shows cell viability of A549 cells as a function of Darapladib, RSL-3, Darapladib and RSL3, Darapladib and Ferrostatin, and Darapladib with RSL3 and Ferrostatin. FIG. 39 shows upstream and downstream protein expression following treatment of A549 under the conditions in FIG. 38.

[0354] These experiments showed that cytotoxicity caused by darapladib in cells under oxidative stress occurred through ferroptosis.

[0355] FIGs. 40-48 show heatmap plots for various cell lines, showing synergy between Darapladib and RSL-3, and rescue via the addition of 1 pM Ferrostatin. Synergy was assessed according to methods described in Example 5(a). c. Combination of Darapladib and Chemotherapeutics

[0356] The tumor-derived cell lines were assessed as a function of inhibition of PLA2G7 (e.g., by darapladib) in combination with various chemotherapeutics (e.g., gemcitabine and vinorelbine).

[0357] FIGs. 49-51 shows cytotoxicity synergy in various cells comparing effects on cell viability with combination of darapladib and gemcitabine, to the effect of each component ( darapladib or gemcitabine) alone. Cytotoxicity was directly quantified following the manufacturer’s protocol. Darapladib concentration was 300 nM and gemcitabine concentration was 10 pM. Mono was the cytotoxicity following single drug exposure. Combo is the toxicity following a combination of darapladib with the chemotherapeutic agent. Cytotoxic synergy is defined as the difference between combo and mono. Statistical analysis was performed using a two-tailed t-test. FIGs. 52-54 show cytotoxic synergy of darapladib with chemotherapeutics gemcitabine and vinorelbine. Cell viability was assessed according to methods described in Example 4(c). Darapladib concentration was 300 nM, gemcitabine concentration was 10 pM, and vinorelbine concentration was 10 pM. .

[0358] FIGs. 55-56 show immunoblots of SF268 and Panc-1 cells. In FIG. 55, cell lines were treated with lipofectamine and siRNA to deliver interfering RNA to knock down the targets listed across the top of the gel. In FIG. 56, top panel shows immunoblots of SF268 and PANC-1 cells lines treated with additional Fenton substrates in media (-F), or with added Fenton and polyunsaturated fatty acids F(PUFA). Bottom panel shows immunoblots following the addition of PUFA substrates in the absence of a Fenton reagent.

[0359] before and after treatment with substrates. Immunoblots were performed and assessed according to methods described in Example 4(a).

[0360] EXAMPLE 6: Assays of PLA2G7 a. Combination of Rilapladib and Modulators of Ferroptosis

[0361] Assays to determine cytotoxicity of inhibitors of PLA2G7 (e.g., by rilapladib) in combination with modulators of ferroptosis were designed and tested as shown and described for in FIGs. 57-59, which show cell death due to the action of Rilapladib, RSL-3, combination of Rilapladib and RSL-3, and rescue via the addition of Ferrostatin in (A) SF268, (B) A549 and (C) PANC-1 cells. Cell viability was assessed according to methods described in Example 4(c). In the Rilapladib and RSL3 combination treatment, cells were treated with equal concentrations of Rilapladib and RSL3. In the rescue experiment, 1 pM of ferrostatin 1 was used.

[0362] These experiments showed that rilapladib inhibits PLA2G7 and induces cell death via ferroptosis. b. Modulation of PLA2G7

[0363] Assays to determine modulation of PLA2G7 expression levels by modulator of oxidative stress and other targets were designed and tested as shown and described for in FIGs. 60-61, which shows immunoblots indicating control of PLA2G7 expression level by a modulator of oxidative stress, nuclear factor erythroid 2-related factor 2 (NRF2), and other antioxidant and lipid metabolic targets. Immunoblots were performed and assessed according to methods described in Example 4(a).

[0364] These experiments showed that PLA2G7 expression levels were controlled by NRF2 and other targets.

EMBODIMENTS

[0365] The following non-limiting embodiments provide illustrative examples of the disclosure, but do not limit the scope of the disclosure.

[0366] Embodiment 1. A method of treating a condition, the method comprising administering to a subject in need thereof a therapeutically-effective amount of a compound that inhibits PLA2G7 and a reduced amount of a second therapy, wherein the reduced amount of the second therapy is therapeutically effective for treating the condition in combination with the therapeutically-effective amount of the compound that inhibits PLA2G7, and wherein the reduced amount of the second therapy is less than an amount of the second therapy that is therapeutically effective for the condition in absence of the therapeutically-effective amount of the compound that inhibits PLA2G7.

[0367] Embodiment 2. The method of embodiment 1, wherein the compound that inhibits PLA2G7 is darapladib.

[0368] Embodiment 3. The method of embodiment 1, wherein the compound that inhibits PLA2G7 is rilapladib.

[0369] Embodiment 4. The method of embodiment 1, wherein the compound that inhibits PLA2G7 is AA39-2.

[0370] Embodiment 5. The method of embodiment 1, wherein the compound that inhibits PLA2G7 is ML256.

[0371] Embodiment 6. The method of embodiment 1, wherein the compound that inhibits PLA2G7 inhibits PAFAH2.

[0372] Embodiment 7. A method of treating a condition, the method comprising administering to a subject in need thereof a therapeutically-effective amount of a compound that inhibits PAFAH2 and a reduced amount of a second therapy, wherein the reduced amount of the second therapy is therapeutically effective for treating the condition in combination with the therapeutically-effective amount of the compound that inhibits PAFAH2, and wherein the reduced amount of the second therapy is less than an amount of the second therapy that is therapeutically effective for the condition in absence of the therapeutically-effective amount of the compound that inhibits PAFAH2.

[0373] Embodiment 8. The method of embodiment 7, wherein the compound that inhibits PLA2G7 is darapladib.

[0374] Embodiment 9. The method of embodiment 7, wherein the compound that inhibits PLA2G7 is rilapladib. [0375] Embodiment 10. The method of embodiment 7, wherein the compound that inhibits PLA2G7 is AA39-2.

[0376] Embodiment 11. The method of embodiment 7, wherein the compound that inhibits PLA2G7 is ML256.

[0377] Embodiment 12. The method of embodiment 7, wherein the compound that inhibits PAFAH2 inhibits PLA2G7.

[0378] Embodiment 13. The method of any one of embodiments 1-12, wherein the second therapy comprises a chemotherapeutic agent or radiation therapy.

[0379] Embodiment 14. The method of any one of embodiments 1-13, further comprising administering to the subject a therapeutically-effective amount of a ferroptosis inducer.

[0380] Embodiment 15. The method of embodiment 14, wherein the ferroptosis inducer exhibits synergy with the compound that inhibits PLA2G7.

[0381] Embodiment 16. The method of embodiment 14, wherein the ferroptosis inducer exhibits synergy with the compound that inhibits PAFAH2.

[0382] Embodiment 17. The method of embodiment 14, wherein the ferroptosis inducer is a cystine-glutamate system Xc- inhibitor.

[0383] Embodiment 18. The method of embodiment 14, wherein the ferroptosis inducer is a glutathione peroxidase (GPX) inhibitor.

[0384] Embodiment 19. The method of any one of embodiments 1-18, further comprising administering to the subject a therapeutically-effective amount of a modulator of oxidative stress.

[0385] Embodiment 20. The method of any one of embodiments 1-19, wherein the reduced amount of the second therapy comprises administering a reduced dosage of the second therapy to the subject.

[0386] Embodiment 21. The method of any one of embodiments 1-19, wherein the reduced amount of the second therapy comprises administering the second therapy to the subject at a reduced frequency.

[0387] Embodiment 22. The method of any one of embodiments 1-19, wherein the reduced amount of the second therapy comprises administering a reduced dosage of the second therapy to the subject at a reduced frequency.

[0388] Embodiment 23. A method of treating a condition, the method comprising administering to a subject in need thereof a therapeutically-effective amount of a first compound that inhibits PLA2G7 and a therapeutically-effective amount of a second compound that exhibits synergy with the first compound that inhibits PLA2G7 for the condition.

[0389] Embodiment 24. A method of treating a condition, the method comprising administering to a subject in need thereof a therapeutically-effective amount of a first compound that inhibits PAFAH2 and a therapeutically-effective amount of a second compound that exhibits synergy with the first compound that inhibits PAFAH2 for the condition

[0390] Embodiment 25. The method of embodiment 23 or 24, wherein the second compound comprises a ferroptosis inducer.

[0391] Embodiment 26. The method of embodiment 25, wherein the ferroptosis inducer is a cystine-glutamate system Xc- inhibitor.

[0392] Embodiment 27. The method of embodiment 25, wherein the ferroptosis inducer is a glutathione peroxidase (GPX) inhibitor.

[0393] Embodiment 28. The method of embodiment 23 or 24, wherein the second compound comprises a modulator of oxidative stress.

[0394] Embodiment 29. The method of any one of embodiments 1-28, wherein the condition is cancer.

[0395] Embodiment 30. The method of embodiment 29, wherein the cancer harbors a RAS mutation.

[0396] Embodiment 31. The method of embodiment 30, wherein the RAS mutation is KRAS.

[0397] Embodiment 32. The method of embodiment 30, wherein the RAS mutation is NRAS.

[0398] Embodiment 33. The method of embodiment 30, wherein the RAS mutation is HR AS.

[0399] Embodiment 34. The method of any one of embodiments 29-33, wherein the RAS mutation is a constitutively active variant.

[0400] Embodiment 35. The method of embodiment 34, wherein the constitutively active variant is a gain of function mutation.

[0401] Embodiment 36. The method of embodiment 34, wherein the constitutively active variant is a duplication.

[0402] Embodiment 37. The method of embodiment 34, wherein the constitutively active variant is a gene amplification. Embodiment 38. The method of embodiment 29, wherein the cancer harbors an STK11 mutation. [0403] Embodiment 39. The method of embodiment 29, wherein the cancer harbors an KE API mutation.

[0404] Embodiment 40. The method of embodiment 29, wherein the cancer harbors an STK11 mutation an KE API mutation.

[0405] Embodiment 4E The method of any one of embodiments 29-37, wherein the cancer further harbors an STK11 mutation.

[0406] Embodiment 42. The method of any one of embodiments 29-37, wherein the cancer further harbors an KEAP 1.

[0407] Embodiment 43. The method of any one of embodiments 29-37, wherein the cancer further harbors an STK11 mutation an KEAP1 mutation.

[0408] Embodiment 44. The method of any one of embodiments 29-43, wherein the cancer cells from the subject exhibit elevated expression or activity of PLA2G7 compared to healthy control cells.

[0409] Embodiment 45. The method of any one of embodiments 29-43, wherein the cancer cells from the subject exhibit elevated expression or activity of PAFAH2 compared to healthy control cells.

[0410] Embodiment 46. The method of any one of embodiments 29-45, wherein the cancer is lung cancer.

[0411] Embodiment 47. The method of any one of embodiments 29-45, wherein the cancer is lymphoma.

[0412] Embodiment 48. The method of any one of embodiments 29-45, wherein the cancer is non-small cell lung.

[0413] Embodiment 49. The method of any one of embodiments 29-45, wherein the cancer is pancreatic cancer.

[0414] Embodiment 50. The method of any one of embodiments 29-45, wherein the cancer is clear-cell renal cell carcinoma.

[0415] Embodiment 51. The method of any one of embodiments 29-45, wherein the cancer is diffuse large B-cell lymphoma.

[0416] Embodiment 52. The method of any one of embodiments 29-45, wherein the cancer is leukemia.

[0417] Embodiment 53. The method of any one of embodiments 29-45, wherein the cancer is astrocytoma.

[0418] Embodiment 54. The method of any one of embodiments 1-53, wherein the administering comprises oral administration. [0419] Embodiment 55. The method of any one of embodiments 1-53, wherein the administering intravenous administration.

[0420] Embodiment 56. The method of any one of embodiments 1-53, wherein the administering intramuscular administration.

[0421] Embodiment 57. A method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a non-wild-type RAS gene; and b) based on the determining that the subject possesses the non-wild type RAS gene, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0422] Embodiment 58. A method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses an elevated level of PLA2G7 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the elevated level of PLA2G7 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0423] Embodiment 59. A method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a decreased level of PLA2G7 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the decreased level of PLA2G7 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0424] Embodiment 60. A method of treating a cancer in a subject in need thereof, the method comprising: a) determining that the cancer in the subject possesses a cell line in which PLA2G7 inhibits ferroptotic cell death; and b) based on the determining that the cancer in the subject possesses a cell line in which PLA2G7 inhibits ferroptotic cell death, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0425] Embodiment 61. A method of treating a condition in a tissue in a subject in need thereof, the method comprising: a) determining that the tissue suffers from oxidative stress; and b) based on the determining that the tissue suffers from oxidative stress, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0426] Embodiment 62. A method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject exhibits a marker associated with the condition, wherein the condition is associated with PLA2G7 activity; and b) based on the determining that the subject exhibits the marker associated with the condition, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7. [0427] Embodiment 63. A method of treating a condition in a subject in need thereof, the method comprising: a) determining based on an assay that the condition is amenable to treatment by a therapeutic agent that synergizes with a compound that inhibits PLA2G7 for the condition; and b) based on the determining that the condition is amenable to treatment by the therapeutic agent that synergizes with the compound that inhibits PLA2G7 for the condition, administering to the subject a therapeutically-effective amount of a compound that inhibits PLA2G7.

[0428] Embodiment 64. The method of any one of embodiments 57-63, wherein the compound that inhibits PLA2G7 is darapladib.

[0429] Embodiment 65. The method of any one of embodiments 57-63, wherein the compound that inhibits PLA2G7 is rilapladib.

[0430] Embodiment 66. The method of any one of embodiments 57-63, wherein the compound that inhibits PLA2G7 is AA39-2.

[0431] Embodiment 67. The method of any one of embodiments 57-63, wherein the compound that inhibits PLA2G7 is ML256.

[0432] Embodiment 68. The method of any one of embodiments 57-63, wherein the compound that inhibits PLA2G7 inhibits PAFAH2

[0433] Embodiment 69. A method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a non-wild type RAS gene; and b) based on the determining that the subject possesses the non-wild type RAS gene, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2.

[0434] Embodiment 70. A method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses an elevated level of PAFAH2 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the elevated level of PAFAH2 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2.Embodiment 71. A method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject possesses a decreased level of PAFAH2 in comparison to a predetermined threshold value; and b) based on the determining that the subject possesses the decreased level of PAFAH2 in comparison to the predetermined threshold value, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2.

[0435] Embodiment 72. A method of treating a cancer in a subject in need thereof, the method comprising: a) determining that the cancer in the subject possesses a cell line in which PAFAH2 inhibits ferroptotic cell death; and b) based on the determining that the cancer in the subject possesses a cell line in which PAFAH2 inhibits ferroptotic cell death, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2.

[0436] Embodiment 73. A method of treating a condition in a tissue in a subject in need thereof, the method comprising: a) determining that the tissue suffers from oxidative stress; and b) based on the determining that the tissue suffers from oxidative stress, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2. [0437] Embodiment 74. A method of treating a condition in a subject in need thereof, the method comprising: a) determining that the subject exhibits a marker associated with the condition, wherein the condition is associated with PAFAH2 activity; and b) based on the determining that the subject exhibits the marker associated with the condition, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2.

[0438] Embodiment 75. A method of treating a condition in a subject in need thereof, the method comprising: a) determining based on an assay that the condition is amenable to treatment by a therapeutic agent that synergizes with a compound that inhibits PAFAH2 for the condition; and b) based on the determining that the condition is amenable to treatment by the therapeutic agent that synergizes with the compound that inhibits PAFAH2 for the condition, administering to the subject a therapeutically-effective amount of a compound that inhibits PAFAH2.

[0439] Embodiment 76. The method of any one of embodiments 69-75, wherein the compound that inhibits PAFAH2 is darapladib.

[0440] Embodiment 77. The method of any one of embodiments 69-75, wherein the compound that inhibits PAFAH2 is rilapladib.

[0441] Embodiment 78. The method of any one of embodiments 69-75, wherein the compound that inhibits PAFAH2 is AA39-2.

[0442] Embodiment 79. The method of any one of embodiments 69-75, wherein the compound that inhibits PAFAH2 is ML256.

[0443] Embodiment 80. The method of embodiments 69-75, wherein the compound that inhibits PAFAH2 inhibits PLA2G7. [0444] Embodiment 81. The method of any one of embodiments 57-80, further comprising administering to the subject therapeutically-effective amount of a second therapy.

[0445] Embodiment 82. The method of embodiment 81, wherein the second therapy exhibits synergy with the compound that inhibits PLA2G7.

[0446] Embodiment 83. The method of embodiment 81, wherein the second therapy exhibits synergy with the compound that inhibits PAFAH2.

[0447] Embodiment 84. The method of embodiment 81, wherein the second therapy is a therapeutically-effective amount of a ferroptosis inducer.

[0448] Embodiment 85. The method of embodiment 84, wherein the ferroptosis inducer is a cystine-glutamate system Xc- inhibitor.

[0449] Embodiment 86. The method of embodiment 84, wherein the ferroptosis inducer is a glutathione peroxidase (GPX) inhibitor.

[0450] Embodiment 87. The method of embodiment 81, wherein the second therapy is a therapeutically-effective amount of a modulator of oxidative stress.

[0451] Embodiment 88. A method of treating a cancer in a subject in need thereof, the method comprising: a) determining that the cancer in the subject possesses a cell line in which PLA2G7 inhibits ferroptotic cell death; and b) based on the determining that the cancer in the subject possesses a cell line in which PLA2G7 inhibits ferroptotic cell death, administering to the subject a therapeutically-effective amount of a compound that operates by a mechanism of action that synergizes with ferroptotic cell death.

[0452] Embodiment 89. The method of embodiment 88, wherein the compound that synergizes with ferroptotic cell death is a compound that inhibits PLA2G7.

[0453] Embodiment 90. A method of treating a cancer in a subject in need thereof, the method comprising: a) determining that the cancer in the subject possesses a cell line in which PAFAH2 inhibits ferroptotic cell death; and b) based on the determining that the cancer in the subject possesses a cell line in which PAFAH2 inhibits ferroptotic cell death, administering to the subject a therapeutically-effective amount of a compound that operates by a mechanism of action that synergizes with ferroptotic cell death.

[0454] Embodiment 91. The method of embodiment 90, wherein the compound that synergizes with ferroptotic cell death is a compound that inhibits PAFAH2.

[0455] Embodiment 92. The method of any one of embodiments 88-91, further comprising administering to the subject a therapeutically-effective amount of a second therapy.

[0456] Embodiment 93. The method of embodiment 92, wherein the second therapy exhibits synergy with the compound that inhibits PLA2G7. [0457] Embodiment 94. The method of embodiment 92, wherein the second therapy exhibits synergy with the compound that inhibits PLA2G7.

[0458] Embodiment 95. The method of embodiment 92, wherein the second therapy is a therapeutically-effective amount of a ferroptosis inducer.

[0459] Embodiment 96. The method of embodiment 95, wherein the ferroptosis inducer is a cystine-glutamate system Xc- inhibitor.

[0460] Embodiment 97. The method of embodiment 95, wherein the ferroptosis inducer is a glutathione peroxidase (GPX) inhibitor.

[0461] Embodiment 98. The method of embodiment 92, wherein the second therapy is a therapeutically-effective amount of a modulator of oxidative stress.

[0462] Embodiment 99. A method comprising: a) determining that a subject exhibits a level of a biomarker, wherein the subject is undergoing a therapeutic regimen of a compound that inhibits PLA2G7; and b) based on the level of the biomarker, determining whether to continue the therapeutic regimen of a compound that inhibits PLA2G7.

[0463] Embodiment 100. The method of embodiment 99, wherein the biomarker is Lp- PLA2.

[0464] Embodiment 101. A method comprising: a) determining that a subject exhibits a level of a biomarker, wherein the subject is undergoing a therapeutic regimen of a compound that inhibits PAFAH2; and b) based on the level of the biomarker, determining whether to continue the therapeutic regimen of a compound that inhibits PAFAH2.

[0465] Embodiment 102. A method comprising determining based on an assay of a subject that the subject has at least a threshold number of genes, wherein the threshold number of genes indicates that a compound that inhibits PLA2G7 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of genes is five. [0466] Embodiment 103. A method comprising determining based on an assay of a subject that the subject has at least a threshold number of genes, wherein the threshold number of genes indicates that a compound that inhibits PAFAH2 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of genes is five. [0467] Embodiment 104. A method comprising determining based on an assay of a subject that the subject expresses at least a threshold number of phospholipases, wherein the threshold number of phospholipases indicates that a compound that inhibits PLA2G7 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of phospholipases is five. [0468] Embodiment 105. A method comprising determining based on an assay of a subject that the subject expresses at least a threshold number of phospholipases, wherein the threshold number of phospholipases indicates that a compound that inhibits PAFAH2 is therapeutically effective for treating a condition that the subject exhibits, wherein the threshold number of phospholipases is five.

[0469] While certain embodiments have been illustrated and described, a person with ordinary skill in the art, after reading the foregoing specification, can effect changes, substitutions of equivalents and other types of alterations. Each aspect and embodiment described above can also have included or incorporated therewith such variations or aspects as disclosed in regard to any or all of the other aspects and embodiments.

[0470] The present technology is also not to be limited in terms of the particular aspects described herein, which are intended as single illustrations of individual aspects of the present technology. Many modifications and variations of this present technology can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. Functionally equivalent methods within the scope of the present technology, in addition to those enumerated herein, will be apparent to those skilled in the art from the foregoing descriptions. Such modifications and variations are intended to fall within the scope of the appended claims. It is to be understood that this present technology is not limited to particular methods, reagents, compounds, compositions, labeled compounds or biological systems, which can, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular aspects only, and is not intended to be limiting. Thus, it is intended that the specification be considered as exemplary only with the breadth, scope and spirit of the present technology indicated only by the appended claims, definitions therein, and any equivalents thereof.

[0471] The embodiments, illustratively described herein may suitably be practiced in the absence of any element or elements, limitation or limitations, not specifically disclosed herein. Thus, for example, the terms “comprising,” “including,” “containing,” etc. shall be read expansively and without limitation. Additionally, the terms and expressions employed herein have been used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the claimed technology. Additionally, the phrase “consisting essentially of’ will be understood to include those elements specifically recited and those additional elements that do not materially affect the basic and novel characteristics of the claimed technology. The phrase “consisting of’ excludes any element not specified. [0472] In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group. Each of the narrower species and subgeneric groupings falling within the generic disclosure also form part of the disclosure. This includes the generic description of the disclosure with a proviso or negative limitation removing any subject matter from the genus, regardless of whether or not the excised material is specifically recited herein.