Small molecule ferroptosis inducers

Information

  • Patent Grant
  • 10947188
  • Patent Number
    10,947,188
  • Date Filed
    Tuesday, June 18, 2019
    5 years ago
  • Date Issued
    Tuesday, March 16, 2021
    3 years ago
Abstract
The present invention provides, inter alia, a compound according to formula (I):
Description
COPYRIGHT NOTICE

A portion of the disclosure of this patent document contains material, which is subject to copyright protection. The copyright owner has no objection to the facsimile reproduction by anyone of the patent document or the patent disclosure, as it appears in the Patent and Trademark Office patent files or records, but otherwise reserves all copyright rights whatsoever.


BACKGROUND

Ferroptosis is a non-apoptotic form of regulated cell death. Its relevance to certain pathological conditions has been reported, such as periventricular leukomalacia, nephrotic tubular death, and Huntington's disease (Skouta et al. 2014, Linkermann et al. 2014). It is distinct from other regulated cell death phenotypes, such as apoptosis and necroptosis (Berghe et al. 2014, Dixon et al. 2014). Ferroptosis is characterized by extensive lipid peroxidation, which can be suppressed by iron chelators or lipophilic antioxidants. Mechanistically, ferroptosis inducers are divided into two classes: (1) inhibitors of cystine import via system xc (e.g., erastin) (Dixon et al. 2012, Dixon et al. 2014), which subsequently causes depletion of glutathione (GSH) (Hayano et al. 2015), and (2) covalent inhibitors (e.g., (1S, 3R)-RSL3) of glutathione peroxidase 4 (GPX4) (Yang et al. 2014). Since GPX4 reduces lipid hydroperoxides using GSH as a co-substrate (Imai et al. 2003), both compound classes ultimately result in loss of GPX4 activity, followed by elevated levels of lipid reactive oxygen species (ROS) and consequent cell death.


There is some crosstalk among distinct regulated cell death phenotypes (Berghe et al. 2014, Linkermann et al. 2014). Each proposed cell death phenotype has generally been studied using different models (Hitomi et al. 2008). However, a universal comparison of different cell death phenotypes would be highly beneficial to understand the mechanisms governing cell death.


In view of the foregoing, there exists an ongoing need to provide new and improved compounds for regulating GPX4 and inducing ferroptosis in cells. The present disclosure is directed towards solving this and other needs.


SUMMARY

One embodiment of the present invention is a compound according to formula (I):




embedded image



wherein:

    • R1 is selected from the group consisting of H, OH, and —(OCH2CH2)xOH;
    • X is an integer from 1 to 6; and
    • R2, R2′, R3, and R3′ independently are selected from the group consisting of H, C3-8cycloalkyl, and combinations thereof, or R2 and R2′ may be joined together to form a pyridinyl or pyranyl and R3 and R3′ may be joined together to form a pyridinyl or pyranyl;


      or an N-oxide, crystalline form, hydrate, or pharmaceutically acceptable salt thereof; with the proviso that the compound is not




embedded image


Another embodiment of the present invention is a method for inducing ferroptosis in a cell. This method comprises contacting the cell with an effective amount of a compound having the structure of formula (I):




embedded image



wherein:

    • R1 is selected from the group consisting of H, OH, and —(OCH2CH2)xOH;
    • X is an integer from 1 to 6; and
    • R2, R2′, R3, and R3′ independently are selected from the group consisting of H, C3-8cycloalkyl, and combinations thereof, or R2 and R2′ may be joined together to form a pyridinyl or pyranyl and R3 and R3′ may be joined together to form a pyridinyl or pyranyl;


      or an N oxide, crystalline form, hydrate, or pharmaceutically acceptable salt thereof.


An additional embodiment of the present invention is a method for decreasing GPX4 in a cell. This method comprising contacting the cell with an effective amount of a compound having the structure of formula (I):




embedded image



wherein:

    • R1 is selected from the group consisting of H, OH, and —(OCH2CH2)xOH;
    • X is an integer from 1 to 6; and
    • R2, R2′, R3, and R3′ independently are selected from the group consisting of H, C3-8cycloalkyl, and combinations thereof, or R2 and R2′ may be joined together to form a pyridinyl or pyranyl and R3 and R3′ may be joined together to form a pyridinyl or pyranyl;


      or an N-oxide, crystalline form, hydrate, or pharmaceutically acceptable salt thereof.





BRIEF DESCRIPTION OF THE DRAWINGS

The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.



FIGS. 1A-1B show that modulatory profiling revealed three types of regulated non-apoptotic cell death.



FIG. 1A shows the experimental scheme to identify regulated non-apoptotic cell death inducers with high modulatability. The numbers in red indicate the number of compounds satisfying each criterion.



FIG. 1B shows the hierarchical clustering of modulatory profiles of 10 CILs with high modulatability and 30 characterized lethal compounds from several classes of lethal mechanisms. Lethal compounds are listed on the right. Ten CILs are indicated by red shading. Forty-six modulators are listed along the bottom (28 death modulators in two cell lines, HT-1080 and BJeLR). Antioxidants (anti-ox) and iron chelators (Fe-chel) are indicated by brown shading. MoA, mechanism of action; Topo, topoisomerase.



FIGS. 2A-2F show that optimization of CIL56 revealed a potent and selective ferroptosis inducer.



FIG. 2A and FIG. 2E show the HRASG12V selectivity. Viability of four engineered BJ cell lines treated with CIL56 (FIG. 2A) or FIN56 (FIG. 2E) for 48 h. mut, cells tumor-transformed as a result of HRASG12V overexpression; wt, isogenic cells without HRASG12V.



FIG. 2B shows the lipid RoS generation. Flow cytometry analysis with BODIPY-581/591 C11 staining in HT-1080 cells incubated with test compounds for 6 h. DFOM, 152 μM deferoxamine.



FIG. 2C and FIG. 2F show the effects of ferroptosis inhibitors on viability of HT-1080 cells cotreated with CIL56 (FIG. 2C) or FIN56 (FIG. 2F) for 48 h. αtoc, 100 μM α-tocopherol; U0126 was applied at 3.8 μM.



FIG. 2D shows the chemical structures of CIL56 and FIN56.


Experiments for FIGS. 2A-2F were performed in biological triplicate, and data are presented as mean±s.e.m. of technical triplicates.



FIGS. 3A-3F show that FIN56-induced ferroptosis decreases GPX4 expression.



FIG. 3A shows the GPX4 enzymatic activity in BJeLR cells after ferroptosis-inducer treatment. GSSG, oxidized glutathione; PC, phosphatidylcholine.



FIG. 3B shows the kinetics of ROS generation after treatment with 0.5 μM (1S, 3R)-RSL3 or 5 μM FIN56, detected with 25 μM H2-DCFDA staining in BJeLR cells.



FIG. 3C shows the GPX4 protein abundance in BJeLR cells after cotreatment with 100 μM α-tocopherol and ferroptosis inducers for 10 h.



FIG. 3D shows the effect of shRNA targeting GPX4 (shGPX4) on FIN56-induced ferroptosis in BJeLR cells. shNT, nontargeting shRNA.



FIG. 3E shows the effects of GFP-GPX4 fusion-protein overexpression on sensitivity to FIN56 in BJeLR cells.



FIG. 3F shows the effects of GFP-GPX4 fusion-protein overexpression on endogenous and exogenous GPX4 protein abundance after FIN56 treatment.


Experiments in FIGS. 3A-3F were done in biological triplicate. Representative results are shown for FIG. 3A, FIG. 3B and FIG. 3E; data in FIG. 3C and FIG. 3F are the mean±s.e.m. of biological triplicates, in which *P<0.05, **P<0.005, ***P<0.0005 (paired two-tailed t-test); data in FIG. 3D are the mean±s.e.m. of technical triplicates.



FIGS. 4A-4D show the SQS encoded by FDFT1 as FIN56's target protein.



FIG. 4A shows the structures of active and inactive FIN56 analogs with PEG linkers.



FIG. 4B shows the effects of five shRNAs against FDFT1 on FIN56. Results in two of the four cell lines are shown. The black line in each graph (median AUC among tested shRNAs) represents a shRNA that had no effect in each cell line.



FIG. 4C shows the summary of proteomic target identification and effect of shRNA screening targeting 70 identified genes on FIN56. Each dot summarizes the result of multiple shRNAs targeting a gene. Each shRNA was considered ‘consistent’ when it exerted the indicated effect (enhancing or suppressing FIN56). X-axis represents the ratio of the number of consistent shRNAs inducing the indicated effect (i.e., enhancing or suppressing FIN56) to the total number of shRNAs targeting the gene. Y-axis shows fold enrichment of protein abundance on active versus inactive probe beads in a pulldown assay.



FIG. 4D shows the effect of siRNAs against loss-of-function candidates on BJeLR cell viability. Cells were grown in the presence of DMSO or α-tocopherol.


shRNA screens in FIG. 4B and FIG. 4C were performed once in four cell lines. The siRNA experiment in FIG. 4D was performed in BJeLR twice, and results indicate the mean of biological replicates.



FIGS. 5A-5H show the results of validating SQS as the functionally relevant target for FIN56's lethality.



FIG. 5A shows the effects of chemical inhibitors of SQS on FIN56's lethality.



FIG. 5B shows the effects of FPP on FIN56's lethality.



FIG. 5C shows the detection of SQS via pulldown assay from HT-1080 whole-cell lysate with active or inactive probes. Note that the probes are the same as those used for chemoproteomic target identification.



FIG. 5D shows the schematic of the mevalonate pathway. The larger font indicates metabolites, and the smaller font indicates enzymes responsible for the reactions or small molecules. Red or blue text indicates molecules (inhibitors or metabolites) that suppressed or enhanced FIN56's lethality, respectively. The detailed results are shown in FIG. 5E and FIG. 5F.



FIG. 5E shows the perturbation of the mevalonate pathway and effects on FIN56's lethality. Concentrations: cerivastatin, 1 μM; metabolites, 100 μM; YM-53601, 5 μM; NB-598, 25 μM; α-tocopherol, 100 μM.



FIG. 5F shows the supplementation of 10 μM end-products of the MVA pathway and their effects on FIN56.



FIG. 5G shows the effect of 10 μM idebenone on FIN56 in HT-1080 cells.



FIG. 5H shows the modulatory profiling between the modulators of the MVA pathway and various lethal compounds inducing oxidative stress. Enh, enhancement; sup., suppression; IPP, isopentenyl-pyrophosphate (PP); DMAPP, dimethylallyl-PP; GPP, geranyl-PP; FPP, farnesyl-PP; GGPP, geranylgeranyl-PP.


Experiments shown in FIG. 5A, FIG. 5B and FIG. 5G were performed in biological replicates, and data are presented as mean±s.e.m. of technical triplicates; experiments shown in FIG. 5E and FIG. 5F were performed in biological duplicates, and data are presented as mean±s.e. of EC50 estimation from sigmoidal curve-fitting; experiment shown in FIG. 5H was performed in singlicate.



FIGS. 6A-6D show that ACC inhibitor prevents GPX4 protein degradation.



FIG. 6A shows the effects of mevalonate pathway modulators and ACC inhibitor on GPX4 abundance with 0.1% DMSO (green) or 5 μM FIN56 (yellow). αTub, α-tubulin. *P<0.05, †† P<0.001 (paired two-tailed t-test); n.s., not significant. Modulator concentrations: DMSO, 0.1%; FPP, 10 μM; YM-53601, 5 μM; idebenone, 10 μM; TOFA, 10 μM.



FIG. 6B shows the lipid peroxide levels after treatment with DMSO, TOFA or α-tocopherol.



FIG. 6C shows the effects of TOFA and α-tocopherol on FIN56 lethality.



FIG. 6D shows the model of FIN56's mechanism of action. Dashed arrows indicate that the mechanistic details are still elusive.


Experiments for FIGS. 6A-6C were performed in biological triplicates. Data in FIG. 6A and FIG. 6C are presented as the mean±s.e.m. of triplicates; single representative results are shown in FIG. 6B.



FIGS. 7A-7D show the screening for caspase-3/7 independent lethal compounds (CIL).



FIG. 7A shows the caspase-3/7 activation assay (Apo-ONE Homogeneous caspase-3/7Assay). Six apopototic and two non-apoptotic inducers were tested to optimize the assay conditions for screening. Grey area represents normalized viability 24 or 48 hrs after lethal compound treatment, and the colored lines indicate Apo-ONE fluorescent signals after indicated time points. Vertical orange lines indicate concentrations shown in FIG. 7B.



FIG. 7B shows the kinetics of Apo-ONE fluorescence intensity upon lethal compound treatment.



FIG. 7C shows the summary of chemical screening of 3,169 compounds for caspase-independent lethality in two cell lines, HT-1080 and BJeLR cells. Blue dots represent CILs.



FIG. 7D shows the statistical summary of screening. The numbers of compounds lethal in both HT-1080 and BJeLR cells and/or activating caspase-3/7 are shown.


Viability and Caspase-activity in FIG. 7A were tested in duplicates; representative results were shown. Screening in FIG. 7C was performed once. Plots in FIG. 7B and FIG. 7D were generated from FIG. 7A and FIG. 7C, respectively.



FIGS. 8A-8E show the modulatory profiling scheme and modulatability.



FIG. 8A shows the scheme of the modulatory profiling experiment. Two cell lines (HT-1080 and BJeLR) were co-treated with a lethal compound and a death modulator.



FIG. 8B shows that the modulator (M)'s effect (EMIL) on the lethal compound (L) was assessed by computing a difference of areas under two dose-response curves (EM|L=AUCmodulator−AUCvehicle).



FIG. 8C shows relationship between modulatory profiles and mechanism of action. In Wolpaw et al. 2011, modulatory profiles of lethals were correlated with their mechanisms of actions.



FIG. 8D shows the modulatability of 56 CILs and 30 characterized compounds. The dashed line represents the median value of the modulatability of characterized compounds. Ten CILs above the line (red dots) were defined as CILs with high modulatability, which were expected to activate specific pathway(s) to induce cell death.



FIG. 8E shows the scheme describing how modulatability of lethal compounds is associated with specific regulated cell-death.


Plot in FIG. 8D was generated from the modulatory profiles (see FIG. 1B) and done once.



FIGS. 9A-9E show that CILs with high modulatability were functionally clustered into three classes.



FIG. 9A shows the chemical structures of the ten CILs with high modulatability. The background colors of the compounds correspond to the clusters in FIG. 1B.



FIG. 9B shows the changes in absorbance spectrum of each compound upon cobalt (II) supplementation in vitro.



FIG. 9C shows the suppression of CIL13, 52 and 64 with the supplementation of CoCl2.



FIG. 9D shows that 19 μM (5 μg/mL) Necrostatin-1 (Nec-1) suppressed CIL62.



FIG. 9E shows that both CIL62 (red) and ferroptosis inducers (orange) were suppressed by 19 μM Nec-1, but CIL62 was not suppressed by the ferroptosis inducers (antioxidants or iron-chelators).


Measurements in FIG. 9B was done once; experiments in FIGS. 9C-9E were extracted from the modulatory profiles (see FIG. 1B). Error bars in FIG. 9C and FIG. 9D are s.e.m. of technical triplicates.



FIGS. 10A-10E show that CIL41/70 induce iron-dependent oxidative stress but not oncogenic-RAS selective in the BJ series. FIG. 10A shows the properties of 6 CILs (41, 56, 69, 70, 75, 79).



FIG. 10B shows that CIL41/70 treatment caused H2-DCFDA-detectable ROS generation.



FIG. 10C shows the suppression of CIL41/70 lethality by ferroptosis inhibitors (the antioxidant α-tocopherol (αToc) and the iron-chelator deferoxamine (DFOM)).



FIG. 10D shows that CIL41/70 do not induce selective lethality in the BJ cell line panel.



FIG. 10E shows the EC50 of CIL56, CIL41/70 and the structural analogs of CIL41/70.


Experiments in FIG. 10B-10D were done in biological triplicates and single representative results are shown; error-bars indicate s.e.m. of technical triplicates; screening in FIG. 10E was done once.



FIG. 11 shows the structure activity relationship study of CIL56 and selective induction of ferroptosis. It shows the structural analogs of CIL56 and their capability of inducing ferroptosis. Piperidine rings and oxime group in CIL56 were substituted by different moieties. EC50 and selectivity (fold suppression by α-tocopherol) of the synthesized analogs were shown. EC50 and selectivity measurements were done in two biological replicates, three technical replicates each.



FIGS. 12A-12N show the mechanisms regulating GPX4 protein abundance.



FIG. 12A shows the cell-line-selectivity of GSH synthesis inhibitors, GPX4 inhibitors, and CIL56 analogs in the NCI60 cell line panel.



FIG. 12B shows the abundance of total glutathione (GSH and GSSG) in HT-1080 cells upon co-treatment with 100 μM αToc and ferroptosis inducers for ten hours. αToc: α-tocopherol.



FIG. 12C shows the GPX4 abundance in BJeLR cells upon 100 μM αToc and ferroptosis inducers. (Corresponding western to FIG. 3C)



FIG. 12D shows the results of GPX1 proteins in BJeLR cells upon co-treatment with 100 μM αToc and ferroptosis inducers for ten hours. GPX protein levels are normalized to α-tubulin protein levels within each sample.



FIG. 12E and FIG. 12F show the effects of GFP-GPX4 fusion protein overexpression on endogenous and exogenous GPX4 protein abundance with or without FIN56 treatment measured using (FIG. 12E) western (corresponding to FIG. 3F), and (FIG. 12F) FACS (green fluorescence).



FIG. 12G and FIG. 12H show the results of with or without pre-treatment of 100 nM selenium for 12 hrs and their effects on (FIG. 12G) FIN56 lethality in HT-1080 cells and (FIG. 12H) GPX4 abundance.



FIG. 12I shows the effects of translation inhibition (CHX) and proteosomal inhibition (MG132) on GPX4 abundance.



FIG. 12J shows the GPX4 transcripts level upon 5 μM FIN56.



FIG. 12K shows the effects of siRNA against GPX4 and TRIT1 on FIN56 lethality.



FIG. 12L shows the effects of GPX4 and TRIT1 knockdown on GPX4 protein level.



FIG. 12M shows the efficiency of knockdown assessed by RT-qPCR.



FIG. 12N shows that Pan-PIM inhibitors, CX-6258 and AZD-1208 were tested in 2-fold dilution series. Although they induce lethality in HT-1080 cells at higher concentrations, the lethality was not suppressed by αToc, indicating the death phenotype was not the same as ferroptosis.


In FIGS. 12C, 12D, 12E, 12H, and 12I, HT-1080 cells were co-treated with FIN56 and αToc. Experiments in FIGS. 12B-12N were done in biological triplicates; single representative results are shown and error-bars indicate s.e.m. of technical triplicates for FIGS. 12F, 12G, 12J, 12K, 12M and 12N; mean and s.e.m. of biological triplicates were shown for FIGS. 12B, 12C, 12D, 12E, 12H, 12I and 12L.



FIG. 13 shows the potency of CIL56 analogs in HT-1080. It shows the potency of CIL56 analogs (see FIG. 4A) in HT-1080 cells in 48 hrs. The experiment was done in biological triplicates. Error-bars indicate s.e.m. of technical triplicates.



FIGS. 14A-14D show the Interpretation of shRNA screen experiment targeting proteins identified in chemoproteomics.



FIG. 14A shows two possible scenarios when FIN56 binds to its target protein. Depending on the scenarios, expected effect of shRNAs against genuine targets on FIN56's lethality is opposite.



FIG. 14B shows the effects of shRNAs targeting 70 different proteins identified in chemoproteomics on FIN56. Two of the four cell lines are shown (See FIG. 4C for the other two). shFDFT1's were featured with polychromatic lines.



FIG. 14C shows the summary of the shRNA screens. Effects of shRNAs targeting the 70 proteins in the four cell lines. Each shRNA's effect in each cell line was represented by AUC, and ranked across the shRNAs in each cell line, The ranks were scaled between 0 and 100. Five shFDFT1 as well as two control shGPX4 are featured; shFDFT1 clones 326, 327, 328, and 325 are constantly within top 10, 20, 30, and 50 percentiles across the four cell lines.



FIG. 14D shows the relationship between the shRNA screen and the protein fold enrichment from the proteomic analysis. Top panel: the shRNA screen (different representation of FIG. 14C). The height of the barplot corresponds to the number of shRNAs targeting the gene. Colors of the segments indicate each shRNA's consistent effect (red—suppression, blue—enhancement, grey—not consistent). Bottom panel: fold enrichment of protein pull-down with active probe vs inactive ones in chemoproteomic analysis. Two plots (see FIG. 4B) show the same result as FIG. 14C, emphasizing the ratio between ‘#consistent enhancer or suppressor shRNAs’ vs ‘#shRNAs targeting each gene’. shRNA treatment and FIN56 treatment was done once in each cell line.



FIG. 15 shows the result of SQS pulldown and competition assay. SQS pull-down using purified truncated SQS, active probe and FIN56 as competitor. 190 μL of 10 ng/mL and 1 ng/mL purified truncated human SQS were preincubated with vehicle (DMSO) or 100 μM FIN56 for two hours, and further incubated with 5 μL of active probe for another two hours. Pull-down and western were done in biological triplicates and a representative result is shown.



FIGS. 16A-16D show the synergy between statins and FIN56.



FIG. 16A shows the synergy between three statins and FIN56 was measured and computed using Bliss independence.



FIG. 16B shows statins' lethality and specificity. Three statins were tested in dilution series in HT-1080 cells with or without mevalonolactone. Lethality of Simvastatin and Lovastatin was not suppressed by mevalonolactone, indicating that they interact with off-targets at higher concentrations while cerivastatin more selectively targets HMG-CoA reductase.



FIG. 16C shows the effects of 1 μM cerivastatin and/or 100 μM mevalonolactone (MVA) in HT-1080 cells.



FIG. 16D shows the effects of supplementing the mevalonate pathway modulators for 24 hrs on GPX4 protein level in HT-1080 cells.


Experiments in FIGS. 16A, 16C and 16D were done in biological triplicates; experiment in FIG. 16B was done in biological duplicate; FIG. 16A and FIG. 16C show single representative results; mean of the technical triplicates were shown for viability and Bliss computation in FIG. 16B; error bars are s.e.m. in FIG. 16C; mean and s.e.m. of the biological replicates were shown in FIG. 16D.



FIGS. 17A-17C show that modulators of the mevalonate pathway do not target respiration chain nor act as lipid antioxidant.



FIG. 17A shows 143B lung adenocarcinoma cells with or without mitochondrial DNA (ρ0 and ρ+ cells).



FIG. 17B shows the confirmation that mitochondrial DNA-encoded genes are not expressed.



FIG. 17C shows the lipid peroxide levels upon the mevalonate pathway modulator treatments and α-tocopherol.


Experiments in FIGS. 17A and 17B were done in biological singlicate and error bars are s.e.m. of three technical replicates; experiments in FIG. 17C was done in biological duplicate.



FIG. 18 shows the effects of the mevalonate pathway modulators and an ACC inhibitor on FIN56-induced GPX4 loss. It is a representative western blot of FIG. 6A. The western was done in three biological replicates.



FIGS. 19A-19C show the modulators' effects on HT-1080 viability. FIG. 19A-19C respectively corresponds to: FIG. 3C; FIG. 5E; and FIG. 5F. Error bars are s.e.m. of technical triplicates.



FIGS. 20A-20J show the full gel images respectively corresponding to: FIG. 12C (left); FIG. 12C (right); FIG. 12D; FIG. 12G; FIG. 12H; FIG. 12K; FIG. 11C; FIG. 15; FIG. 16D; FIG. 18.



FIG. 21 shows the workflow of Target Enrichment Analysis (TEA).



FIG. 22 shows the dual mechanism of CIL56 and ferroptosis induced by FIN56.



FIGS. 23A-23B show that FIN56-induced ferroptosis involves Ca(II) signaling.



FIG. 23A shows the flow cytometry to monitor intracellular Ca(II) levels in Fluo-4 stained HT-1080 cells.



FIG. 23B shows the effects of Ca(II) inducers and depleters on FIN56 lethality in HT-1080 cells.





DETAILED DESCRIPTION OF THE INVENTION

One embodiment of the present invention is a compound according to formula (I):




embedded image



wherein:

    • R1 is selected from the group consisting of H, OH, and —(OCH2CH2)xOH;
    • X is an integer from 1 to 6; and
    • R2, R2′, R3, and R3′ independently are selected from the group consisting of H, C3-8cycloalkyl, and combinations thereof, or R2 and R2′ may be joined together to form a pyridinyl or pyranyl and R3 and R3′ may be joined together to form a pyridinyl or pyranyl;


      or an N-oxide, crystalline form, hydrate, or pharmaceutically acceptable salt thereof; with the proviso that the compound is not




embedded image


In one aspect of this embodiment, the compound has the structure of formula (II):




embedded image



wherein:

    • R1 is selected from the group consisting of OH and —(OCH2CH2)xOH;
    • X is an integer from 1 to 6; and
    • R4 and R5 are independently selected from the group consisting of CH2 and O;


      or an N-oxide, crystalline form, hydrate, or pharmaceutically acceptable salt thereof.


Preferably, R1 is OH. Also preferably, R4 and R5 are each CH2.


In another aspect of this embodiment, the compound is selected from the group consisting of:




embedded image



and combinations thereof, or an N-oxide, crystalline form, hydrate, or pharmaceutically acceptable salt thereof.


Preferably, the compound is FIN56:




embedded image



or an N-oxide, crystalline form, hydrate, or pharmaceutically acceptable salt thereof.


Another embodiment of the present invention is a composition. This composition comprises a pharmaceutically acceptable carrier, adjuvant, or vehicle, and one or more compounds according to the present invention.


The term “alkyl” refers to the radical of saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl-substituted cycloalkyl groups, and cycloalkyl-substituted alkyl groups. In certain embodiments, a straight chain or branched chain alkyl has 10 or fewer carbon atoms in its backbone (e.g., C1-C10 for straight chains, C3-C10 for branched chains). Likewise, certain cycloalkyls have from 3-8 carbon atoms in their ring structure, including 5, 6 or 7 carbons in the ring structure.


Moreover, unless otherwise indicated, the term “alkyl” (or “cycloalkyl”) as used throughout the specification, examples, and claims is intended to include both “unsubstituted alkyls” and “substituted alkyls”, the latter of which refers to alkyl moieties having substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone. Such substituents can include, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, an aromatic, or heteroaromatic moiety. It will be understood by those skilled in the art that the moieties substituted on the hydrocarbon chain can themselves be substituted, if appropriate. For instance, the substituents of a substituted alkyl may include substituted and unsubstituted forms of amino, azido, imino, amido, phosphoryl (including phosphonate and phosphinate), sulfonyl (including sulfate, sulfonamido, sulfamoyl and sulfonate), and silyl groups, as well as ethers, alkylthios, carbonyls (including ketones, aldehydes, carboxylates, and esters), —CF3, —CN and the like. Exemplary substituted alkyls are described below. Cycloalkyls can be further substituted with alkyls, alkenyls, alkoxys, alkylthios, aminoalkyls, carbonyl-substituted alkyls, —CF3, —CN, and the like.


The term “cycloalkyl” means a univalent group derived from cycloalkanes by removal of a hydrogen atom from a ring carbon atom.


The term “Cx-y” when used in conjunction with a chemical moiety, such as, alkyl and cycloalkyl, is meant to include groups that contain from x to y carbons in the chain. For example, the term “Cx-yalkyl” refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups that contain from x to y carbons in the chain, including haloalkyl groups such as trifluoromethyl and 2,2,2-tirfluoroethyl, etc.


The term “substituted” refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with the permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this invention, the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. Substituents can include any substituents described herein, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinatean amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic moiety. It will be understood by those skilled in the art that the moieties substituted on the hydrocarbon chain can themselves be substituted, if appropriate.


As set forth previously, unless specifically stated as “unsubstituted,” references to chemical moieties herein are understood to include substituted variants. For example, reference to an “aryl” group or moiety implicitly includes both substituted and unsubstituted variants.


As used herein, an “N-oxide” means a compound containing an N—O bond with three additional hydrogen and/or side chains attached to N, so that there is a positive charge on the nitrogen. The N-oxides of compounds of the present invention may be synthesized by simple oxidation procedures well known to those skilled in the art. For example, the oxidation procedure described by P. Brougham et al. (Synthesis, 1015-1017, 1987), allows the two nitrogen of a piperazine ring to be differentiated, enabling both the N-oxides and N,N′-dioxide to be obtained. Other oxidation procedures are disclosed in, e.g., U.S. Patent Publication No. 20070275977; S. L. Jain, J. K. Joseph, B. Sain, Synlett, 2006, 2661-2663; A. McKillop, D. Kemp, Tetrahedron, 1989, 45, 3299-3306; R. S. Varma, K. P. Naicker, Org. Lett., 1999, 1, 189-191; and N. K. Jana, J. G. Verkade, Org. Left., 2003, 5, 3787-3790. Thus, the present invention includes these and other well-known procedures for making N-oxides, so long as the end product is sufficiently effective as set forth in more detail below.


The term “crystalline form”, as used herein, refers to the crystal structure of a compound. A compound may exist in one or more crystalline forms, which may have different structural, physical, pharmacological, or chemical characteristics. Different crystalline forms may be obtained using variations in nucleation, growth kinetics, agglomeration, and breakage. Nucleation results when the phase-transition energy barrier is overcome, thereby allowing a particle to form from a supersaturated solution. Crystal growth is the enlargement of crystal particles caused by deposition of the chemical compound on an existing surface of the crystal. The relative rate of nucleation and growth determine the size distribution of the crystals that are formed. The thermodynamic driving force for both nucleation and growth is supersaturation, which is defined as the deviation from thermodynamic equilibrium. Agglomeration is the formation of larger particles through two or more particles (e.g., crystals) sticking together and forming a larger crystalline structure.


As used herein, a “hydrate” means a compound that contains water molecules in a definite ratio and in which water forms an integral part of the crystalline structure of the compound. Methods of making hydrates are known in the art. For example, some substances spontaneously absorb water from the air to form hydrates. Others may form hydrates upon contact with water. In most cases, however, hydrates are made by changes in temperature or pressure. Additionally, the compounds of the present invention as well as their salts may contain, e.g., when isolated in crystalline form, varying amounts of solvents, such as water. Included within the scope of the invention are, therefore, all hydrates of the compounds and all hydrates of salts of the compounds of the present invention, so long as such hydrates are sufficiently effective as set forth in more detail below.


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


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


The compositions of the invention are pharmaceutically acceptable and may comprise one or more active ingredients in admixture with one or more pharmaceutically-acceptable carriers and, optionally, one or more other compounds, drugs, ingredients and/or materials. Regardless of the route of administration selected, the agents/compounds of the present invention are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art. See, e.g., Remington, The Science and Practice of Pharmacy (21 st Edition, Lippincott Williams and Wilkins, Philadelphia, Pa.). More generally, “pharmaceutically acceptable” means that which is useful in preparing a composition that is generally safe, non-toxic, and neither biologically nor otherwise undesirable and includes that which is acceptable for veterinary use as well as human pharmaceutical use.


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


Another embodiment of the present invention is a method of inducing ferroptosis in a cell. This method comprises contacting the cell with an effective amount of a compound having the structure of formula (I):




embedded image



wherein:

    • R1 is selected from the group consisting of H, OH, and —(OCH2CH2)xOH;
    • X is an integer from 1 to 6; and
    • R2, R2′, R3, and R3′ independently are selected from the group consisting of H, C3-8cycloalkyl, and combinations thereof, or R2 and R2′ may be joined together to form a pyridinyl or pyranyl and R3 and R3′ may be joined together to form a pyridinyl or pyranyl;


      or an N-oxide, crystalline form, hydrate, or pharmaceutically acceptable salt thereof.


As used herein, “ferroptosis” means regulated cell death that is iron-dependent. Ferroptosis is characterized by the overwhelming, iron-dependent accumulation of lethal lipid reactive oxygen species. Ferroptosis is distinct from apoptosis, necrosis, and autophagy. Assays for ferroptosis are as disclosed, for instance, in Dixon et al., 2012.


Preferred compounds that fall within the structure of formula I are as set forth above.


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


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


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


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


As used herein, ex vivo refers to a process performed in an artificial environment outside the organism on living cells or tissue which are removed from an organism and subsequently returned to an organism.


Another embodiment of the present invention is a method for decreasing GPX4 in a cell. This method comprises contacting the cell with an effective amount of a compound having the structure of formula (I):




embedded image



wherein:

    • R1 is selected from the group consisting of H, OH, and —(OCH2CH2)xOH;
    • X is an integer from 1 to 6; and
    • R2, R2′, R3, and R3′ independently are selected from the group consisting of H, C3-8cycloalkyl, and combinations thereof, or R2 and R2′ may be joined together to form a pyridinyl or pyranyl and R3 and R3′ may be joined together to form a pyridinyl or pyranyl;


      or an N-oxide, crystalline form, hydrate, or pharmaceutically acceptable salt thereof.


Preferred compounds that fall within the structure of formula I are as set forth above.


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


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


As used herein, “GPX4” refers to glutathione peroxidase 4, a glutathione metabolism enzyme.


The compositions of the invention may, optionally, contain additional ingredients and/or materials commonly used in such compositions. These ingredients and materials are well known in the art and include (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and silicic acid; (2) binders, such as carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, hydroxypropylmethyl cellulose, sucrose and acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, sodium starch glycolate, cross-linked sodium carboxymethyl cellulose and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as cetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, and sodium lauryl sulfate; (10) suspending agents, such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth; (11) buffering agents; (12) excipients, such as lactose, milk sugars, polyethylene glycols, animal and vegetable fats, oils, waxes, paraffins, cocoa butter, starches, tragacanth, cellulose derivatives, polyethylene glycol, silicones, bentonites, silicic acid, talc, salicylate, zinc oxide, aluminum hydroxide, calcium silicates, and polyamide powder; (13) inert diluents, such as water or other solvents; (14) preservatives; (15) surface-active agents; (16) dispersing agents; (17) control-release or absorption-delaying agents, such as hydroxypropyl methyl cellulose, other polymer matrices, biodegradable polymers, liposomes, microspheres, aluminum monosterate, gelatin, and waxes; (18) opacifying agents; (19) adjuvants; (20) wetting agents; (21) emulsifying and suspending agents; (22), solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan; (23) propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane; (24) antioxidants; (25) agents which render the formulation isotonic with the blood of the intended recipient, such as sugars and sodium chloride; (26) thickening agents; (27) coating materials, such as lecithin; and (28) sweetening, flavoring, coloring, perfuming and preservative agents. Each such ingredient or material must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the subject. Ingredients and materials suitable for a selected dosage form and intended route of administration are well known in the art, and acceptable ingredients and materials for a chosen dosage form and method of administration may be determined using ordinary skill in the art.


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


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


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


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


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


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


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


The rate of absorption of the active agent/drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally-administered agent/drug may be accomplished by dissolving or suspending the active agent/drug in an oil vehicle. Injectable depot forms may be made by forming microencapsule matrices of the active ingredient in biodegradable polymers. Depending on the ratio of the active ingredient to polymer, and the nature of the particular polymer employed, the rate of active ingredient release can be controlled. Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue. The injectable materials can be sterilized for example, by filtration through a bacterial-retaining filter.


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


It is understood that the disclosure of a compound herein encompasses all stereoisomers of that compound. As used herein, the term “stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures which are not interchangeable. The three-dimensional structures are called configurations. Stereoisomers include enantiomers and diastereomers.


The terms “racemate” or “racemic mixture” refer to a mixture of equal parts of enantiomers. The term “chiral center” refers to a carbon atom to which four different groups are attached. The term “enantiomeric enrichment” as used herein refers to the increase in the amount of one enantiomer as compared to the other.


It is appreciated that to the extent compounds of the present invention have a chiral center, they may exist in and be isolated in optically active and racemic forms. Some compounds may exhibit polymorphism. It is to be understood that the present invention encompasses any racemic, optically-active, diastereomeric, polymorphic, or stereoisomeric form, or mixtures thereof, of a compound of the invention, which possess the useful properties described herein, it being well known in the art how to prepare optically active forms (for example, by resolution of the racemic form by recrystallization techniques, by synthesis from optically-active starting materials, by chiral synthesis, or by chromatographic separation using a chiral stationary phase).


Examples of methods to obtain optically active materials are known in the art, and include at least the following:

    • i) physical separation of crystals—a technique whereby macroscopic crystals of the individual enantiomers are manually separated. This technique can be used if crystals of the separate enantiomers exist, i.e., the material is a conglomerate, and the crystals are visually distinct;
    • ii) simultaneous crystallization—a technique whereby the individual enantiomers are separately crystallized from a solution of the racemate, possible only if the latter is a conglomerate in the solid state;
    • iii) enzymatic resolutions—a technique whereby partial or complete separation of a racemate by virtue of differing rates of reaction for the enantiomers with an enzyme;
    • iv) enzymatic asymmetric synthesis—a synthetic technique whereby at least one step of the synthesis uses an enzymatic reaction to obtain an enantiomerically pure or enriched synthetic precursor of the desired enantiomer;
    • v) chemical asymmetric synthesis—a synthetic technique whereby the desired enantiomer is synthesized from an achiral precursor under conditions that produce asymmetry (i.e., chirality) in the product, which may be achieved using chiral catalysts as disclosed in more detail herein or chiral auxiliaries;
    • vi) diastereomer separations—a technique whereby a racemic compound is reacted with an enantiomerically pure reagent (the chiral auxiliary) that converts the individual enantiomers to diastereomers. The resulting diastereomers are then separated by chromatography or crystallization by virtue of their now more distinct structural differences and the chiral auxiliary later removed to obtain the desired enantiomer;
    • vii) first- and second-order asymmetric transformations—a technique whereby diastereomers from the racemate equilibrate to yield a preponderance in solution of the diastereomer from the desired enantiomer or where preferential crystallization of the diastereomer from the desired enantiomer perturbs the equilibrium such that eventually in principle all the material is converted to the crystalline diastereomer from the desired enantiomer. The desired enantiomer is then released from the diastereomer;
    • viii) kinetic resolutions—this technique refers to the achievement of partial or complete resolution of a racemate (or of a further resolution of a partially resolved compound) by virtue of unequal reaction rates of the enantiomers with a chiral, non-racemic reagent or catalyst under kinetic conditions;
    • ix) enantiospecific synthesis from non-racemic precursors—a synthetic technique whereby the desired enantiomer is obtained from non-chiral starting materials and where the stereochemical integrity is not or is only minimally compromised over the course of the synthesis;
    • x) chiral liquid chromatography—a technique whereby the enantiomers of a racemate are separated in a liquid mobile phase by virtue of their differing interactions with a stationary phase. The stationary phase can be made of chiral material or the mobile phase can contain an additional chiral material to provoke the differing interactions;
    • xi) chiral gas chromatography—a technique whereby the racemate is volatilized and enantiomers are separated by virtue of their differing interactions in the gaseous mobile phase with a column containing a fixed non-racemic chiral adsorbent phase;
    • xii) extraction with chiral solvents—a technique whereby the enantiomers are separated by virtue of preferential dissolution of one enantiomer into a particular chiral solvent;
    • xiii) transport across chiral membranes—a technique whereby a racemate is placed in contact with a thin membrane barrier. The barrier typically separates two miscible fluids, one containing the racemate, and a driving force such as concentration or pressure differential causes preferential transport across the membrane barrier. Separation occurs as a result of the non-racemic chiral nature of the membrane which allows only one enantiomer of the racemate to pass through.


The stereoisomers may also be separated by usual techniques known to those skilled in the art including fractional crystallization of the bases or their salts or chromatographic techniques such as LC or flash chromatography. The (+) enantiomer can be separated from the (−) enantiomer using techniques and procedures well known in the art, such as that described by J. Jacques, et al., Enantiomers, Racemates, and Resolutions”, John Wiley and Sons, Inc., 1981. For example, chiral chromatography with a suitable organic solvent, such as ethanol/acetonitrile and Chiralpak AD packing, 20 micron can also be utilized to effect separation of the enantiomers.


The following examples are provided to further illustrate the methods of the present invention. These examples are illustrative only and are not intended to limit the scope of the invention in any way.


EXAMPLES

The invention is further illustrated by the following examples, which are offered for illustrative purposes, and are not intended to limit the invention in any manner. Those of skill in the art will readily recognize a variety of noncritical parameters, which can be changed or modified to yield essentially the same results.


Example 1
Materials and Methods

Chemicals.


3,169 uncharacterized lethal compounds, as well as structural analogs of CIL41/70, were purchased from Asinex, Chem Bridge, Chem Div, Enamine, InterBioScreen, MayBridge, TimTec, Vitas M Labs, and Zelinsky. Chemical modulators (see Table 2) used in modulatory profiling were obtained as previously described (Wolpaw et al. 2011). U0126 was obtained from LC Laboratories. Deferoxamine mesylate, α-tocopherol, sodium selenite, zaragozic acid A, mevalonolactone and simvastatin were purchased from Sigma-Aldrich. Cerivastatin was obtained from Waterstone Technology. Lovastatin was obtained from Santa Cruz Biotechnology. YM-53601 and TOFA were purchased from Cayman Chemical. Isoprenoid pyrophosphate (isopentenyl-PP, dimethylallyl-PP, geranyl-PP, farnesyl-PP and geranylgeranyl-PP) ammonium salts were purchased from Isoprenoids. Idebenone was purchased from Tocris Bioscience. Pim kinase inhibitors (CX-6258 and AZD-1208) were purchased from Selleck Chemicals. Erastin and (1S,3R)-RSL3 were synthesized as described previously (Yang et al. 2014; Yagoda et al. 2007). NB-598 maleate was purchased from ChemScene. CIL56, FIN56, and their structural analogs were synthesized as described by Cholody et al. in the international patent application PCT/US2008/006015 (published as WO2008140792A1). Building blocks for these compounds were purchased from Matrix Scientific and Sigma-Aldrich. The purity of the purchased compounds was not assessed. Structures of 56 CILs and CIL41/70 analogs are presented in Tables 4 and 5. The total synthesis of CIL56 analogs and their 1H NMR data are described in Example 8.


Cell Lines and Media.


Four engineered BJ cell lines (BJeLR, DRD, BJeHLT, and BJeH) were obtained from Robert Weinberg (Whitehead Institute). 143B cells (osteosarcoma) were from Eric Schon (Columbia University). Calu-1 (lung adenocarcinoma) and HT-1080 (fibrosarcoma) cells were from American Type Culture Collection. The four BJ cell lines were grown in DMEM high-glucose media (Life Technologies), 20% Medium 199 (Sigma), and 15% heat-inactivated FBS (FBS). HT-1080 cells were grown in DMEM high-glucose media with 1% non-essential amino acids (Life Technologies) and 10% FBS. 143B cells were grown in DMEM high-glucose media with 1% glutamine and 10% FBS. Calu-1 cells were grown in McCoy's 5A media (Life Technologies) supplemented with 10% FBS. All the cell lines were grown at 37° C. under 5% CO2. Cell lines were not tested for mycoplasma.


Cell Viability Assay.


1,000 cells per 36 μl were seeded in each well in 384-well plates. Lethal compounds were dissolved, and a twofold, 12-point dilution series was prepared in DMSO. Compound solutions were further diluted with media at 1:25, and 4 μl per well of the diluted solutions was added to cell cultures immediately after cells were seeded. When ferroptosis inhibitors (100 μM α-tocopherol, 152 μM deferoxamine, or 10 μM U0126) were used in cotreatments with lethal inducers, they were supplemented to cell culture when the lethal compounds were added, and the cells were then incubated for 24 h. When other cell-death-modulating compounds (100 nM sodium selenite, 1 μM cerivastatin, 100 μg/mL mevalonic acid) were used in cotreatments, they were supplemented to cell culture for 24 h before lethal compounds were added to cell culture, after which cells were further incubated for 24 h at 37° C. under 5% CO2. On the day of the viability measurement, we added 10 μl per well of 50% Alamar Blue diluted in media (Life Technologies) and further incubated the cells at 37° C. for 6 h. Fluorescence intensity (excitation/emission: 530/590) was measured with a Victor 3 plate reader (PerkinElmer), and the normalized viability was calculated as VL=(IL−I0)/(IV−I0), where VL, I0, IV, and IL are the normalized viability and the raw fluorescence intensities from the wells containing media, cells treated with a vehicle (negative control), and cells with the lethal compound (L), respectively. When the effect of a chemical modulator (M) on L was calculated, we instead used the equation VL|M=(IM,L−I0)/(IM,V−I0), where VL|M, IM,L and IM,V are the normalized viability, and fluorescence intensity from cells treated with M and V, and from cells with M and L, respectively. The viability was typically measured in biological triplicate unless otherwise specified. A representative dose-response curve, the mean and standard error of normalized viability from one replicate were plotted. HT-1080 viability after modulator treatments corresponding to FIG. 3C and FIGS. 5E-5F is available in FIGS. 19A-19C.


Caspase-3/7 Activation Assay.


The Apo-ONE Homogeneous Caspase-3/7 Assay (Promega) was used according to the manufacturer's protocol, with a minor modification. First, we optimized the assay (FIG. 7A-7B). HT-1080 cells were seeded at 1,000 cells per 40 μl in each well of a 384-well plate, incubated for 1 h, and treated with test compounds for different durations (from 3.5 to 48 h). 15 μl of culture media was aspirated from each well, and 5 μl of a mixture of lysis buffer and caspase-3/7 fluorogenic substrate from the kit was added. Plates were kept in the dark at room temperature for 16 h, and the fluorescence (excitation/emission: 490/535) of each well was measured using a Victor 3 plate reader. In the optimization, the fluorescence of apoptosis-inducer-treated cells started increasing after 6-12 h, and lethal compounds that induced strong positive signals in the end (i.e., apoptosis inducers) were distinguished from non-apoptotic inducers as early as after 18 h. We therefore treated cells with lethal compounds for 18 h in further analyses. In the screening mode, we incubated HT-1080 cells with screening molecules for 18 h, processed cells, and measured the fluorescence. Compounds inducing fluorescence with similar intensity as vehicle (DMSO) treatments were defined as caspase-independent lethals.


Discovery of Novel Ferroptosis Inducers Using Modulatory Profiling.


Collection of CILs. First, collected uncharacterized synthetic compounds from various vendors for different screening purposes. Of those, we found 3,169 compounds to be lethal in BJeLR cells. We first sought compounds that induced non-apoptotic cell death in two ferroptosis-susceptible cell lines, HT-1080 and BJeLR. We tested the 3,169 compounds at 5.3 μg/mL in those two cell lines, and 451 compounds showed (i) an EC80<2.8 μg/mL in both cell lines and (ii) no activation of caspase-3/7 at 5.3 μg/mL. Cell viability and caspase-3/7 activity assays were performed as described above. The 95th percentile of Apo-ONE fluorescence from 0.13% DMSO-treated cells was set as the threshold of caspase-independent lethality, and the raw fluorescent value of each well was divided by the threshold for normalization; lethal compounds were considered caspase-3/7-independent when the normalized fluorescent values were less than one. These compounds were defined as CILs. Next, we computed the structural similarity among the 451 compounds based on Pubchem's fingerprint (Backman et al. 2011) and removed structurally similar compounds (cutoff: Tanimoto coefficient of 0.9). We also removed compounds that did not satisfy Lipinski's rule of five with a minor modification (no more than five hydrogen bond donors, no more than ten hydrogen bond acceptors, molecular weight ranging from 250 to 500, a partition coefficient log P not greater than five) and compounds whose biological activities were known. Finally, we retested the selected compounds in twofold dilution series in the two cell lines and closely examined 56 compounds with low EC50 in modulatory profiling.


Modulatory profiling of CILs. Examined 56 CILs using modulatory profiling (Wolpaw et al. 2011) (FIG. 8). HT-1080 and BJeLR cells were seeded at 1,000 cells per 40 μl in each well of a 384-well plate. They were cotreated with a lethal compound (L) and a death modulator (M) in technical triplicates. We added 56 CILs to cells in a twofold, 14-point dilution series. Death modulators were reagents known to perturb cell death signaling pathways and were treated at a single concentration, as described previously (Wolpaw et al. 2011). Cells were incubated with lethal compounds and modulators for 48 h, and the normalized viability was measured as described above. For each combination of L and M, the area under the dose-response curve (AUC) was computed. An effect of M on L (EM|L) was represented by the difference in AUCs between the modulator (AUCM,L) and the vehicle (AUCV,L), or EM|L=AUCM,L−AUCV,L (see FIG. 8B). When M suppresses or enhances the effect of L, EM|L>0 or EM|L<0, respectively.


The modulatability of each L (mL) was an average of the absolute value of the effect of all M's on L, or mLM|EM|L|/nM, where nM is the number of modulators, 46. Large mL indicates that L induces a selective cell death phenotype (Wolpaw et al. 2011). Computed mL for each CIL and each known lethal compound from different classes of mechanism of action (i.e., HDAC inhibitors, proteosomal inhibitors, mitochondrial uncouplers, topoisomerase I and II inhibitors, microtubule destabilizers, and ferroptosis inducers). mL of CILs larger than the median value of known lethal compounds' mL were considered ‘high’; we identified high-modulatability CILs. For generating hierarchical clustering of the modulatory profiles, the distance between modulatory profiles (P) of two compounds Pi and Pj was defined by dij=1−CorrPearson(Pi,Pj), and hierarchical clustering with an average-linkage method was used to generate dendrograms.


Cobalt (II) Binding Assay In Vitro.


For each cluster I compound (CIL13, −52, and −64), prepared two-fold dilution series with DMSO. We added 30 μL per well of either water or 10 μM cobalt (II) chloride solution to each well of a 384-well plate. Dilution series of each compound in DMSO were added by 10 μL per well. After solutions had been mixed by shaking of the plate using a Victor 3 plate reader (PerkinElmer), absorbance was scanned between 300 and 700 nm for each compound. Finally the following Absorbance value for each compound was plotted (see FIG. 9B):









ΔΔΔAbsorbance
=




Δ






Abs

CIL
,


Co





2

+




-


Δ

Abs


CIL
,
water









=




(



Ab

s


CIL
,


Co





2

+



-

Abs

DMSO
,


Co





2

+




)

-










(



Ab

s


CIL
,
water


-


Ab

s


DMSO
,
water



)









where Abs is the absorbance at each wavelength.


Search of Commercially Available Structural Analogs of CIL41/70.


Structural analogs possessing the core scaffold of CIL41/70 (SMILES: C(═O)([*])O/N═C([*])/N) were searched for in eMolecules (see, e.g., http://www.emolecules.com) and purchased from the vendors listed above. Their lethality was tested once in HT-1080 cells in technical triplicates.


Analysis of ROS Generation.


Grew 200,000 HT-1080 or BJeLR cells in six-well plates at 37° C. for 16 h. Cells were treated with test compounds, trypsinized, pelleted, and washed once with PBS. For lipophilic or aqueous ROS detection, cells were re-suspended in Hanks' Balanced Salt Solution (HBSS; Life Technologies) containing test compounds as well as C11-BODIPY(581/591) (2 μM) or H2-DCFDA (25 μM), respectively (Life Technologies), and incubated for 10 min at 37° C. Cells were then pelleted, re-suspended in 500 μL HBSS, strained through a 40-μm cell strainer (BD Falcon), and analyzed using a BD Accuri C6 flow cytometer (BD Biosciences). Both dyes were measured in the FL1 channel. Experiments were done in biological triplicates.


Glutathione-Quantification Assay.


Seeded 500,000 HT-1080 cells in a 10-cm dish. Cells were grown at 37° C. for 16 h. On the day of the analysis, cells were cotreated with 100 μM α-tocopherol and either vehicle (DMSO) or a ferroptosis inducer (10 μM erastin, 0.5 μM (1S,3R)-RSL3, or 5 μM FIN56) and incubated for 10 h. Cells were then trypsinized, pelleted, washed once with 400 μL of ice-cold PBS containing 1 mM EDTA, and sonicated. After the cell debris had been pelleted and removed, both oxidized and reduced glutathione in 120 μL of sample was quantified in technical triplicates using the QuantiChrome glutathione assay kit (BioAssay Systems). The glutathione quantity was normalized to the protein concentration measured via Bradford assay (Bio-Rad).


GPX4 Enzymatic Activity Assay.


Seeded 17 million BJeLR cells in 225-cm2 tissue culture flasks (Corning). We then added vehicle (0.1% DMSO for 11 h) or test compound (10 μM erastin for 11 h, 0.5 μM (1S,3R)-RSL3 for 2 h, or 5 μM FIN56 for 10 h). The GPX4 enzymatic activity assay was performed as described previously (Yang et al. 2014). Briefly, 10 million cells were resuspended in cell lysis buffer. Cells were sonicated to make cell lysates that were then cleared by centrifugation at 14,000 r.p.m. for 10 min. The protein concentration of the cleared cell lysates was determined via Bradford protein assay. Mixed 200 μg of cellular protein with phosphatidyl choline hydroperoxide, a GPX4-specific substrate, and reduced glutathione, a GPX4 cofactor. The mixture was incubated at 37° C. for 30 min and then subjected to lipid extraction using chloroform:methanol (2:1) solution. The lipid extract was evaporated using a Rotavap and re-dissolved in 100% ethanol before being injected into the LC-MS instrument for 2-linoleoyl1-palmitoyl-sn-glycero-3-phosphocholine (PLPC) quantitation.


Reverse-Transcription Quantitative PCR.


Trypsinized 0.2 to 1 million cells grown in six-well dishes, pelleted them, resuspended them in Buffer RLT (Qiagen), and homogenized them with QlAshredder (Qiagen). RNA was further extracted using the RNeasy Mini Kit (Qiagen). We converted 2 μg of extracted RNA from each sample into cDNA using TaqMan reverse-transcription reagents (Life Technologies). qPCR primers were designed to detect all splicing variants using Primer Express 2.0 (Applied Biosystems). The designed primers were confirmed to amplify only the designated gene transcripts using in silico PCR (see, e.g., http://genome.ucsc.edu/cgi-bin/hgPcr). For qPCR reactions, primers, cDNA, and Power SYBR Green PCR Master Mix (Applied Biosystems) were mixed, and quantitation was performed using a StepOnePlus real-time PCR system (Applied Biosystems). Experiments were done in biological triplicates.


Western Blotting.


Seeded 300,000 cells (HT-1080 or BJeLR) per well in six-well plates. For cotreatment, ferroptosis inducers (10 μM erastin for 11 h, 0.5 μM (1S, 3R)-RSL3 for 2 h, or 5 μM FIN56) and 100 μM α-tocopherol were added to cell culture at the same time, and cells were then incubated for 10 h (FIG. 2E). When other death-modulating compounds (100 nM sodium selenite, 1 μM cerivastatin, 100 μg/mL mevalonic acid, 30 μM C75, or 2 μM cerulenin) were used in cotreatments, cells were pre-incubated with the modulators for 24 h before being treated with ferroptosis inducers for 6 h. Cell lysis, SDS-PAGE and protein transfer to PVDF membrane were performed as previously described (Dixon et al. 2012). We used the following antibodies: anti-human α-tubulin (Santa Cruz Biotechnology, sc-32293, 1:10,000 dilution), anti-human GPX4 (Abcam, ab41787, 1:2,000 dilution), anti-human GPX1 (R&D Systems, AF3798, 1:1,000 dilution), and anti-human SQS (Abcam, ab109723 for full-length, ab195046 for truncated, both at 1:1,000 dilution). Secondary antibodies were from LI-COR (1:3,000 dilution). The PVDF membranes labeled with primary and secondary antibodies were scanned using an Odyssey Imaging System (LI-COR). Experiments were performed in biological triplicates, and the mean±s.e. of intensity was plotted. Full gel images are shown in FIGS. 20A-20J.


Gene-Knockdown Experiment.


shRNAs designed by the RNAi Consortium were used, and gene knockdown was performed as described previously (9). For siRNA-mediated gene-knockdown experiments, HT-1080 cells were reverse-transfected with 5 nM siRNA. siRNA targeting GPX4 (Dharmacon, #L-011676-00) or TRIT1 (Dharmacon, #L-018831-02) or nontargeting siRNA (Qiagen) was mixed with 2 μL Lipofectamine RNAiMAX (Invitrogen) in a well of a 12-well plate. After incubation for 30 min at room temperature, 30,000 cells were added to each well, and knockdown was allowed to proceed for 48 h. Cells were then harvested and re-seeded for RT-qPCR, viability assay, and western blotting. For RT-qPCR, cells were re-seeded into 12 well plates and harvested the following day as described above. For viability assay, 1,000 cells per well were reseeded into 384-well plates for 24 h and were then incubated with compounds for another 24 h before the addition of Alamar Blue. Compounds were added in a 12-point twofold dilution series, with the highest concentrations of compound being 36.5 μM (erastin), 10 μM ((1S,3R)-RSL3), and 38.7 μM (FIN56). For western blotting, cells were harvested 48 h after knockdown (no re-seeding).


FIN56 Target Identification.


Conjugation of active and inactive probes with Profinity epoxide resin. Incubated 6 μmol of active (SRS11-31) or inactive (SRS11-66) probes dissolved in 500 μL DMSO and 300 mg of Profinity epoxide resin (Bio-Rad) in saturated sodium bicarbonate at 45° C. for 3 d. The conjugation reaction was ended by adding 120 μL of 1 M ethanolamine to the reaction mixture. The conjugated probe beads were used for further protein pulldown assay.


Affinity chromatography with active and inactive probe beads. Seeded 8 million HT-1080 cells in two 15-cm polystyrene tissue culture dishes and allowed the cells to grow overnight. Culture media was removed from dishes, and cells were washed five times with cold PBS. After the PBS had been completely removed, the cells were treated with 2 mL of lysis buffer (25 mM MOPS (pH 7.2), 15 mM EGTA, 15 mM MgCl2, 2 mM DTT, 1 mM sodium orthovanadate, 1 mM sodium fluoride, 0.5% NP-40, 60 mM β-glycerophosphate, protease inhibitor cocktail (Sigma-Aldrich P8340)) per plate, scraped, and collected. Cells were then agitated at 4° C. for 15 min, after which insoluble components were precipitated at 14,000 g at 4° C. for 10 min, the supernatant was removed, and the protein concentration was measured via Bradford assay.


Incubated 250 μg of protein (up to 400 μL of the whole-cell lysate) with active and inactive probe beads, added 20 mL of bead buffer (50 mM Tris-HCl (pH 7.4), 250 mM NaCl, 5 mM EDTA, 5 mM EGTA, 5 mM NaF, 0.1% NP-40), and incubated the mixture at 4° C. for 12 h. Beads were then collected using Poly-prep chromatography columns (Bio-Rad). Beads were then washed with 8 mL of bead buffer three times and transferred to Eppendorf tubes.


Bacterial expression and purification of truncated human SQS for competition assay. The plasmid encoding the truncated human squalene synthase (SQS 31-370) in pET28a expression vector, kindly provided by Chia-I Liu and Andrew H. J. Wang (Academia Sinica, Taiwan), was confirmed by DNA sequencing (Gene Wiz, Inc.) and then used to transform Escherichia coli BL21-Gold (DE3) competent cells (Agilent Technologies). The cells with the construct were grown in LB media supplemented with 100 μg/mL ampicillin at 37° C. until the OD600 reached 1. Protein expression was induced with 0.5 mM isopropyl β-D-thiogalactoside at 17° C. overnight (12-13 h). Cells were pelleted (4,000 g, 20 min, 4° C.) and lysed by sonication in SQS buffer (50 mM Tris, pH 7.4, 250 mM NaCl, 5 mM imidazole, 5 mM MgCl2, 1 mM TCEP). Cell lysate was then centrifuged at 15,000 g for 45 min at 4° C. The supernatant was loaded onto a chromatography column containing Ni Sepharose 6 Fast Flow beads (GE Life Sciences) equilibrated with SQS buffer. After two washes and one nonspecific wash of the beads, the bound SQS was eluted with 250 mM imidazole in the same buffer. The purity of eluted fractions was verified by SDS-PAGE as more than 90% pure. The fractions containing SQS were concentrated, flash-frozen, and stored at −80° C. Protein concentration was determined using absorbance at 280 nm with a molar extinction coefficient (ε) of 42,860 M−1 cm−1 (for reduced SQS with an N-terminal His12 tag as calculated from the amino acid sequence by ExPASy ProtParam).


SQS competition assay with purified truncated SQS, FIN56, and active probe. Purified truncated SQS was prepared as described above. The protein solution was diluted with bead buffer (50 mM Tris-HCl, pH 7.4, 250 mM NaCl, 5 mM EDTA, 5 mM EGTA, 5 mM NaF, 0.1% NP-40, 1 mM TCEP). We mixed 190 μL of 1 or 10 ng/mL SQS solution with 10 μL of DMSO or competitor solution (2 mM FIN56 solution in DMSO), with DMSO and FIN56 at final concentrations of 5% and 100 μM, respectively. This mixture was rotated and incubated at 4° C. for 2 h. We added 15 μL of active probe-bead solution (33% slurry) to the solution and incubated it for another 2 h. The beads were spun down (500 r.p.m., 4° C., 1 min), and supernatant was removed and washed with 500 μL of bead buffer three times. After the third wash, 20 μL of 3×SDS sample loading buffer was added, and the mixture was boiled at 95° C. for 5 min. Samples were spun for 1 min at 13,000 r.p.m. in a table-top centrifuge, and 10 μL of each sample was loaded on gels for SDS-PAGE. SQS was confirmed by immunoblotting.


Proteomic analysis for target identification. At Quantitative Proteomics Center at Columbia University, the proteins in pulldown samples with active (SRS11-31) and inactive (SRS11-66) beads were eluted at 80° C. in 50 mM ammonium bicarbonate with 0.1% Rapigest detergent with protease inhibitor cocktail P8340 (Sigma-Aldrich). Biological triplicate culture and affinity pulldowns for bound active and inactive compound were prepared. Cysteines in the protein samples were reduced with dithiothreitol and alkylated with iodoacetamide, and proteins were digested with trypsin (6 ng/μL, Promega Corp, #V511A, in 50 mM ammonium bicarbonate). A digest of yeast alcohol dehydrogenase (50 fmol) was added as an internal detection control.


Three chromatograms were recorded for each of six biological replicates (three active, three inactive), yielding 12 chromatograms. Analytical separation was done on a NanoAcquity UPLC (Waters), with a 120-min chromatogram on a 75-μm inner diameter×25-cm HSS T3 1.8-μm-particle-diameter reverse-phase C18 column at a flow rate of 300 nL/min with an acetonitrile/formic acid gradient at 45° C. Identification and quantitation of proteins bound to the beads was done by label-free proteomic profiling on a Synapt G2 HDMS (quadrupole-time-of-flight) mass spectrometer (Waters) using data-independent scanning (MSE) as described previously (Yang et al. 2014), except that spectra were recorded in positive-ion sensitivity mode without ion mobility. Spectra were searched against a human UniProt complete proteome with ProteinLynx Global Server version 2.5 RC9 (Waters) and post-processed with Elucidator software version 3.3.0.1.SP3_CRE52.21 (Ceiba Solutions, Inc.) as described previously (Alegre-Aguarón et al. 2014).


Target validation using custom shRNA library. Custom lentiviral shRNA libraries were generated using plasmids encoding the first generation of the RNAi Consortium shRNAs targeting 70 genes identified by chemoproteomics according to the RNAi Consortium's instructions. These lentiviral shRNAs were laid out in an arrayed format using 384-well plates and were infected in four cell lines (HT-1080, BJeLR, Calu-1, and 143B). Our rationale for the screening design was that if shRNAs target the bona fide FIN56 targets, those shRNAs should show ‘consistent’ FIN56-enhancing or -suppressing effects in all four cell lines, depending on how FIN56 acts on the target protein (FIG. 14A). If the shRNAs act on off-targets to change sensitivity to FIN56, their effects should be less consistent. Gene knockdown was performed as described above; 400 cells per well in 384-well plates were seeded and incubated for a day, lentivirus-containing shRNAs were infected in them on the next day, and puromycin was added 2 d after infection. Cells were treated with twofold eight-point dilution series of FIN56 after 24 h and incubated for another 48 h before Alamar Blue was added, after which the cells were incubated for 6 h.


Data analysis for discovering functionally relevant targets. Alamar Blue fluorescence intensity data were normalized as described above. For each cell line, dose-response curves for FIN56 for each shRNA treatment were plotted and overlaid on a single plot first. From among eight tested concentrations of FIN56, extreme (low or high) concentrations that did not kill or completely kill cells treated with any shRNAs were removed; eventually four or five more informative concentrations were used for further analysis, as in FIG. 4B and FIG. 14B. For each shRNA treatment, the AUC after FIN56 treatment in dilution series was computed and rank-ordered across all tested shRNAs in each cell line. Based on their ‘consistency’ across four cell lines and the magnitude of effects, shRNAs were classified into 11 categories (5 consistently suppressing groups (top 10%, top 20%, etc.), 5 consistently enhancing groups (top 10%, etc.), and nonconsistent). Note that ‘nonconsistent’ indicates that the shRNA induced FIN56-enhancing effects in some cell lines and FIN56-suppressing effects in others. A gene was ranked according to the consistent effects of shRNAs targeting it. When at least one of the shRNAs targeting a gene showed consistent FIN56-enhancing or -suppressing effects, the gene was considered a potentially functionally relevant target of FIN56, through loss-of-function or gain-of-function scenarios, respectively.


Validation of loss-of-function targets using pooled siRNAs. Candidate loss-of-function targets of FIN56 were further targeted by pooled siRNAs. Previously, pooled siRNA against GPX4, a target of RSL3, was shown to phenocopy RSL3; siGPX4 induced ferroptosis that was suppressed by α-tocopherol as well as oncogenic Ras selectivity across BJ series. Expecting that siRNAs against bona fide loss-of-function targets would induce ferroptosis, we treated siRNAs against the candidates as well as GPX4 as a positive control. siRNAs were transfected into BJeLR cells. We incubated 1 mL of Opti-MEM (Life Technologies) mixed with 20 μmol of pooled siRNA and 5 μL of RNAiMAX (Life Technologies) to form a complex for 15 min, and we then aliquoted 500 μL per well into six-well dishes. Next, 120,000 BJeLR cells per 1.5 mL in each well were seeded and grown for 2 d. Cells were then trypsinized and seeded again at 120,000 per well and grown with or without supplementation of α-tocopherol. After 2 d, cells were trypsinized and cell density was measured using an automated cell counter (Vicell, Beckman Coulter).


Statistical Analysis and Data Visualization.


Dose-response curve plotting and EC50 computation were done with Prism 5.0c. P values for the differences in EC50 values were computed on the basis of model comparison with Akaike's information criterion. The rest of the statistics and plotting were done using R language and the following R packages and functions: ChemmineR package for Pubchem's fingerprint and Tanimoto coefficient computation, heatmap.2 function in the gplots package for plotting heat maps, and flowCore and flowViz packages modified for plotting .fcs files in flow cytometry. The statistical significance of protein expression (on western blots) was calculated using paired two-tailed t-tests.









TABLE 1







Small molecule screening data.









Category
Parameter
Description





Assay
Type of assay
Cell-based assay



Target
Casepase-independent lethality in HT-1080 and




BJeLR.



Primary measurement
i) Lethality: Alamar Blue (Life Technologies)




Viability in both HT-1080 and BJeLR cells. ii)




Apo-ONE Homogeneous Caspase-3/7 Assay




(Promega)



Key reagents
Alamar Blue (Life Technologies); Apo-ONE




Homogeneous Caspase-3/7 Assay reagents




(Promega)



Assay protocol
Grow cells treated with lethal compounds. Lysed




cells and measured Apo-ONE Caspase-3/7




fluorescence after 16 hours. Separately, HT-1080




and BJeLR were incubated with compounds for 48




hours and added Alamar Blue, incubated for




another 6 hours.



Additional comments



Library
Library size
3,169 compounds



Library composition
Uncharacterized compounds pre-selected for




lethality in BJ-eLR cell line.



Source
Diversity-oriented library was purchased from the




following companies: TimTec, InterBioScreen,




ChemBridge.



Additional comments



Screen
Format
384 well plate



Concentration(s) tested
5.3 ug/mL



Plate controls
DMSO (0.4%) as non-lethal control. No lethal




control



Reagent/compound dispensing system
Beckman Coulter Biomek FX Workstation



Detection instrument and software
Perkin Elmer Victor3



Assay validation/QC




Correction factors
None



Normalization
i) Viability: median of 28 wells with DMSO treated




cells in each 384-well plate was set as one. All the




wells were scaled proportionally, ii) Caspase-3/7




activity: 95 percentile of HMSO treated cells were




set as one. All the wells were scaled proportionally.



Additional comments



Post-HTS
Hit criteria
Compounds that induces i) Viability < 0.2 in both


analysis

HT-1080 and BJeLR cell lines, and ii) Caspase-3/7




activity < 1 were considered caspase-independent




lethal,



Hit rate
451/3,169 = 14.2%



Additional assay(s)
Of 451 hits, 56 structurally diverse and potent




compounds were selected, purchased and tested for




Modulatory Profiling.



Confirmation of bit purity and structure
The selected 56 compounds were further reordered




from another vendors, tested for caspase




independent lethality in 12-point 2-fold dilution




series.



Additional comments
All 56 compounds were confirmed positive.
















TABLE 2







Death modulators used in modulatory profiling.













Chemical or Genetic




conc
Ferroptosis


Modulator
Abbreviation
Mechanism
HT1080
BJeLR
(μM)
suppressor
















α-tocophenol
Atoc
Antioxidant
x
x
100
x


Butylated
BHA
Antioxidant
x

140
x


hydroxyanisole








Butylated
BHT
Antioxidant
x

115
x


hydroxytoluene








(+)-6-Hydroxy-2,5,7,8-
Trolox
Antioxidant
x
x
150
x


tetramethylchromane-








2-carboxylic acid








Cobalt (II)
Co2
Calcium channels
x
x
1200





blocker






Gadolinium (III)
Gd3
Calcium channels
x
x
30





blocker (via








sodium leak








channel, stretch








activated channel)






Ethyleneglycol-O,O′-
EGTA
Calcium ion
x

2000



bis(2-amino ethyl)-

chelator






N,N,N′,N′-tetraacetic








acid








Digoxin
Dig
Inhibitor of Na/K
x

0.13





ATPase (increase








calcium ion influx)






Deteroxamine
dtom
iron ion chelator
x

150
x


L-mimosine
Lmim
iron ion chelator,
x
1*
175
x




inhibitor of cell








cycle at G1-S






Cycloheximide
CHX
Inhibitor of protein
x
x
1.5





synthesis






Actinomycin D
ActD
Inhibitor of RNA
x

0.0012





synthesis (RNAPII)






Calpain Inhibitor I
ALLN
Inhibitor of calpain
x
x
25





I/II and cathepsins








B/L






Na-tosyl-lys-
TLCK
Inhibitor of trypsin-
x
x
135



chloromethylketone

like serine proteasus






Pepstatin
Pepstatin
Inhibitor of
x
x
1





cathepsin D






NG-Monomethyl-D-
NMMA
Inhibitor of nitric
x
x
7



arginine

oxide synthase






NG-Nitro-L-arginine-
LNAME
Inhibitor of nitric
x
x
300



methyl ester

oxide synthase






1,4-diamino-2,3-
U0126
Inhibitor of
x

11



dicyano-1,4-bis[2-

Mitogen-activated






aminophenylthio]

protein kinase






butadiene

kinase (MEK) 1/2






Anthra(1,9-
SP600125
Inhibitor of c-Jun
x

10



cd)pyrazol-6(2H)-one

N-terminal kinase






1,9-Pyrazoloanthrone

1/2/3






Obz-val-ala-
zVAO
Inhibitor of pan-
x
x
45



asp(OMe)-

caspases (apoptosis)






fluormethylketone








t-butoxycarbonyl-asp-
BocD
Inhibitor of pan-
x
x
50



fluormethylketone

caspases (apoptosis)






3,4-dihydro-5-[4-(1-
DPQ
Inhibitor of poly-
x
x
10



pipendinyl)butoxyl]-

ADP ribose






1(2H)-isoquinolinone

polymerase








1(PARP1;








PARP1-








dependent cell








death)






3-methyladenine
3MA
Inhibitor of
x
x
5000





autophagosome








formation








(macroautophagy)






Necrostatin-1
Nec1
Inhibitor of receptor
x
x
20





interactin protein








kinase 1








(RIPK1)








(necroptosis) and








idoleamine 2,3-








dioxygenase (IDO)






Aunntricarboxylic
ATA
Inhibitor of
x

40



Acid

ribonuclease






Nicotinamide adenine
NAD
Activities sirtuins,
x
1*
2000



dinucleotide, oxidized

endogenious








electrons carrier






β-Carolene
Bcarotene
Vitamin A precursor
x

0.2



Cyclosporin A
CspA
Binds cyclophilin D
x

5





1*: used both individually and as a combination (NAD+ and Lmim)













TABLE 3





EC50s of CILs classified with other ferroptosis inducers and the effects of ferroptosis suppressors on the CILs.





















CIL41
CIL56
CIL69





HT-1080
DMSO
5.67 (4.13 to 7.78)
0.443 (0.287 to 0.683)
0.551 (0.337 to 0.902)


iron-chelators
Deferoxamine
 124 (96.3 to 159)
 1.36 (0.946 to 1.94)
 9.29 (6.23 to 13.9)



Lmim
69.8 (65.9 to 73.8)
 1.53 (1.22 to 1.94)
 4.79 (3.52 to 6.52)


antioxidants
Atoc
  50 (44 to 56.8)
 2.49 (2.22 to 2.8)
 6.13 (4.79 to 7.86)



Trolox
23.4 (20 to 27.3)
 1.31 (1.08 to 1.58)
 4.26 (3.45 to 5.26)



BHA
  26 (18.3 to 36.9)
 2.65 (2.02 to 3.47)
 4.79 (3.39 to 6.76)



BHT
12.6 (9.47 to 16.7)
 1.69 (1.09 to 2.61)
 3.28 (2.32 to 4.62)


MEK inh
U0126
20.9 (16.6 to 26.4)
 2.81 (2.2 to 3.58)
 5.75 (4.01 to 8.24)


BJeLR
DMSO
3.17 (2.58 to 3.88)
0.213 (0.164 to 0.276)
0.193 (0.126 to 0.294)


iron-chelators
Lmim
22.4 (14.7 to 34.1)
0.925 (0.646 to 1.32)
0.976 (0.652 to 1.46)



Lmim_NAD
22.6 (14.7 to 34.6)
 1.3 (0.884 to 1.91)
0.889 (0.696 to 1.13)


antioxidants
Trolox
23.2 (16 to 33.5)
 2.03 (1.5 to 2.73)
 2.05 (1.6 to 2.63)



atoc
36.6 (27.2 to 49.3)
 2.49 (1.9 to 3.26)
 1.78 (1.45 to 2.2)







CIL70
CIL75
CIL79





HT-1080
DMSO
4.44 (2.98 to 6.62)
 0.144 (0.101 to 0.205)
0.271 (0.198 to 0.371)


iron-chelators
Deferoxamine
 157 (114 to 216)
 4.82 (3.75 to 6.2)
 32.4 (23.7 to 44.2)



Lmim
47.8 (34.6 to 66)
 2.47 (1.67 to 3.64)
  12 (9.01 to 15.9)


antioxidants
Atoc
32.2 (21.8 to 47.5)
 2.94 (2.23 to 3.89)
 11.7 (9.82 to 13.9)



Trolox
11.1 (10.2 to 12.2)
 2.56 (2.22 to 2.95)
 10.2 (7.42 to 14.1)



BHA
13.3 (10 to 17.7)
 2.75 (1.97 to 3.85)
 10.1 (7.02 to 14.4)



BHT
 6.5 (4.63 to 9.12)
 1.72 (1.27 to 2.34)
 6.3 (4.62 to 8.58)


MEK inh
U0126
10.3 (8.06 to 13.2)
 2.53 (1.72 to 3.7)
 14.3 (10.7 to 19.1)


BJeLR
DMSO
1.76 (1.29 to 2.4)
0.0598 (0.0411 to 0.0868)
0.157 (0.121 to 0.204)


iron-chelators
Lmim
13.1 (9.02 to 18.9)
 0.341 (0.198 to 0.589)
0.667 (0.385 to 1.16)



Lmim_NAD
14.8 (11.7 to 18.7)
 0.314 (0.209 to 0.472)
0.566 (0.469 to 0.684)


antioxidants
Trolox
 7.1 (5.19 to 9.71)
  1.1 (0.824 to 1.48)
 8.32 (6.41 to 10.8)



atoc
18.6 (15.5 to 22.3)
 1.04 (0.857 to 1.26)
 7.93 (6.09 to 10.3)





* unit is μM; values indicate ‘mean (range within 1 s.d.)’






The data in Table 3 are pulled out from the modulatory profiling. Values are point estimates as well as 95% confidential intervals in parentheses computed using sigmoidal curve-fitting in Prism are shown from co-treatment of modulaotrs (rows) and lethal compounds (columns) in two cell lines (HT-1080 and BJeLR).









TABLE 4







Structures of 56 CILs.









Compound
Pubchem CID
Structure












CIL01
5771107


embedded image







CIL02
698314


embedded image







CIL04
2927276


embedded image







CIL05
2802124


embedded image







CIL06
776372


embedded image







CIL07
3113288


embedded image







CIL09
791069


embedded image







CIL10
3221079


embedded image







CIL11
96149


embedded image







CIL13
972880


embedded image







CIL15
2215161


embedded image







CIL16
2221997


embedded image







CIL17
1316704


embedded image







CIL18
659917


embedded image







CIL20
609767


embedded image







CIL22
779573


embedded image







CIL23
779863


embedded image







CIL25
796505


embedded image







CIL26
3732278


embedded image







CIL27
1717988


embedded image







CIL28
2855628


embedded image







CIL33
5333738


embedded image







CIL34
781106


embedded image







CIL36
2057920


embedded image







CIL40
2837694


embedded image







CIL41
2841722


embedded image







CIL44
2259838


embedded image







CIL46
5728915


embedded image







CIL47
2904443


embedded image







CIL48
1263400


embedded image







CIL49
5286934


embedded image







CIL50
5308990


embedded image







CIL51
2946035


embedded image







CIL52
2857031


embedded image







CIL55
6623618


embedded image







CIL56
654092


embedded image







CIL58
699693


embedded image







CIL60
779647


embedded image







CIL62
991372


embedded image







CIL63
779402


embedded image







CIL67
3351416


embedded image







CIL66
657546


embedded image







CIL67
6659101


embedded image







CIL69
818740


embedded image







CIL70
9551837


embedded image







CIL71
658723


embedded image







CIL72
658341


embedded image







CIL74
4406635


embedded image







CIL75
818737


embedded image







CIL76
3774616


embedded image







CIL77
6298513


embedded image







CIL79
6545175


embedded image







CIL80
1187467


embedded image







CIL87
NA


embedded image







CIL88
NA


embedded image







CIL89
NA


embedded image


















TABLE 5







CIL41/70 structural analogs.








Compound
Structure





CB122 (0.98 ug/mL)


embedded image







CB70 (1.59 ug/mL)


embedded image







VM1 (2 ug/mL)


embedded image







CB69 (2.09 ug/mL)


embedded image







CB115 (2.42 ug/mL)


embedded image







MB14 (2.6 ug/mL)


embedded image







CIL41 (2.8 ug/mL)


embedded image







B02 (3.3 ug/mL)


embedded image







CB56 (4.05 ug/mL)


embedded image







CB98 (4.46 ug/mL)


embedded image







CB147 (4.88 ug/mL)


embedded image







A05 (5.04 ug/mL)


embedded image







CB116 (5.49 ug/mL)


embedded image







MB6 (6.48 ug/mL)


embedded image







CB49 (7.23 ug/mL)


embedded image







CB140 (8.56 ug/mL)


embedded image







VM3 (8.51 ug/mL)


embedded image







CB62 (8.63 ug/mL)


embedded image







CB126 (9.3 ug/mL)


embedded image







CB18 (10.21 ug/mL)


embedded image







EN2 (10.51 ug/mL)


embedded image







CB111 (10.71 ug/mL)


embedded image







AS2 (12.26 ug/mL)


embedded image







CB14 (13.98 ug/mL)


embedded image







ZE2 (15.63 ug/mL)


embedded image







CB58 (17.61 ug/mL)


embedded image







AS1 (>20 ug/mL)


embedded image







CB1 (>20 ug/mL)


embedded image







CB2 (>20 ug/mL)


embedded image







CB3 (>20 ug/mL)


embedded image







CB4 (>20 ug/mL)


embedded image







CB5 (>20 ug/mL)


embedded image







CB6 (>20 ug/mL)


embedded image







CB7 (>20 ug/mL)


embedded image







CB8 (>20 ug/mL)


embedded image







CB9 (>20 ug/mL)


embedded image







CB10 (>20 ug/mL)


embedded image







CB11 (>20 ug/mL)


embedded image







CB12 (>20 ug/mL)


embedded image







CB13 (>20 ug/mL)


embedded image







CB15 (>20 ug/mL)


embedded image







CB16 (>20 ug/mL)


embedded image







CB17 (>20 ug/mL)


embedded image







CB19 (>20 ug/mL)


embedded image







CB20 (>20 ug/mL)


embedded image







CB21 (>20 ug/mL)


embedded image







CB22 (>20 ug/mL)


embedded image







CB23 (>20 ug/mL)


embedded image







CB24 (>20 ug/mL)


embedded image







CB25 (>20 ug/mL)


embedded image







CB26 (>20 ug/mL)


embedded image







CB27 (>20 ug/mL)


embedded image







CB28 (>20 ug/mL)


embedded image







CB29 (>20 ug/mL)


embedded image







CB30 (>20 ug/mL)


embedded image







CB31 (>20 ug/mL)


embedded image







CB32 (>20 ug/mL)


embedded image







CB33 (>20 ug/mL)


embedded image







CB34 (>20 ug/mL)


embedded image







CB35 (>20 ug/mL)


embedded image







CB36 (>20 ug/mL)


embedded image







CB37 (>20 ug/mL)


embedded image







CB38 (>20 ug/mL)


embedded image







CB39 (>20 ug/mL)


embedded image







CB40 (>20 ug/mL)


embedded image







CB41 (>20 ug/mL)


embedded image







CB42 (>20 ug/mL)


embedded image







CB43 (>20 ug/mL)


embedded image







CB44 (>20 ug/mL)


embedded image







CB45 (>20 ug/mL)


embedded image







CB46 (>20 ug/mL)


embedded image







CB47 (>20 ug/mL)


embedded image







CB48 (>20 ug/mL)


embedded image







CB50 (>20 ug/mL)


embedded image







CB51 (>20 ug/mL)


embedded image







CB52 (>20 ug/mL)


embedded image







CB53 (>20 ug/mL)


embedded image







CB54 (>20 ug/mL)


embedded image







CB55 (>20 ug/mL)


embedded image







CB57 (>20 ug/mL)


embedded image







CB59 (>20 ug/mL)


embedded image







CB60 (>20 ug/mL)


embedded image







CB61 (>20 ug/mL)


embedded image







CB63 (>20 ug/mL)


embedded image







CB64 (>20 ug/mL)


embedded image







CB65 (>20 ug/mL)


embedded image







CB66 (>20 ug/mL)


embedded image







CB67 (>20 ug/mL)


embedded image







CB68 (>20 ug/mL)


embedded image







CB70 (>20 ug/mL)


embedded image







CB71 (>20 ug/mL)


embedded image







CB72 (>20 ug/mL)


embedded image







CB73 (>20 ug/mL)


embedded image







CB74 (>20 ug/mL)


embedded image







CB75 (>20 ug/mL)


embedded image







CB76 (>20 ug/mL)


embedded image







CB77 (>20 ug/mL)


embedded image







CB78 (>20 ug/mL)


embedded image







CB79 (>20 ug/mL)


embedded image







CB80 (>20 ug/mL)


embedded image







CB81 (>20 ug/mL)


embedded image







CB82 (>20 ug/mL)


embedded image







CB83 (>20 ug/mL)


embedded image







CB84 (>20 ug/mL)


embedded image







CB85 (>20 ug/mL)


embedded image







CB86 (>20 ug/mL)


embedded image







CB87 (>20 ug/mL)


embedded image







CB88 (>20 ug/mL)


embedded image







CB89 (>20 ug/mL)


embedded image







CB90 (>20 ug/mL)


embedded image







CB91 (>20 ug/mL)


embedded image







CB92 (>20 ug/mL)


embedded image







CB93 (>20 ug/mL)


embedded image







CB94 (>20 ug/mL)


embedded image







CB95 (>20 ug/mL)


embedded image







CB96 (>20 ug/mL)


embedded image







CB97 (>20 ug/mL)


embedded image







CB99 (>20 ug/mL)


embedded image







CB100 (>20 ug/mL)


embedded image







CB101 (>20 ug/mL)


embedded image







CB102 (>20 ug/mL)


embedded image







CB103 (>20 ug/mL)


embedded image







CB104 (>20 ug/mL)


embedded image







CB105 (>20 ug/mL)


embedded image







CB106 (>20 ug/mL)


embedded image







CB107 (>20 ug/mL)


embedded image







CB108 (>20 ug/mL)


embedded image







CB109 (>20 ug/mL)


embedded image







CB110 (>20 ug/mL)


embedded image







CB112 (>20 ug/mL)


embedded image







CB113 (>20 ug/mL)


embedded image







CB114 (>20 ug/mL)


embedded image







CB117 (>20 ug/mL)


embedded image







CB118 (>20 ug/mL)


embedded image







CB119 (>20 ug/mL)


embedded image







CB120 (>20 ug/mL)


embedded image







CB121 (>20 ug/mL)


embedded image







CB123 (>20 ug/mL)


embedded image







CB124 (>20 ug/mL)


embedded image







CB125 (>20 ug/mL)


embedded image







CB127 (>20 ug/mL)


embedded image







CB128 (>20 ug/mL)


embedded image







CB129 (>20 ug/mL)


embedded image







CB130 (>20 ug/mL)


embedded image







CB131 (>20 ug/mL)


embedded image







CB132 (>20 ug/mL)


embedded image







CB133 (>20 ug/mL)


embedded image







CB134 (>20 ug/mL)


embedded image







CB135 (>20 ug/mL)


embedded image







CB136 (>20 ug/mL)


embedded image







CB137 (>20 ug/mL)


embedded image







CB138 (>20 ug/mL)


embedded image







CB139 (>20 ug/mL)


embedded image







CB141 (>20 ug/mL)


embedded image







CB142 (>20 ug/mL)


embedded image







CB143 (>20 ug/mL)


embedded image







CB144 (>20 ug/mL)


embedded image







CB145 (>20 ug/mL)


embedded image







CB146 (>20 ug/mL)


embedded image







CB148 (>20 ug/mL)


embedded image







CD1 (>20 ug/mL)


embedded image







EN1 (>20 ug/mL)


embedded image







EN3 (>20 ug/mL)


embedded image







EN4 (>20 ug/mL)


embedded image







EN5 (>20 ug/mL)


embedded image







EN6 (>20 ug/mL)


embedded image







EN7 (>20 ug/mL)


embedded image







EN8 (>20 ug/mL)


embedded image







EN9 (>20 ug/mL)


embedded image







EN10 (>20 ug/mL)


embedded image







EN11 (>20 ug/mL)


embedded image







MB1 (>20 ug/mL)


embedded image







MB2 (>20 ug/mL)


embedded image







MB3 (>20 ug/mL)


embedded image







MB4 (>20 ug/mL)


embedded image







MB5 (>20 ug/mL)


embedded image







MB7 (>20 ug/mL)


embedded image







MB8 (>20 ug/mL)


embedded image







MB9 (>20 ug/mL)


embedded image







MB10 (>20 ug/mL)


embedded image







MB11 (>20 ug/mL)


embedded image







MB12 (>20 ug/mL)


embedded image







MB13 (>20 ug/mL)


embedded image







MB15 (>20 ug/mL)


embedded image







MB16 (>20 ug/mL)


embedded image







MB17 (>20 ug/mL)


embedded image







VM2 (>20 ug/mL)


embedded image







VM4 (>20 ug/mL)


embedded image







VM5 (>20 ug/mL)


embedded image







ZE1 (>20 ug/mL)


embedded image







ZE3 (>20 ug/mL)


embedded image







ZE4 (>20 ug/mL)


embedded image







ZE5 (>20 ug/mL)


embedded image







ZE6 (>20 ug/mL)


embedded image







ZE7 (>20 ug/mL)


embedded image







ZE8 (>20 ug/mL)


embedded image







ZE9 (>20 ug/mL)


embedded image







ZE10 (>20 ug/mL)


embedded image







ZE11 (>20 ug/mL)


embedded image







ZE12 (>20 ug/mL)


embedded image







ZE13 (>20 ug/mL)


embedded image







ZE14 (>20 ug/mL)


embedded image







ZE15 (>20 ug/mL)


embedded image







ZE16 (>20 ug/mL)


embedded image







ZE17 (>20 ug/mL)


embedded image











Example 2
Modulatory Profiling Revealed Three Kinds of Cell Death

To map the landscape of cell death, small-molecule inducers of regulated, non-apoptotic cell death were searched (FIG. 1A). Total of 3,169 lethal compounds were tested for induction of caspase-independent cell death in HT-1080 fibrosarcoma cells and BJeLR engineered transformed fibroblasts, cell lines used previously for modulatory profiling experiments (Hahn et al. 1999) (see Table 1). It was found that 451 compounds (14%) triggered cell death without activation of caspases 3 and 7, detected using a fluorogenic substrate. These compounds were defined as ‘caspase-3/7-independent lethals’ (CILs) (FIG. 7, and Tables 4 and 5). Thus, although most lethal compounds activate caspase activity (irrespective of whether caspase activity is required for their lethality), it was found that a considerable number of compounds were lethal without activating cleavage of the fluorogenic caspase-activity probe.


Fifty-six structurally diverse and potent (EC80<2.8 μg ml−1) CILs were further examined using a modulatory profiling strategy. Previously, the clustering of modulatory profiles, or changes in the potency and efficacy of a lethal compound induced by cotreatment with chemical and genetic death modulators (Table 2), revealed that compounds with the same mechanism of action share similar modulatory profiles (Wolpaw et al. 2011). For evaluation of diverse regulated cell death programs, modulatory profiling should facilitate the grouping of pharmacological agents by their induction of specific regulated cell death programs. It was observed that ten compounds induced three types of regulated non-apoptotic cell death. A specific ferroptosis inducer was further identified through lead optimization and its mechanism of action was uncovered. In this analysis, 10 of the 56 CIL compounds exhibited ‘high modulatability’, meaning their lethality was considerably suppressed or enhanced by specific pharmacological or genetic agents (Wolpaw et al. 2011) (FIG. 8 and Tables 6-9); high modulatability correlates with activity through a specific lethal pathway. When analyzed along with other well-characterized lethal compounds, these CILs fell into three classes (FIG. 1B and FIG. 9A). The first class (CIL13, CIL52, and CIL64) acted via metal chelation, as these compounds were inhibited by cobalt (II) and bound to cobalt (II) in vitro (FIGS. 9B-9C). The second class (CIL62) induced cell death that was suppressed by necrostatin-1 (Degterev et al. 2005) (FIGS. 9D-9E; note that this does not necessarily imply necroptosis, as necrostatin-1 has necroptosis-independent effects (Takahashi et al. 2012)), and the third class (CIL41, CIL56 (1), CIL69, CIL70, CIL75, and CIL79) comprised ferroptosis inducers, as suggested by their suppression by canonical ferroptosis inhibitors (iron chelators and lipophilic antioxidants; Table 3) and clustering with known ferroptosis inducers. Six CIL compounds that clustered with ferroptosis inducers were selected for further studies.


Of the six ferroptotic CILs, three (CIL69, CIL75, and CIL79) are putative electrophiles and clustered most closely with known electrophilic ferroptosis inducers, such as (1S, 3R)-RSL3 (FIG. 10A). The remaining CILs consisted of two novel scaffolds: CIL41/70 and CIL56. CIL41/70 induced ROS accumulation, detected using H2-DCFDA (FIG. 10B), and cell death that was strongly suppressed by ferroptosis inhibitors (i.e., the lipophilic antioxidant α-tocopherol and the iron chelator deferoxamine) (FIG. 10C).


All known ferroptosis inducers induce selective lethality in a BJ engineered cell line series, namely, BJeH, BJeHLT, DRD, and BJeLR cells. These cells were initially created to demonstrate that normal human fibroblasts can be transformed into tumor cells by the introduction of defined genetic elements (human telomerase, SV40 small and large T antigens, and oncogenic HRASG12V) (Hahn et al. 1999). BjeLR cells and DRD cells, which overexpress oncogenic RAS, were found to be more sensitive to ferroptosis inducers than BJeHLT cells and BJeH cells, which do not express oncogenic RAS. CIL41/70, unlike all other ferroptosis-inducing compounds reported to date, did not exhibit oncogenic RAS selectivity in the BJ engineered cell line series (Yang et al. 2008) (FIG. 10D). Moreover, 203 commercially available structural analogs of CIL41/70 were tested and they were less potent than CIL56 itself (Tables 4-5 and FIG. 10E). Given that CIL56 was the most potent compound, and that it retained some degree of selectivity toward oncogenic-RAS-expressing cells in the BJ series (FIG. 2A), CIL56 was believed to be more likely than CIL41/70 to yield a potent and selective probe of ferroptosis. Thus, CIL56 was selected for more detailed characterization.









TABLE 6







List of lethal compounds.










name
known mechanism of action







MS275
HDAC inhibitor



Scriptaid
HDAC inhibitor



TrichostatinA
HDAC inhibitor



Echinomycin
HIF1a inhibitor



Colchicine
microtubule inhibitor



NPC25
microtubule inhibitor



NPC4
microtubule inhibitor



NPC7
microtubule inhibitor



Podophyllotoxin
microtubule inhibitor



Rotenone
microtubule inhibitor



Vinblastine
microtubule inhibitor



Vincristine
microtubule inhibitor



DPI3
ferroptosis (electrophile)



DPI4
ferroptosis (electrophile)



DPI6
ferroptosis (electrophile)



RSL3
ferroptosis (electrophile)



Bortezomib
Proteasome inhibitor



MG132
Proteasome inhibitor



MG262
Proteasome inhibitor



Erastin
ferroptosis



Camptothecin
Topol inhibitor



Irinotecan
Topol inhibitor



Daunorubicin
Topoll inhibitor



Doxorubicin
Topoll inhibitor



Etoposide
Topoll inhibitor



Mitoxantrone
Topoll inhibitor



Cycloheximide
Translational inhibitor



Dinitrophenol
mitochondrial uncoupler



NaN3
mitochondrial uncoupler



Valinomycin
mitochondrial uncoupler



CIL1
NA



CIL2
NA



CIL4
NA



CIL5
NA



CIL6
NA



CIL7
NA



CIL9
NA



CIL10
NA



CIL11
NA



CIL13
Transition metal chelator



CIL15
NA



CIL16
NA



CIL17
NA



CIL18
NA



CIL20
NA



CIL22
NA



CIL23
NA



CIL25
NA



CIL26
NA



CIL27
NA



CIL28
NA



CIL33
NA



CIL34
NA



CIL36
NA



CIL40
NA



CIL41
ferroptosis



CIL44
NA



CIL46
NA



CIL47
NA



CIL48
NA



CIL49
NA



CIL50
NA



CIL51
NA



CIL52
Transition metal chelator



CIL55
NA



CIL56
ferroptosis



CIL58
NA



CIL60
NA



CIL62
Nec-1 dependent cell death



CIL63
NA



CIL64
Transition metal chelator



CIL66
NA



CIL67
NA



CIL69
ferroptosis (electrophile)



CIL70
ferroptosis



CIL71
NA



CIL72
NA



CIL74
NA



CIL75
ferroptosis (electrophile)



CIL76
NA



CIL77
NA



CIL79
ferroptosis (electrophile)



CIL80
NA



CIL87
NA



CIL88
NA



CIL89
NA

















TABLE 7







List of chemical death modulators.
















tested





conc


chemicals
full name
known biological function
(μM)













atoc
α-tocopherol
Antioxidant
100


bCarotene
β-carotene
Antioxidant
0.19


BHA
Butylated hydroxyanisole
Antioxidant
139


BHT
Butylated hydroxytoluene
Antioxidant
113


trolox
(±)-6-Hydroxy-2,5,7,8-tetramethylchromane-
Antioxidant
150



2-carboxylic acid




CspA
Cyclosporin A
Binds cyclophilin
5


Co2+
Cobalt (II)
calcium channel blocker
656


Gd3+
Gadolinium (III)
calcium channel blocker
20


EGTA
Ethyleneglycol-O,O′-bis(2-amino ethyl)-
calcium(II) chelator
2000



N,N,N′,N′-tetraacetic acid




Pepstatin
Pepstatin
cathepsin D inhibitor
1


Lmim
L-mimosine
G1-S cell-cycle inhibitor, iron chelator
175


3MA
3-methyladenine
Inhibitor of autophagosome formation
5000


ALLN
Calpain Inhibitor 1
Inhibitor of calpain land II, cathepsins B,I
6.3


Nec-1
Necrostatin-1
Inhibitor of necroptosis
19


deferox
Deferoxamine
iron chelator
152


SP600125
Anthra(1,9-cd)pyrazol-6(2H)-one1,9-
JNK inhibitor
10



Pyrazoloanthrone




U0126
1,4-diamino-2,3-dicyano-1,4-bis[2-
MEK1/2 inhibitor
13.1



aminophenylthio)]butadiene




digoxin
Digoxin
Na+/K+ ATPase inhibitor
0.13


LNAME
NG-Nitro-L-arginine-methyl ester
Nitric oxide synthase inhibitor
300


NMMA
NG-Monomethyl-D-arginine
Nitric oxide synthase inhibitor
20


ATA
Aurintricarboxylic Acid
Nuclease Inhibitor
38


BocD
t-butoxycarbonyl-Asp-fluomethylketone
pan-caspase inhibitor
50


zVAD
Cbz-Val-Ala-Asp(OMe)-fluormethylketone
pan-caspase inhibitor
45


DPQ
3,4-dihydro-5-[4-(1-piperidinyl)butoxy]-
PARP1 inhibitor
10



1(2H)-isoquinolinone




CHX
Cycloheximide
protein synthesis inhibitor
1.5


ActD
Actinomycin D
RNA polymerase inhibitor
0.002


TLCK
Nα-tosyl-L-Lys-chloro methyl ketone
serine protease inhibitor
135


NAD+
Nicotinamide Adenine Dinucleotide
Sirtuin activator, prevents ATP depletion
2000


L/mim/NAD+
L-mimosine and NAD+ co-treatment

175/2000
















TABLE 8







List of cell lines.









cells
full name
description





BJ
BJeLR
BJ fibroblast overexpressing hTert/




SV40LT/SV40ST/HrasG12V


HT
HT-1080
fibrosarcoma harboring NrasQ61K
















TABLE 9





Modulatory profiles of characterized and uncharacterized lethal compounds.





















BJ.3MA
BJ.ALLN
BJ.Boc D
BJ.CHX
BJ.Co2+





MS275
−0.026059742
−0.039317112
 0.089435721
 0.226620516
 0.232568502


Scriptaid
−0.020600458
−0.078560038
 0.031670618
 0.268137819
 0.240617478


TrichostatinA
−0.004309497
−0.087739323
 0.096123823
 0.258751821
 0.309126126


Echinomycin
 0.345698628
 0.02426368
 0.049246386
 0.201520598
 0.093210997


Colchicine
 0.003025932
−0.033558465
−0.000908197
 0.160992151
 0.168765369


NPC25
−0.025141082
−0.011859418
 0.0284768
 0.338638606
 0.205559338


NPC4
−0.044629775
−0.022535714
−0.009005742
 0.231995694
 0.151230708


NPC7
−0.023899287
−0.018930504
−0.00265144
 0.211134268
 0.080494659


Podophyllotoxin
−0.01267149
−0.003672686
 0.003541897
 0.122809732
 0.094971921


Rotenone
−0.026596072
−0.034348313
−0.016740744
 0.106622385
 0.083795618


Vinbiastine
 0.013104608
−0.072276828
 0.001717399
 0.232808367
 0.187592903


Vincristine
−0.03566309
−0.031708431
−0.005015919
 0.195548157
 0.180588109


DPI3
 0.095722195
−0.072359809
−0.00375885
−0.044207783
 0.155678052


DPI4
 0.081304775
−0.075963448
−0.020600089
−0.044823823
 0.115974336


DPI6
 0.061477039
−0.054000329
−0.018112304
−0.069274018
 0.10520246


RSL3
 0.091035174
 0.012662457
 0.038324402
 0.184102513
 0.109106536


Bortezomib
 0.001927724
−0.103124775
 0.030321225
 0.298426073
 0.237919799


MG132
−0.043463292
−0.270072286
 0.042157364
 0.247614973
 0.184937083


MG262
−0.036195527
−0.198870912
 0.017555267
 0.243797991
 0.110875339


Erastin
 0.093482003
−0.057509042
−0.000616511
 0.090809688
−0.170369309


Camptothecin
−0.020270594
 0.033675205
 0.079833441
 0.123280564
 0.028994781


Irinotecan
−0.046340941
 0.015018658
 0.070216151
 0.124862147
 0.028550947


Daunorubicin
 0.037934083
 0.028460875
 0.033216728
 0.149732833
 0.212207875


Doxorubicin
 0.031127831
 0.015547275
 0.049656193
 0.14122675
 0.197065807


Etoposide
−0.061955521
 0.060991381
 0.061201223
 0.125509938
 0.095103567


Mitoxantrone
 0.033515749
 0.062035067
 0.037464053
 0.143696892
 0.453843256


Cycloheximide
 0.038465272
 0.094614356
 0.02242339
 0.361284466
 0.237543606


Dinitrophenol
−0.008182185
−0.038382592
−0.01714167
 0.189038538
−0.008315819


NaN3
−0.095593638
−0.003819896
−0.007272953
 0.072620828
−0.021225548


Vatinomycin
 0.033259329
 0.020633509
 0.020464639
 0.357294884
 0.095951635


CIL1
 0.094702761
−0.033187356
−0.046498523
−0.08681018
 0.095722858


CIL2
 0.064313223
−0.039784978
−0.03807957
 0.03999122
−0.136186971


CIL4
 0.083161827
−0.003082963
−0.018283858
−0.009993254
−0.17469724


CIL5
 0.085235042
−0.116283699
−0.07609025
−0.032403457
 0.047047513


CIL6
 0.119061436
−0.049603536
−0.052467687
 0.135265612
 0.069605114


CIL7
 0.091880431
−0.028761721
−0.049458392
−0.010559719
 0.09998614


CIL9
 0.046502556
−0.120198756
−0.061705214
 0.03696055
−0.009162007


CIL10
 0.134017231
−0.017134117
−0.052123672
−0.018050509
 0.114193588


CIL11
 0.090044215
 0.015376964
−0.043226998
 0.123905521
 0.077708109


CIL13
 0.090834135
−0.039328117
−0.029889688
−0.126427664
 0.528659723


CIL15
 0.027611875
−0.06757043
−0.053211843
 0.12872254
 0.096097693


CIL16
 0.05063654
−0.034913398
−0.046915645
 0.012206749
 0.072067228


CIL17
 0.079269682
−0.074781483
−0.031155382
−0.046204279
−0.047640395


CIL18
 0.06540499
−0.045068104
−0.018410753
−0.015819111
 0.089434829


CIL20
 0.055003817
−0.090980632
−0.063733658
−0.009676654
 0.047634171


CIL22
−0.043294993
−0.032323012
−0.040599287
 0.113573387
 0.076154164


CIL23
−0.016130596
−0.045611913
−0.01959298
−0.023034675
−0.015941685


CIL25
 0.023740381
−0.07208946
−0.009758131
−0.02730202
−0.003244445


CIL26
 0.05123851
−0.058398468
−0.05573532
−0.001732868
 0.048705995


CIL27
 0.040295075
−0.066042535
−0.080491998
 0.139881194
 0.059305657


CIL28
 0.043299829
−0.020647414
−0.025487472
−0.011194165
 0.034165753


CIL33
 0.0619933
−0.112365796
−0.032294484
 0.102623158
 0.043114331


CIL34
 0.077008824
−0.043739268
−0.024624589
 0.121487032
 0.07311128


CIL36
 0.062946001
−0.032362618
−0.047302904
−0.030872077
−0.036468619


CIL40
 0.113089292
−0.025923038
−0.056067715
−0.1422925
 0.033483801


CIL41
 0.116955468
−0.082112839
−0.063738367
−0.057101897
−0.023784749


CIL44
 0.05755856
−0.058803806
−0.039629515
 0.155313574
 0.023867911


CIL46
 0.188432008
−0.013738553
−0.005903073
 0.15984289
−0.138932367


CIL47
 0.150522382
−0.026277586
 0.053970843
−0.02690468
−0.137946997


CIL48
 0.097888892
−0.100752292
−0.022253324
 0.161924385
 0.053915436


CIL49
 0.045036307
−0.058638439
−0.025704178
 0.169190352
 0.009716111


CIL50
−0.00161299
−0.008923829
 0.031127244
 0.141459593
−0.017803771


CIL51
 0.059081237
−0.203188928
−0.000882839
 0.169570765
 0.063992863


CIL52
 0.011896568
 0.010918623
 0.018402924
 0.037243011
 0.7977955


CIL55
 0.063543912
−0.046846172
−0.014987852
 0.037300771
 0.03522058


CIL56
 0.145895802
 0.058308053
−0.010427691
−0.072214121
 0.126264931


CIL58
 0.034027705
 0.018458023
−0.007995419
−0.049112036
 0.238560717


CIL60
 0.067356791
−0.004196565
 0.026257063
 0.180280969
 0.038358824


CIL62
 0.009761947
−0.062646512
−0.008657967
 0.516222526
 0.015737354


CIL63
 0.040754174
 0.05440433
−0.010529835
 0.095083008
 0.021772384


CIL64
 0.02464908
 0.014202785
 0.011077585
 0.047118757
 0.940237984


CIL66
 0.041676948
 0.087467313
−0.03145846
 0.083694083
−0.031407325


CIL67
 0.023124894
 0.021494083
 0.005780137
 0.308266858
 0.037082001


CIL69
 0.077127838
−0.000522215
−0.003227696
−0.03495043
 0.172373945


CIL70
 0.101356041
−0.07035852
 0.00241214
 0.130415593
−0.000492503


CIL71
 0.011512823
−0.055893307
 0.015201388
−0.063520521
 0.026760788


CIL72
 0.017140014
 0.004589548
 0.026716057
 0.25849909
 0.117570684


CIL74
 0.036151004
−0.025360323
 0.043242954
 0.014664752
 0.213514501


CIL75
 0.046533481
 0.013011291
 0.03091594
−0.009102995
 0.204879929


CIL76
−0.036382826
−0.053597155
 0.032366316
 0.260846556
 0.167625358


CIL77
−0.057281353
 0.018263122
 0.056223402
 0.25480136
 0.186570503


CIL79
 0.07979903
 0.027169014
 0.029379264
−0.046505369
 0.176254231


CIL80
 0.006540475
−0.004769237
 0.008783432
 0.135118471
 0.095062234


CIL87
−0.002763545
−0.08720998
−0.013398191
 0.295017043
 0.141646385


CIL88
 0.030934243
−0.142376485
−0.01445462
 0.235378859
 0.053789327


CIL89
−0.01648407
−0.120355803
 0.03243382
 0.19676153
 0.017205043






BJ.DPQ
BJ.Gd3+
BJ.LNAME
BJ.Lmin
BJ.Lmin/NAD+





MS275
−0.0096557
 0.02299609
 0.02320658
−0.0011826
 0.156946048


Scriptaid
 0.00489165
 0.0289045
 0.03611375
 0.11129574
 0.211416066


TrichostatinA
−0.0095226
−0.0088019
−0.0420318
 0.08422628
 0.159662164


Echinomycin
−0.0043253
 0.00204084
 0.01198552
 0.12050248
 0.235093929


Colchicine
−0.0305343
−0.010606
−0.0085479
 0.05808173
 0.140911627


NPC25
−0.0020323
 0.00764977
 0.01842301
 0.15871171
 0.181745644


NPC4
−0.0121288
 0.01108163
 0.03470683
 0.12282908
 0.189076249


NPC7
−0.0111977
 0.00040178
 0.02663201
 0.10580065
 0.20079283


Podophyllotoxin
−0.0389641
 0.01784317
 0.00586807
 0.05456671
 0.101224933


Rotenone
−0.022785
 0.01421305
−0.011312
 0.06491961
 0.143857129


Vinbiastine
−0.0311986
 0.00835127
−0.0006349
 0.0658354
 0.171479031


Vincristine
−0.030439
 0.00794382
−0.0071703
 0.06034138
 0.172069929


DPI3
 0.05023093
 0.05621465
 0.0011647
 0.15320194
 0.19306387


DPI4
 0.02148024
 0.06864379
 0.00316962
 0.14433436
 0.14432113


DPI6
 0.03271803
 0.03553869
 0.02698599
 0.12687654
 0.086661743


RSL3
 0.03115236
 0.02447626
 0.02931886
 0.34623405
 0.385813695


Bortezomib
−0.0073919
−0.03227
−0.0086492
 0.09993315
 0.149126821


MG132
−0.0132706
 0.03162845
 0.01788625
 0.14869399
 0.206416573


MG262
 0.00506418
 0.02134418
 0.02271737
 0.14366619
 0.152015466


Erastin
−0.0050575
−0.0387997
−0.0397056
 0.11149699
 0.153795532


Camptothecin
−0.0111881
−0.007898
 0.00320584
 0.0294683
 0.116436878


Irinotecan
 0.04602433
−0.0076941
 0.01821737
 0.018475
 0.080917834


Daunorubicin
−0.0352683
−0.0052472
 0.00552589
 0.05475302
 0.143361824


Doxorubicin
 0.00446182
−0.0110536
 0.01381765
 0.05475337
 0.147679846


Etoposide
−0.0468961
 0.01316612
 0.00451468
 0.04682368
 0.101123735


Mitoxantrone
−0.0581416
 0.0106939
 0.00757779
 0.0436762
 0.116200284


Cycloheximide
 0.02122104
 0.02359332
 0.00769629
 0.07750166
 0.105686189


Dinitrophenol
−0.0325051
−0.0141327
−0.0202006
−0.0204979
 0.076205151


NaN3
−0.0694736
 0.01113264
−0.0183243
−0.0841719
−0.007837039


Vatinomycin
−0.0220075
−0.0004394
−0.0132299
−0.0078469
 0.152525695


CIL1
−0.01655
−0.0015775
−0.0210343
−0.0142017
−0.019472966


CIL2
 0.0039287
 0.01321642
−0.004423
 0.0083184
−0.019443953


CIL4
−0.0039688
 0.05533373
−0.0128845
−0.0028673
 0.086739015


CIL5
 0.00287621
 0.01145922
−0.0097869
−0.036977
−0.017375505


CIL6
 0.01365177
 0.00684778
−0.0263723
−0.0387383
−0.063801914


CIL7
 0.06720757
−0.0189741
−0.0318209
 0.01256103
 0.017714492


CIL9
−0.0122469
−0.0103755
 0.01335948
−0.0396671
 0.014992517


CIL10
−0.0269473
 0.00792172
 0.01363382
−0.0234054
−0.013959025


CIL11
−0.0156617
 0.02904746
−0.0003217
−0.0403399
 0.013439329


CIL13
 0.00739191
 0.04262745
 0.0154421
−0.0341183
−0.054563077


CIL15
−0.0123768
 0.02534821
−0.0245318
−0.0004619
−0.01163706


CIL16
−0.0173163
−0.0250626
−0.0127813
−0.0223794
−0.05229458


CIL17
 0.00467941
−0.0400009
−0.0080537
−0.02464
−0.032086352


CIL18
−0.0131845
−0.0165879
−0.0162754
−0.0550532
−0.050203511


CIL20
−0.0269595
 0.04265912
−0.0017472
−0.0260292
−0.03733821


CIL22
 0.00575155
 0.00589901
 0.01229865
 0.002645
 0.072783496


CIL23
 0.0043789
 0.03265453
−0.0212857
−0.0161655
 0.004125464


CIL25
 0.00185155
 0.04216016
−0.0044279
 0.0162808
 0.09245744


CIL26
 0.01829107
 0.00454073
−0.033216
−0.013902
 0.091113219


CIL27
 0.01710026
−0.0240573
−0.0438093
 0.0147174
 0.023647992


CIL28
 0.00923459
−0.0774082
−0.0485835
−0.052998
−0.025777883


CIL33
 0.02548824
−0.0653931
−0.0315674
 0.01529408
−0.012603667


CIL34
 0.03951199
−0.0319954
−0.016312
−0.0234022
 0.042574773


CIL36
 0.13933756
 0.01469476
 0.0008728
 0.02335875
−0.094025018


CIL40
 0.01886595
−0.0047375
 0.00219723
−0.0527296
−0.06069381


CIL41
 0.02160659
−0.0067674
 0.00306986
 0.20320555
 0.150431554


CIL44
 0.05603523
 0.00555618
 0.00898119
 0.03065188
−0.049754257


CIL46
 0.03979168
−0.0560629
−0.0218477
 0.25833718
 0.18247054


CIL47
 0.06122321
−0.0287511
−0.024789
−0.0169586
−0.014373272


CIL48
 0.01111515
−0.0043066
−0.0103879
−0.0266405
 0.063259258


CIL49
−0.002084
−0.0060484
−0.0078571
 0.03689207
 0.070671325


CIL50
−0.0386937
−0.0211267
 0.03227487
 0.03122751
 0.016756946


CIL51
 0.04077456
 0.03262832
−0.0414541
−0.0741306
 0.034681874


CIL52
 0.03648385
 0.08326996
−0.0157785
 0.0077833
 0.082646149


CIL55
 0.01735295
 0.00355544
−0.0200804
 0.08142775
 0.192397007


CIL56
 0.03219032
 0.00499655
 0.00597842
 0.1358207
 0.219400696


CIL58
 0.00992247
 0.02130798
−0.0281144
−0.0091724
 0.027134971


CIL60
 0.02056594
 0.02704229
−0.0154202
 0.04196132
 0.058879954


CIL62
−0.0057516
 0.00294553
−0.0205525
 0.01862207
 0.087356675


CIL63
 0.00030462
 0.01459045
−0.0068124
 0.01214827
 0.077758217


CIL64
 0.00939684
 0.02721015
 0.03248984
 0.0277705
 0.075703433


CIL66
−0.0072839
−0.0038385
 0.02705047
−0.0519479
 0.064436594


CIL67
−0.0050886
 0.01429813
 0.01176843
−0.0878664
 0.159935957


CIL69
 0.02521299
 0.02982599
 0.00520114
 0.17994869
 0.144850955


CIL70
 0.02973309
 0.04884126
−0.0060271
 0.20617772
 0.263347395


CIL71
 0.03052411
 0.01393906
 0.00120448
−0.0213785
 0.001502008


CIL72
−0.0108661
 0.0301285
 0.00105307
 0.00484398
 0.071016136


CIL74
−0.0290634
 0.02962909
 0.00161925
 0.00776593
 0.071540234


CIL75
−1.616E−05
 0.04589641
 0.01183514
 0.14596013
 0.229245422


CIL76
 0.00735075
 0.01564981
 0.00434473
−0.0058295
−0.026560841


CIL77
−0.0092733
−0.0073443
 0.0334856
−0.0484787
 0.049815598


CIL79
 0.00396667
 0.00956035
 0.01543874
 0.20485377
 0.213581808


CIL80
−0.033684
 0.01802279
 0.0290131
 0.01638807
 0.092706078


CIL87
−0.0320238
 0.01489917
 0.02319253
 0.00077144
 0.076394758


CIL88
−0.0360775
−0.0365172
 0.0201045
−0.0124052
 0.033993856


CIL89
−0.0259805
−0.0703761
 0.03646289
 0.03550987
 0.040691428






BJ.NAD+
BJ.NMMA
BJ.Nec-1
BJ.Pepstatin
BJ.TLCK





MS275
 0.12689755
−0.0037791
−0.0132868
−0.01823965
 0.08473737


Scriptaid
 0.13840299
 0.02102042
 0.02803847
−0.02820409
 0.07545029


TrichostatinA
 0.05543793
−0.0186055
−0.0475068
−0.01284083
 0.0287911


Echinomycin
 0.22918456
 0.01082334
−0.0190088
−0.00436871
−0.001724


Colchicine
 0.11282645
−0.0052205
−0.0444276
 0.00366931
−0.0133804


NPC25
 0.07747315
−0.0077502
 0.02693649
 0.00041587
 0.00565247


NPC4
 0.00303668
−0.0106621
 0.05591668
−0.00309854
−0.0010738


NPC7
 0.03954194
 0.00518631
 0.07596296
 0.0081013
−0.0072504


Podophyllotoxin
 0.0962667
−0.0070434
−0.0416706
 0.02214061
−0.0204248


Rotenone
 0.10368941
 0.00580134
−0.0288816
 0.0200717
−0.0216911


Vinbiastine
 0.13733224
 0.00810716
−0.0617589
 0.00239815
−0.0036716


Vincristine
 0.12547697
−0.0033537
−0.0557183
−0.00252147
−0.0125536


DPI3
 0.02391881
 0.01402271
 0.13878977
 0.0339671
 0.23010879


DPI4
 0.03153366
 0.00496096
 0.12949554
 0.03965648
 0.16121299


DPI6
 0.01054535
−0.0078233
 0.16668804
 0.01992931
 0.1418306


RSL3
 0.04136024
 0.00780566
 0.23792845
−0.00080934
 0.27316475


Bortezomib
 0.05174981
−0.0002968
 0.00229317
 0.00069802
 0.07108188


MG132
 0.04273584
 0.02073008
−0.0504018
−0.02291037
 0.1241401


MG262
 0.06083716
−0.0069281
−0.0134217
 0.00662351
 0.09405199


Erastin
−0.0233386
−0.0562522
 0.16649238
 0.18917929
 0.01550359


Camptothecin
 0.11203217
−0.006852
 0.02477381
 0.01408015
 0.02306401


Irinotecan
 0.08680983
−0.0151973
 0.05502616
 0.01485256
 0.01158128


Daunorubicin
 0.12262643
−0.0186743
−0.020549
 0.02538848
 0.02855852


Doxorubicin
 0.13394249
−0.0074862
−0.0036772
 0.0177527
 0.03306748


Etoposide
 0.08358535
−0.0367759
−0.0435391
 0.01632262
 0.01398922


Mitoxantrone
 0.08732533
 0.00443081
−0.0303752
 0.00326268
 0.02288009


Cycloheximide
 0.08265474
 0.00253528
 0.03326419
 0.01474665
 0.03152874


Dinitrophenol
 0.069856
−0.0273529
−0.0019545
 0.05394491
−0.0425959


NaN3
 0.06959726
 0.00587385
−0.0522878
 0.01610826
 0.03199676


Vatinomycin
 0.11006403
−0.0263396
 0.01733615
 0.04946152
 0.00660171


CIL1
 0.039405
−0.0249794
 0.08160063
 0.0763918
 0.01374571


CIL2
 0.03383486
−0.0052336
 0.03180184
 0.05923483
 0.06974481


CIL4
 0.02730981
−0.0156562
 0.10151633
 0.08230554
 0.06929196


CIL5
 2.99E−05
−0.0270966
 0.03195004
 0.06042127
−0.0260298


CIL6
 0.02657028
 0.00628736
 0.01237172
 0.0371055
−0.0153571


CIL7
 0.00904591
−0.0153267
 0.02534044
 0.06230936
−0.0237259


CIL9
 0.00071813
−0.0154109
 0.02689145
 0.01819724
−0.0175244


CIL10
−0.001182
 0.00619197
 0.07272333
 0.00104409
−0.0583071


CIL11
−0.0391938
−0.0037798
 0.02973909
 0.04124625
−0.0115517


CIL13
 0.01377788
−0.0317716
 0.0473837
 0.03783902
 0.3411674


CIL15
 0.01286697
−0.0362316
 0.02390543
 0.02560014
−0.0056295


CIL16
−0.0217694
−0.0193843
 0.06902775
 0.02480838
−0.0251636


CIL17
−0.0163243
−0.0168812
 0.02959089
 0.03911776
−0.0786355


CIL18
 0.00749025
−0.0237147
 0.02259594
 0.04723511
−0.0382445


CIL20
 0.0304319
−0.010461
 0.01149984
−0.00251563
−0.0074299


CIL22
−0.005902
−0.007412
−0.0163259
 0.02723931
−0.0132068


CIL23
 0.00510439
−0.0124185
 0.03269009
−0.00166279
−0.0078056


CIL25
 0.061062
 0.01682877
 0.03297461
−0.00414972
−0.0588887


CIL26
 0.0056784
 0.00678111
−0.0397433
 0.02267646
−0.0613286


CIL27
−0.0135746
 0.00947413
−0.0545169
 0.00650879
−0.0306493


CIL28
 0.06230594
 0.00026396
 0.00573701
 0.06165907
−0.0941654


CIL33
 0.05051157
 0.04580018
−0.0895292
 0.07671621
−0.1077058


CIL34
−0.0043717
 0.03325703
−0.0385173
 0.04418365
−0.1072627


CIL36
−0.0182644
 0.03927146
−0.1729066
−0.01890624
−0.0433688


CIL40
 0.00558711
 0.05460608
 0.02441649
−0.01642406
−0.0535803


CIL41
−0.0197993
 0.0421872
 0.09368387
−0.03127794
−0.0246306


CIL44
−0.0416701
 0.02894312
−0.0111888
 0.00245544
−0.039631


CIL46
−0.0237067
 0.03362854
 0.08455105
−0.01312611
−0.097147


CIL47
−0.0635645
 0.04023428
−0.0220279
 0.0545364
−0.0376769


CIL48
−0.0424572
 0.01412088
−0.0443391
−0.01271153
−0.0133895


CIL49
 0.02344296
 0.00689813
 0.00197172
 0.02852214
−0.0354854


CIL50
−0.0848204
−0.0045535
−0.0568274
 0.00383848
−0.052247


CIL51
−0.0173703
 0.0030163
 0.03450537
−0.06742194
−0.0442069


CIL52
 0.09321461
−0.0061184
 0.03887959
 0.02195686
 0.28402574


CIL55
 0.02771794
−0.0027585
 0.09849299
 0.02873006
 0.07377767


CIL56
 0.02644617
 0.00049055
 0.23684095
 0.03544769
 0.19862485


CIL58
 0.02238111
−0.0529865
 0.09830113
 0.07532329
 0.15046424


CIL60
 0.05735354
−0.0333761
 0.08176375
 0.13594441
−0.0670478


CIL62
 0.04148582
−0.0555592
 0.19290773
 0.06831212
−0.0317476


CIL63
 0.02351722
 0.00271866
 0.02815163
 0.02881969
−0.0437018


CIL64
 0.14219284
 0.0019661
 0.05532962
 0.04261861
 0.0650817


CIL66
−0.0264041
−0.0255773
 0.0369439
 0.04468669
−0.0793466


CIL67
−0.0727219
−0.0160091
−0.0201768
 0.08702793
 0.0614094


CIL69
 0.02202981
−0.0213286
 0.17205944
 0.05309819
 0.19627093


CIL70
 0.03107613
−0.0085471
 0.12929417
 0.07714716
 0.0630101


CIL71
−0.036561
−0.000118
−0.0295579
−0.01111712
 0.01513793


CIL72
−0.0366009
 0.01422321
−0.024636
 0.01988225
 0.03640616


CIL74
−0.0590576
 0.01058899
−0.024894
 0.08294342
−0.0294162


CIL75
−0.0164272
−0.0144916
 0.18828932
 0.07275235
 0.23105427


CIL76
 0.00489521
−0.0193662
−0.0283021
 0.02129585
−0.0086886


CIL77
−0.0567652
−0.0277058
−0.0654344
 0.07337511
−0.0556501


CIL79
−0.0063695
−0.0085941
 0.22575501
 0.04062847
 0.19843153


CIL80
−0.0085154
−3.885E−05
−0.0159272
 0.03142114
−0.0060943


CIL87
−0.0329284
−0.0073839
−0.0481974
−0.0328253
 0.00320572


CIL88
−0.0355936
−0.0121722
−0.0364896
−0.06126086
 0.02089869


CIL89
−0.0393202
−0.0234351
−0.0295688
−0.00629215
−0.066762






BJ.atoc
BJ.trolox
BJ.zVAD
HT.3MA
HT.ALLN





MS275
−0.0084515
−0.0011371
 0.14146731
−0.0920374
−0.1325215


Scriptaid
−0.0053962
−0.0185016
 0.12050225
−0.1243573
−0.1402561


TrichostatinA
−0.0180623
−0.0267223
 0.11977296
−0.1241009
−0.1481335


Echinomycin
 0.16805955
−0.043222
 0.00597268
 0.30826032
 0.08399808


Colchicine
−0.02812
−0.0042293
 0.0570233
 0.01860588
 0.19321494


NPC25
−0.0050705
 0.00975904
 0.07171837
 0.08924394
 0.17592665


NPC4
 0.00195392
 0.01365968
 0.04442037
 0.06849702
 0.17149162


NPC7
 0.01389367
 0.02619418
 0.05045047
 0.04871941
 0.00130774


Podophyllotoxin
 0.00707233
−0.012817
 0.04960196
 0.0013506
 0.15724862


Rotenone
 0.05115769
−0.0107482
 0.04408484
 0.06288513
 0.07973252


Vinbiastine
 0.14869923
−0.0114541
 0.01551736
 0.05248911
 0.15265143


Vincristine
 0.04717344
−0.0330489
 0.01856344
 0.02990414
 0.18689078


DPI3
 0.43896545
 0.43427174
−0.0465249
−0.007409
−0.1136639


DPI4
 0.26106654
 0.25470923
−0.0217944
−0.0014583
−0.1647522


DPI6
 0.31847172
 0.32883594
−0.0118015
 0.00819515
−0.1107117


RSL3
 0.36011958
 0.37310011
 0.00586004
−0.0747144
−0.0380195


Bortezomib
 0.01647258
 0.01237058
 0.00508615
−0.0717141
−0.085871


MG132
−0.0023264
−0.0144852
−0.0845176
−0.0339243
−0.21489


MG262
 0.05198126
 0.00808713
−0.0549816
−0.0656271
−0.1355736


Erastin
 0.32756245
 0.24919798
−0.0053013
−0.0457811
−0.068557


Camptothecin
 0.01158062
 0.04873997
 0.08240716
 0.01920351
 0.03354113


Irinotecan
 0.03987423
0.07779279
 0.07984183
 0.00553604
−0.0552172


Daunorubicin
 0.00414462
−0.0202568
 0.05593995
 0.02823215
 0.00869204


Doxorubicin
 0.01689052
 0.01163726
 0.05543158
 0.03088374
 0.03102276


Etoposide
−0.0103765
−0.0180785
 0.06084183
 0.00720806
 0.03276835


Mitoxantrone
 0.02585399
−0.0186233
 0.04970382
 0.02955197
 0.0735492


Cycloheximide
 0.00016722
 0.01855954
 0.02186279
 0.00551244
 0.3818123


Dinitrophenol
 0.02317705
 0.00995695
−0.0001198
−0.1332127
 0.11473914


NaN3
−0.005135
−0.0383723
−0.0010651
−0.0571935
 0.06335229


Vatinomycin
 0.28498
 0.06867276
 0.03769032
−0.1652966
 0.04228034


CIL1
 0.05385054
 0.08301673
−0.0470499
−0.0019681
 0.10528473


CIL2
−0.0114945
 0.00165699
−0.0039592
 0.07177774
 0.0529217


CIL4
 0.00430676
 0.0084059
 0.01241555
 0.02219194
 0.02482089


CIL5
−0.0027101
 0.01829519
−0.0205413
 0.01320494
 0.07446486


CIL6
 0.01482359
−0.0067769
−0.0031922
 0.06095929
 0.04648241


CIL7
 0.00084946
 0.04239953
−0.0583405
−0.0046258
 0.11359317


CIL9
 0.02754398
 0.05396583
−0.0270786
 0.04492152
−0.0207669


CIL10
 0.04670042
 0.05526964
−0.0073328
−0.0198166
 0.10670073


CIL11
−0.020819
−0.0178186
−0.0045166
 0.00011437
 0.01794211


CIL13
−0.0056464
−0.0066491
−0.0045682
 0.02534783
 0.0164632


CIL15
−0.0114947
−0.0102551
 0.01242501
 0.02629949
 0.00012453


CIL16
−0.0335072
 0.00368936
−0.0061661
 0.02993016
−0.032855


CIL17
 0.00645085
−0.001174
−0.0231259
 0.01558063
 0.03631301


CIL18
−0.0004228
 0.03949592
−0.0054018
 0.01461487
 0.07303251


CIL20
−0.0051132
 0.02308064
−0.0403597
 0.00488149
 0.09673045


CIL22
 0.0101546
 0.0183972
 0.03093116
−0.0160623
 0.02848605


CIL23
 0.00231459
−0.0215153
 0.0469053
 0.01608989
 0.00152136


CIL25
 0.02909253
−0.0010292
 0.03879171
 0.01604736
−0.0257036


CIL26
 0.05008099
 0.0314732
−0.0032595
 0.03209618
 0.00159167


CIL27
 0.00240378
 0.00049489
−0.0035753
 0.07660012
 0.16362907


CIL28
 0.05294996
 0.06039075
 0.00623431
 0.09370094
 0.04718566


CIL33
 0.04139859
−0.0533898
−0.0267524
 0.08789041
 0.04123474


CIL34
 0.0578079
 0.00702937
 0.00599705
 0.1254804
 0.15508365


CIL36
 0.06821973
−0.0232232
 0.0096601
 0.08326909
 0.14219062


CIL40
 0.0502196
 0.03943469
 0.00797747
 0.08070401
 0.17614854


CIL41
 0.27286743
 0.18714539
 0.03607308
 0.04520507
 0.09324127


CIL44
 0.00911865
−0.0398862
−0.0168219
 0.07356209
 0.13923637


CIL46
 0.10512996
 0.04647304
−0.0515263
 0.08623972
 0.03766758


CIL47
 0.03518558
−0.028058
−0.0149664
 0.07293753
 0.22270776


CIL48
 0.01164047
−0.0340715
−0.0180689
 0.08539756
 0.16107782


CIL49
−0.0083242
−0.0233678
−0.0017621
 0.0543506
 0.06619736


CIL50
 0.00564663
 0.00486512
 0.02532048
−0.0235396
 0.18885435


CIL51
 0.02105244
 0.05843873
 0.03693029
−0.1102537
−0.0787411


CIL52
 0.08208107
 0.01188166
 0.02643387
−0.0155768
 0.2564788


CIL55
 0.18175017
 0.19779608
−0.0135613
 0.00703667
 0.02517231


CIL56
 0.29616775
 0.2616025
 0.03467497
−0.033086
−0.1793586


CIL58
 0.01276141
 0.07255912
 0.00460643
 0.01140406
 0.11609447


CIL60
 0.03955105
 0.04379633
−0.0011813
 0.02575321
−0.0158789


CIL62
 0.02712651
 0.03186254
 0.03040708
−0.0090173
 0.17019511


CIL63
 0.01699604
 0.02098909
 0.04472039
 0.00891738
 0.07446255


CIL64
 0.02331446
 0.03467968
 0.00817202
 0.08632164
 0.00995343


CIL66
 0.01039015
 0.03146974
 0.00222834
 0.03556215
−0.0328894


CIL67
−0.0150944
−0.0044336
 0.01614338
 0.01770618
 0.12391603


CIL69
 0.26783116
 0.3133252
 0.04233348
 0.01872961
−0.1005245


CIL70
 0.30465448
 0.191108
 0.0332095
−0.0366043
−0.1854615


CIL71
 0.01578726
 0.00497539
 0.06648208
 0.00307792
−0.0126326


CIL72
−0.0141221
 0.00226961
 0.08776565
 0.01189674
 0.0857353


CIL74
−0.0263184
−0.0174426
 0.01184939
−0.0832325
 0.12935179


CIL75
 0.36061903
 0.37638163
 0.01441857
−0.0336856
−0.0704404


CIL76
−0.0202205
−0.0290381
 0.05031848
−0.0137338
 0.08674041


CIL77
−0.0058929
−0.0185567
 0.06873383
−0.0134697
 0.111967


CIL79
 0.47068744
 0.50412672
 0.06601613
−0.0171877
−0.0838722


CIL80
−0.0062044
−0.0006860
 0.07245022
 0.03564047
 0.10205804


CIL87
 0.00201163
−0.0357952
 0.07239753
−0.0233802
 0.13886251


CIL88
 0.02299092
−0.0325654
 0.03654718
−0.0294199
 0.13385996


CIL89
 0.02137483
 0.0079497
 0.01685657
−0.054211
 0.02598681






HT.ATA
HT.ActD
HT.BHA
HT.BHT
HT.BocD





MS275
0.02120396
−0.1981516
−0.048682
−0.1794314
0.02448207


Scriptaid
−0.0387325
−0.1631887
−0.0588253
−0.1030388
0.02827963


TrichostatinA
−0.0004817
−0.1678342
−0.0695579
−0.1351852
0.04109121


Echinomycin
0.04087497
0.06603729
0.02424152
−0.0407598
0.07384558


Colchicine
0.13248515
0.01342724
0.01506112
−0.0067467
0.12639405


NPC25
0.02995861
0.09537898
0.03736291
0.08587469
0.16302178


NPC4
0.03000496
0.07284775
0.04586726
−0.0223846
0.08053185


NPC7
0.01607661
0.05634308
0.0520777
0.00336857
0.10095325


Podophyllotoxin
0.12060383
−0.0009195
0.02739229
0.00085858
0.09840235


Rotenone
0.05899383
0.10621410
−0.0218389
0.02466811
0.05103128


Vinbiastine
0.09963106
0.05584248
−0.0165122
0.01379476
0.12562051


Vincristine
0.13037176
0.05976018
−0.0047543
−0.0156826
0.14006758


DPI3
−0.0428149
−0.0002473
0.40702448
0.34683128
−0.0039141


DPI4
−0.0362229
−0.0068307
0.28495694
0.24859229
−0.0140724


DPI6
−0.0419376
−0.0416732
0.35257524
0.28208053
−0.0011769


RSL3
−0.0453945
−0.0857906
0.37982898
0.24437145
−0.0118449


Bortezomib
0.05409902
−0.0722867
−0.0498515
−0.1309809
0.02873805


MG132
0.00576757
−0.0641667
−0.1238156
−0.1271213
0.00205102


MG262
0.03320394
−0.0585361
−0.0764988
−0.1219041
0.00758177


Erastin
−0.1314893
−0.1110882
0.17477284
0.12434413
−0.0033408


Camptothecin
−0.00483
−0.1286722
0.04861159
−0.0841072
0.03455369


Irinotecan
0.00598208
−0.1215199
0.0146573
−0.0826371
0.04301941


Daunorubicin
−0.0018602
0.01441852
0.01070136
−0.0560879
0.05219776


Doxorubicin
0.02840528
0.00799312
0.02167254
−0.0681508
0.05959192


Etoposide
−0.0363022
−0.0994447
0.04757315
−0.0005481
0.05886376


Mitoxantrone
0.02421666
−0.0207278
0.02090636
−0.0249046
0.06801715


Cycloheximide
−0.0105671
0.06056867
0.06705416
−0.0329721
0.04129835


Dinitrophenol
−0.0389289
0.11863548
−0.1811047
−0.1843367
0.05040775


NaN3
0.03107002
0.11974286
−0.0406068
−0.1432331
0.00899593


Vatinomycin
0.04038739
0.0778977
−0.0126288
−0.0523299
0.01495644


CIL1
−0.0478044
−0.0405747
0.02690843
−0.0849008
−0.0164407


CIL2
0.03084107
−0.0052281
−0.0376678
−0.1327033
0.13949477


CIL4
0.0280594
−0.0014633
−0.0093098
−0.0221211
0.12719391


CIL5
0.00058927
−0.0075302
−0.0850186
−0.0991686
0.01524426


CIL6
0.00765427
−0.0089279
−0.0076359
−0.0611365
0.07600404


CIL7
−0.0167092
−0.0119692
−0.0635192
−0.103029
0.07799454


CIL9
−0.0031435
−0.0639955
−0.0447691
−0.1803047
−0.0237571


CIL10
0.01742637
−0.0186466
−0.0286273
−0.086629
0.13845853


CIL11
0.01479608
−0.0342959
−0.0190179
−0.0964126
0.07910764


CIL13
0.04671792
−0.0432671
−0.0628773
−0.1040152
0.09941468


CIL15
−0.0028092
0.01241754
−0.0399345
−0.1488868
0.10209544


CIL16
0.00396522
0.01875074
−0.0593716
−0.1721118
0.14140992


CIL17
−0.0157027
−0.0398214
−0.0205226
−0.1380266
0.03130279


CIL18
0.00141458
0.01840477
−0.0870338
−0.1394373
0.0660702


CIL20
−0.0012756
−0.0066082
0.00017152
−0.080748
0.01009418


CIL22
0.05907837
−0.0240194
0.00192474
−0.0523839
0.00862815


CIL23
0.01792035
0.0196907
0.00701969
−0.0276208
−0.0125905


CIL25
0.06488994
−0.035853
−0.0154723
−0.1041257
0.05182572


CIL26
0.04191602
−0.0024876
−0.1441072
−0.121522
0.00899186


CIL27
0.04844866
0.03171132
−0.0155971
−0.0553673
0.0032882


CIL28
0.06113466
−0.0135206
−0.1362427
−0.1108463
0.01461547


CIL33
0.04655447
−0.0326933
−0.0293756
−0.1089134
0.00630687


CIL34
0.05661395
−0.0322385
0.01628473
−0.1101646
−0.0074755


CIL36
0.05273856
−0.0440921
0.00539978
−0.1272189
0.00216899


CIL40
0.06692048
−0.0654416
−0.0937176
−0.150511
0.01176086


CIL41
0.02642687
−0.0934252
0.13123971
0.00933821
−0.0167806


CIL44
0.0592013
0.01649407
−0.0255872
−0.1006627
−0.0552012


CIL46
0.01619574
−0.0291586
−0.1014536
−0.1471088
−0.0437101


CIL47
0.07043108
0.01587837
−0.0447825
−0.1469266
−0.0096169


CIL48
0.04683652
0.01672523
−0.0696577
−0.1546714
−0.0382283


CIL49
0.04767921
−0.0187376
−0.0143278
−0.0829684
−0.0224007


CIL50
0.06517532
−0.0077502
0.00665487
−0.06029
0.00401667


CIL51
0.00357883
0.02697499
−0.0796469
−0.0848758
−0.0145762


CIL52
0.03416515
0.07378954
−0.0130181
−0.0671898
0.00858206


CIL55
0.05194555
−0.0078999
0.0959274
0.0491901
0.0156815


CIL56
−0.0193853
−0.1240014
0.1773734
0.01460369
−0.0127039


CIL58
0.05141171
0.02044398
−0.0398906
−0.1354996
−0.020815


CIL60
0.07051541
0.04832397
−0.0006407
−0.1303369
−0.0405115


CIL62
0.0294142
−0.0114286
−0.0073935
−0.1437226
0.0024554


CIL63
0.07925366
−0.0032543
0.01783848
−0.1088494
0.01854753


CIL64
0.1978537
0.04801805
−0.1468952
−0.3128485
−0.107814


CIL66
0.02652209
−0.0327722
−0.0208457
−0.154242
−0.0578802


CIL67
0.08909056
0.06498217
−0.0355783
−0.1041323
−0.0237794


CIL69
−0.0053581
−0.0042653
0.24891501
0.22271496
−0.0345747


CIL70
−0.0043783
−0.0291397
0.10618587
−0.0354134
−0.030043


CIL71
0.0348115
0.00070178
−0.0562239
−0.0583947
0.01088871


CIL72
0.0553314
0.02923746
−0.0401283
−0.1001862
−0.0241548


CIL74
0.01362072
0.02636443
−0.0865918
−0.0457196
0.01242029


CIL75
−0.0171453
0.01891564
0.31310244
0.28280708
−0.0121888


CIL76
0.04513457
0.04285579
0.00057803
−0.0865509
−0.027674


CIL77
0.05491925
0.09589947
−0.0087136
−0.079983
−0.0282833


CIL79
−0.0530223
−0.0402145
0.39088563
0.36443077
−0.0023848


CIL80
0.05780885
0.08002129
0.00945222
−0.0197054
−0.0233656


CIL87
0.11366272
−0.0075862
−0.0033579
−0.0831667
−0.0071337


CIL88
0.08121765
−0.0310215
0.00710891
−0.0998787
0.02036709


CIL89
0.05896078
0.03977145
−0.0061467
−0.1187912
−0.0046673






HT.CHX
HT.Co2+
HT.CspA
HT.DPQ
HT.EGTA





MS275
−0.1188969
−0.102694
−0.1885541
−0.0105775
−0.0058107


Scriptaid
−0.1048316
−0.193896
−0.1413332
−0.00813
−0.0176747


TrichostatinA
−0.096958
−0.0277054
−0.1436955
−0.0015533
−0.0171128


Echinomycin
0.14361174
0.1491729
−0.084261
0.00691846
0.00691633


Colchicine
0.04333171
0.10984848
−0.1208652
0.01511414
−0.000119


NPC25
0.1519485
0.13013485
−0.0243371
0.01957461
0.03077107


NPC4
0.12317457
0.06041622
−0.0446754
−0.0070462
0.00516068


NPC7
0.13739696
0.01909726
−0.0268684
0.01267774
0.03356135


Podophyllotoxin
0.07196985
0.11023663
−0.0759222
0.00596443
0.01145102


Rotenone
0.07793169
0.20027
−0.1721788
−0.0054033
0.00225841


Vinbiastine
0.06308695
0.22685265
−0.1334704
0.00368754
0.0105502


Vincristine
0.08269379
0.21695164
−0.1876483
0.01267252
0.00923087


DPI3
0.03770847
0.23127886
−0.0304777
0.04997748
0.04922409


DPI4
−0.0086992
0.17414425
5.8801E−05
0.05528113
0.0361033


DPI6
0.07054851
0.16240191
−0.0172826
0.06163326
0.04864836


RSL3
−0.0509933
−0.1947562
−0.1267995
0.16532291
0.03695606


Bortezomib
0.02630913
0.06444914
−0.2224122
0.01078526
−0.006629


MG132
0.04853848
−0.0570457
−0.3220782
−0.0240645
0.00777542


MG262
−0.0039772
−0.0442528
−0.2144673
0.00810399
−0.0011524


Erastin
0.05504404
−0.2142077
0.02448894
0.05672269
0.01444339


Camptothecin
0.05619607
0.14156517
−0.1551597
0.00814331
0.01652399


Irinotecan
0.0667595
0.02045732
−0.1317706
0.00907953
0.01365217


Daunorubicin
0.05854213
0.16142753
−0.079195
0.0088822
0.03262034


Doxorubicin
0.08993251
0.26270189
−0.1098208
0.01106636
0.03492068


Etoposide
0.11300892
0.39257154
−0.1769875
0.00642178
0.02456941


Mitoxantrone
0.04002351
0.38167687
−0.112673
0.00650421
0.01801996


Cycloheximide
0.14620902
0.09947986
0.04639214
0.01424647
0.0138591


Dinitrophenol
0.20975707
0.0433563
−0.1730059
0.0815659
−0.0959985


NaN3
0.04722344
0.03940525
−0.0477731
−0.0079698
−0.002548


Vatinomycin
0.10235561
−0.0618283
−0.0378938
0.17016661
−0.0448288


CIL1
0.09163445
0.20478977
−0.0454163
0.04202848
−0.0066024


CIL2
0.1414035
−0.1320389
0.01223619
−0.0031255
−0.0056273


CIL4
0.10959238
−0.1439922
−0.0469582
0.00735917
0.01533189


CIL5
0.0972613
0.08697891
−0.0215948
0.00964073
−0.0092153


CIL6
0.16102241
0.04207798
−0.0127452
0.04323431
0.01026344


CIL7
0.05109611
0.10624303
−0.0644051
0.00525397
0.00645645


CIL9
0.07226888
0.0175425
−0.0307928
−0.0033167
−0.0068483


CIL10
0.05131796
0.12255069
0.00487182
0.0043051
−0.0077762


CIL11
0.00705209
0.05357729
−0.0054982
0.00390362
0.0040021


CIL13
0.03998706
0.29237304
0.06242739
−0.0151495
0.08353385


CIL15
0.06342084
0.13972534
−0.0070316
−0.0239385
−0.0006158


CIL16
0.08542601
0.1550268
0.00838857
−0.022048
−0.0002723


CIL17
0.06579474
−0.0084365
−0.0335367
−0.0230258
0.0018567


CIL18
0.11789261
0.15978519
−0.0482687
−0.0127573
−0.0094606


CIL20
0.12124292
0.12049185
−0.0564837
−0.0377921
−0.018046


CIL22
0.01937859
−0.0347775
−0.0361099
−0.0093401
−0.0125444


CIL23
0.08773004
0.0331646
−0.0232587
0.01052194
−0.0132254


CIL25
−0.0160441
−0.0612815
0.01072928
0.02628733
0.01623521


CIL26
−0.0205236
−0.0274445
−0.0422466
0.0298704
0.00999786


CIL27
0.04575936
0.046063
−0.0363878
0.02306718
0.02603826


CIL28
0.03280038
−0.0329175
−0.0185839
0.04403759
0.00586771


CIL33
0.06763145
0.14175785
−0.0321298
0.02727875
−0.0074608


CIL34
0.04610462
0.09925752
−0.0218141
0.03329763
−0.0020351


CIL36
−0.0664531
0.02820654
0.01593074
0.05193466
−0.0131102


CIL40
−0.0286309
0.04910502
−0.0469721
0.04908281
−0.0264168


CIL41
−0.0545941
0.06388608
9.4409E−05
0.0428151
0.0294626


CIL44
0.02873779
−0.0465752
−0.0158637
−0.0166894
−0.0072668


CIL46
−0.0190914
−0.1082153
0.00033561
−0.0016923
−0.0169929


CIL47
0.00787398
−0.2380995
0.01760547
−0.0203835
0.00105146


CIL48
0.01479799
0.03070109
0.00268882
−0.0003728
0.01715062


CIL49
−0.0094838
0.00880832
0.04470562
−0.0241787
0.02251689


CIL50
−0.0482491
−0.0868821
−0.0127092
−0.0158756
−0.0224416


CIL51
0.08663358
0.0390822
0.00675035
0.01090346
0.04897337


CIL52
0.14735761
0.41547366
0.05641151
−0.0133024
0.09077934


CIL55
−0.0103853
0.11519832
−0.0078827
0.0140465
0.04876502


CIL56
−0.0723393
0.01295837
0.01564299
0.0624478
0.023781


CIL58
−0.0567432
0.09509314
0.06761309
−0.0059223
0.05183692


CIL60
0.11203194
−0.0800334
−0.0048312
0.02072829
0.05295873


CIL62
0.08707299
0.03422233
−0.0013468
0.01016954
0.04610858


CIL63
0.07744277
0.02639089
0.00306287
−0.0217193
0.06222147


CIL64
0.06684839
0.41607903
0.08661477
−0.1350243
0.18424717


CIL66
0.08144049
−0.0603373
−0.0546913
−0.0110529
0.03093079


CIL67
0.14582237
0.03257119
0.01638443
−0.0109218
0.12341795


CIL69
0.03395238
0.07131741
0.00984813
−0.0065882
0.06031548


CIL70
−0.0263072
0.019784
−0.0467956
0.0269571
0.01851476


CIL71
0.07610494
0.02342185
−0.032331
−0.0035051
−0.0229848


CIL72
0.11473513
0.11632921
−0.0191176
−0.0315719
0.00960798


CIL74
0.02524432
0.03243519
−0.0046218
0.04846659
−0.0267434


CIL75
0.04020367
0.12168756
−0.0096738
0.01971005
−0.0070184


CIL76
0.15052401
0.15314755
−0.0059982
0.01553764
−0.0215115


CIL77
0.1436028
0.12610682
−0.0579089
0.0034956
−0.0034501


CIL79
0.02793995
0.29484557
−0.0338926
0.04763562
0.02262115


CIL80
0.09740519
0.11202522
0.01161471
−0.0171717
0.01990089


CIL87
0.06730386
0.03163242
−0.0535776
−0.0256244
0.00530494


CIL88
0.04477773
0.01651643
−0.0584225
−0.0143108
0.01878024


CIL89
−0.0086929
0.00837676
−0.0690578
−0.0052866
0.04226482






HT.Gd3+
HT.LNAME
HT.Lmim
HT.NAD+
HT.NMMA





MS275
−0.0102689
0.00420427
−0.0972784
0.00087937
−0.0033302


Scriptaid
−0.0217556
−0.0026369
−0.0558337
0.01064252
0.00886933


TrichostatinA
−0.0033979
0.0035016
−0.0502135
0.03432383
0.00433465


Echinomycin
0.01526225
0.01177721
0.06784009
0.17795514
−0.0009733


Colchicine
0.01351349
0.00823575
0.11991903
0.1471024
0.00858507


NPC25
0.00874193
0.01017305
0.22821299
0.16858337
0.01131088


NPC4
0.01314254
0.00258833
0.20365836
0.11357064
0.01570669


NPC7
0.02479755
0.00219407
0.12362177
0.09896023
−0.0072181


Podophyllotoxin
0.00915994
−0.0049258
0.10975693
0.13182264
0.00047202


Rotenone
0.01050591
−0.0073773
0.14131642
0.15629326
0.00783177


Vinbiastine
−0.0025566
−0.0123136
0.12599319
0.16821378
−0.0035162


Vincristine
−0.0009641
0.00177863
0.11545731
0.1698731
0.00483074


DPI3
−0.009591
−0.0039604
0.39279429
0.00238937
−0.0015082


DPI4
−0.0212108
0.01547048
0.30856618
0.02440851
−0.0007723


DPI6
−0.0227614
−0.0043042
0.38620976
0.00907448
−0.0110443


RSL3
−0.012653
−0.0108087
0.33020083
0.00788309
−0.0055098


Bortezomib
−0.028227
0.00992421
0.01347348
0.13902598
0.01353813


MG132
−0.0110019
5.1006E−05
0.00133838
0.03798455
0.01063618


MG262
−0.0031676
0.00700243
0.00145721
0.07678843
−0.0042292


Erastin
−0.0132633
−0.0087309
0.21777043
−0.0299568
0.00682629


Camptothecin
0.02887279
0.01135258
0.07220067
0.060785
0.01978231


Irinotecan
0.01373612
0.01215253
0.05380647
0.03220192
0.00973208


Daunorubicin
0.01529138
−0.0098523
0.06111651
0.13051521
0.01829966


Doxorubicin
0.03740828
0.00668466
0.08822325
0.18719506
0.02832305


Etoposide
0.02667925
0.00544411
0.05001251
0.05270336
0.01602895


Mitoxantrone
0.03226623
0.01297692
0.05677663
0.1502147
0.02360459


Cycloheximide
0.00299079
0.02188041
0.10894658
0.07007128
0.0198614


Dinitrophenol
−0.0282895
−0.0088594
0.02524142
0.11635416
0.01660839


NaN3
−0.0046043
−0.0135112
−0.0761287
0.06682519
0.00282199


Vatinomycin
−0.0045237
−0.0384324
0.1162672
0.07832788
−0.002681


CIL1
−0.0321493
0.03977103
0.08631064
0.07889481
−0.0481967


CIL2
−0.0230384
0.02625109
−0.0301281
0.04925344
−0.0302901


CIL4
−0.0099527
0.01005304
−0.0192819
−0.0138124
−0.007501


CIL5
−0.0256984
0.0226272
0.00357399
0.03421081
−0.003405


CIL6
−0.0289684
0.01570488
−0.0106374
0.04899418
−0.0229193


CIL7
−0.029815
0.01998017
−0.0026743
0.07707444
−0.024423


CIL9
−0.0435849
0.02123914
−0.0546891
0.02617067
−0.0373288


CIL10
−0.0278814
0.02616127
−0.020647
0.0408689
−0.009286


CIL11
−0.0298047
0.02953801
−0.0340013
0.02581914
−0.0255366


CIL13
−0.030717
−0.0796178
−0.061016
−0.0446201
−0.0411831


CIL15
−0.0388128
0.00130904
−0.0483689
0.01762367
−0.0194345


CIL16
−0.0407027
0.01973692
−0.0554627
0.05126088
−0.0315355


CIL17
−0.0431802
0.02242474
−0.0132
0.04423236
−0.0263893


CIL18
−0.0330227
0.0132922
−0.0049868
0.09492877
−0.0291809


CIL20
−0.02607
0.01702871
0.00291412
0.05984741
−0.0208969


CIL22
−0.0045341
0.04607751
0.01658507
0.01013702
0.01444063


CIL23
−0.000221
0.00562841
0.01608214
−0.0002222
−0.0010838


CIL25
0.01712542
0.02662905
0.01684027
−0.0305125
−0.0066394


CIL26
−0.0053343
0.00121517
−0.0040003
−0.0140608
−0.0096924


CIL27
−0.016795
0.00120376
−0.0010549
−0.0033996
−0.0133115


CIL28
−0.0258257
−0.0045004
0.00131087
0.0070887
−0.0097629


CIL33
−0.0218244
0.00304734
−0.0070678
−0.018901
−0.0022478


CIL34
−0.0228317
−0.0051696
−0.002139
−0.0055345
−0.0030313


CIL36
−0.042672
−0.0279853
0.00421383
−0.0637848
−0.0095011


CIL40
−0.0185952
−0.0199914
0.00048251
−0.0474936
−0.0069008


CIL41
−0.0259176
−0.0121037
0.13770074
−0.0086566
−0.0102875


CIL44
−0.0114691
−0.0250548
−0.0076141
−0.0714428
−0.0419036


CIL46
−0.0086171
−0.0280471
−0.0316559
−0.0675446
0.0015569


CIL47
−0.0226292
−0.0128778
−0.0417274
−0.0278389
−0.0119475


CIL48
−0.003556
−0.0408723
−0.0175732
−0.0546542
−0.0190249


CIL49
0.00090697
0.01289374
−0.0368471
−0.0141297
0.00042903


CIL50
−0.0387716
0.01805957
−0.0053598
0.00979941
0.00419608


CIL51
0.00328775
0.0109103
0.01293031
0.0439374
0.01561505


CIL52
−0.006922
−0.0094118
0.02653731
0.01944592
−0.011955


CIL55
−0.0147883
0.00717105
0.07004625
0.03996362
0.00476077


CIL56
−0.0154241
0.05374763
0.10168238
0.0231475
−0.0147499


CIL58
−0.0046743
−0.0998179
−0.0725714
−0.0653865
−0.0908298


CIL60
−0.016299
0.00439718
0.0346384
0.01525644
−0.0109061


CIL62
−0.0257331
−0.0035685
0.00805599
−0.016889
−0.0175391


CIL63
−0.0316366
0.04201046
0.03961757
0.03315753
−0.0022977


CIL64
−0.0263028
<0.2159453
−0.2704681
−0.1970983
−0.1741564


CIL66
−0.0175703
0.00403506
0.01651998
−0.0110611
−0.014205


CIL67
−0.0194487
−0.0075145
0.01911998
−0.0179485
−0.0134891


CIL69
−0.0144554
−0.045383
0.17920333
0.03637312
−0.0404809


CIL70
−0.0135663
0.00599279
0.21979664
0.04321272
−0.0058179


CIL71
−0.0155194
0.02366266
−0.0026093
0.01545888
−0.0007859


CIL72
−9.532E−05
0.01225977
−0.0018208
0.00419487
−0.0044427


CIL74
−0.008213
0.0192517
0.02034016
0.07663993
9.3282E−05


CIL75
−0.022849
−0.0173727
0.24992353
−0.0025939
−0.0279395


CIL76
−0.0292987
0.02220486
0.00796408
0.00780997
−0.0079421


CIL77
−0.0367999
0.01693235
0.02572953
0.00335673
−0.0150852


CIL79
−0.0257937
0.01224348
0.40517143
−0.0042922
0.00319361


CIL80
−0.0322647
0.01972379
0.05448637
0.01251178
−0.0125563


CIL87
−0.0275554
0.00525664
−0.0021155
−0.0019259
−0.012374


CIL88
−0.036514
0.02198995
0.01907149
0.00747768
−0.000615


CIL89
−0.0459432
0.00306109
0.02342444
0.02661864
−0.0090411






HT.Nec-1
HT.Pepstatin
HT.SP600125
HT.TLCK
HT.U0126





MS275
−0.0555719
−0.0165421
−0.0990255
−0.0240373
−0.0188407


Scriptaid
−0.0561729
−0.0129585
−0.0700007
−0.0557541
−0.0422842


TrichostatinA
−0.0728913
−0.0039853
−0.0686523
−0.0259878
−0.0646838


Echinomycin
0.02092974
0.00686125
0.08228039
0.0064356
0.01676216


Colchicine
0.01660958
0.00441257
−0.0285789
0.03060033
0.22370553


NPC25
0.03331695
0.00907117
0.04752536
0.01191743
0.14555698


NPC4
0.0515934
−0.0097394
0.0594831
−0.0170816
0.10968643


NPC7
0.02936184
0.0140623
0.05675053
−0.0171298
0.03515321


Podophyllotoxin
−0.0001825
−0.0005129
−0.0276787
0.06977949
0.13541903


Rotenone
0.00392091
−0.0225529
−0.019636
0.0438025
0.13841211


Vinbiastine
−0.0058737
−0.0004474
0.0028618
0.00236221
0.13717582


Vincristine
0.02187585
0.00026198
0.00089979
−0.0063942
0.19262685


DPI3
0.06021328
0.02311469
−0.058705
0.30781927
0.43976324


DPI4
0.05134598
−0.003295
−0.0428398
0.17609432
0.34227684


DPI6
0.05782935
0.0120804
−0.0586102
0.2543766
0.38311656


RSL3
0.07095265
−0.0132658
−0.1318706
0.31517178
0.39718399


Bortezomib
−0.0618561
−8.033E−05
−0.0302754
0.0719481
−0.0318738


MG132
−0.0972845
−0.0273265
−0.0343148
0.01546369
−0.0709958


MG262
−0.0681715
−0.0060693
−0.0359956
0.03565513
−0.0666432


Erastin
−0.0255149
−0.0257345
−0.0368797
0.11665403
0.15248389


Camptothecin
0.01101588
0.00591775
0.02319051
0.0438257
0.00027174


Irinotecan
0.0216686
0.0147477
0.00621888
0.02566275
0.00698441


Daunorubicin
0.02173842
0.0056488
−0.0113019
0.06428773
−0.0158987


Doxorubicin
0.00757748
0.00895778
−0.0214841
0.06329923
−0.0203643


Etoposide
−0.0059495
−0.001737
0.00478042
0.1489187
−0.0262697


Mitoxantrone
0.01072807
0.0206681
−0.0157506
0.06003033
−0.0320525


Cycloheximide
0.03735921
0.0061635
−0.0033266
0.01836327
0.03847567


Dinitrophenol
0.03763391
0.01716386
−0.1203227
0.03576826
−0.0215633


NaN3
−0.0296822
−0.0043368
−0.0040791
0.02260372
0.02567325


Vatinomycin
0.08393792
0.01970083
−0.1610157
0.02961013
0.00548191


CIL1
0.02294445
0.00147358
−0.0491857
0.04647174
0.14912375


CIL2
−0.0127254
0.00881644
−0.0036839
0.02036906
0.02089377


CIL4
0.00249863
0.00928023
−0.006297
0.01720083
0.00056242


CIL5
0.00335096
0.01964795
−0.0214887
−0.0040572
−0.0294352


CIL6
0.01299035
0.01879984
−0.0119461
−0.0054596
0.0484597


CIL7
0.00713771
−0.00124
−0.0415509
−0.0111335
0.0155565


CIL9
0.02132694
−0.0021878
0.01016425
0.00551409
0.03914784


CIL10
0.03697698
−0.0108718
−0.0092266
−0.0016305
0.01126286


CIL11
0.01983171
−0.0030957
−0.028758
0.01902218
0.06089128


CIL13
0.04319318
−0.0304073
−0.0797293
0.01350303
0.0592947


CIL15
0.02067279
−0.0224727
−0.0239724
−0.0173117
0.08141342


CIL16
0.00654015
−0.0234635
−0.0219493
−0.0286311
0.05201472


CIL17
0.03321398
−0.0210351
−0.0131421
−0.0026391
0.04847215


CIL18
0.01177279
−0.032163
−0.0346469
−0.030163
0.01191696


CIL20
−0.0008504
0.00016309
−0.025792
−0.0033667
0.02994872


CIL22
−0.0022834
0.00057166
−0.0017349
0.02243652
0.06274399


CIL23
−0.0091711
−0.0054941
0.00961612
0.01135686
0.02324317


CIL25
−0.0148096
0.0109388
0.02383204
0.02361905
−0.0319668


CIL26
−0.0120433
0.00129487
−0.0105174
−0.006743
−0.0264817


CIL27
−0.0027517
0.01174891
0.0163087
−0.006607
0.02757687


CIL28
−0.0302207
0.00126773
0.01064584
0.00513542
−0.0399002


CIL33
−0.0021233
0.0082125
0.00235928
0.01726866
0.01835922


CIL34
0.00686593
0.00474178
−0.0184715
0.01693744
0.04868298


CIL36
−0.011675
0.00332247
−0.0200721
0.0152678
0.05534488


CIL40
−0.0051299
−0.00194
−0.0132347
0.00416077
−0.0001902


CIL41
0.03738938
0.0042815
−0.0717017
0.02281886
0.12025054


CIL44
−0.0057439
−0.0039999
−0.0210979
0.02136815
0.02689059


CIL46
−0.0019823
0.00167494
−0.0076843
0.01132234
−0.0067813


CIL47
−0.0087651
0.00218279
−0.0100931
0.04658627
0.02649361


CIL48
0.00165808
−0.0099776
−0.0024138
7.0978E−05
0.02169423


CIL49
0.00344753
−0.0033156
0.02246486
0.00922985
0.0342359


CIL50
0.00128911
−0.0014853
−0.0064271
−0.0179113
0.01111688


CIL51
0.00509238
0.01542012
−0.0031793
0.01271012
0.00143333


CIL52
0.02401098
0.007127
−0.0081211
0.02434163
−0.0228692


CIL55
0.04440885
0.00539614
−0.0374682
0.01360865
0.11427443


CIL56
0.0959978
0.0126835
−0.0879927
0.13656299
0.20114056


CIL58
0.00775971
−0.0126765
−0.0598748
−0.0460517
−0.0318612


CIL60
−0.0006376
0.01083176
0.00027971
−0.0200117
0.01614973


CIL62
0.10375755
−0.008295
−0.011172
−0.0300719
−0.0151454


CIL63
0.0101043
0.01385659
−0.0072659
−0.0038932
0.03264314


CIL64
0.08111458
−0.0268927
−0.2293971
−0.0559641
−0.0191125


CIL66
0.02128777
0.00624294
0.03361222
0.0051314
0.01890169


CIL67
0.02262162
0.01839263
−0.0186259
0.00417225
0.00260433


CIL69
0.06650416
0.01186413
−0.0908984
0.19370261
0.32267551


CIL70
0.03638609
0.01868934
−0.0889235
−0.0310472
0.09641879


CIL71
0.00574381
−0.0008016
−0.020035
−0.0018413
0.00294299


CIL72
0.0135119
−0.0136805
−0.0144137
0.02906901
0.04439213


CIL74
0.00310431
0.00621638
0.00195294
0.00303427
0.00203069


CIL75
0.07488577
0.01122834
−0.0700855
0.26036826
0.35244208


CIL76
0.01775768
0.00123696
0.00722099
0.01034265
0.07720638


CIL77
0.00338189
0.00162175
−0.0188939
0.00547294
0.00650936


CIL79
0.07472586
0.01455371
−0.0471988
0.3457588
0.4407363


CIL80
0.00204116
−0.0123765
0.0200436
0.0260488
−0.0104464


CIL87
−0.0074561
−0.0048804
−0.0099304
−0.0010104
0.00227418


CIL88
0.00351822
0.0070051
−0.0211583
0.00992871
0.00960663


CIL89
0.00283255
0.01266621
−2.169E−05
0.00753856
0.01092111






HT.atoc
HT.bCarotene
HT.deferox
HT.digoxin
HT.trolox





MS275
−0.0101635
−0.030054
−0.1652583
−0.1792504
−0.0259902


Scriptaid
−0.0093307
−0.0187192
−0.1469224
−0.0879295
−0.0298598


TrichostatinA
−0.0050621
−0.0347195
−0.1597123
−0.1260736
−0.0338165


Echinomycin
0.14638974
−0.0049676
0.1664907
0.09969184
−0.0242328


Colchicine
−0.0173042
0.01722509
0.29837397
0.13591788
0.01590927


NPC25
0.02932732
−0.0021003
0.36869446
0.11404384
0.00377491


NPC4
−0.0046734
−0.0040981
0.30213611
0.1271105
−0.0147566


NPC7
0.09995549
−0.0175912
0.22103456
0.09504661
0.00125913


Podophyllotoxin
−0.0202323
0.0009225
0.31039865
0.10979609
0.01030472


Rotenone
0.04769067
0.00359379
0.34697219
0.16171241
0.00389493


Vinbiastine
0.15731554
0.00194469
0.28465035
0.1244578
−0.0019595


Vincristine
0.01852742
0.01182267
0.32174322
0.14200631
−0.0159878


DPI3
0.48830882
−0.0093433
0.50494601
−0.0771195
0.43071723


DPI4
0.35227875
−0.0126362
0.40953431
−0.0634303
0.31508429


DPI6
0.43573174
−0.0104991
0.41669672
−0.0498795
0.36282227


RSL3
0.44706907
−0.0134625
0.35079526
−0.1463351
0.42129597


Bortezomib
0.00702342
−0.0191437
−0.0167225
0.07916355
−0.0041833


MG132
0.04884257
7.0861E−05
−0.1161718
0.08352183
−0.0161622


MG262
0.05222066
−0.0092548
−0.0846482
0.05596798
−0.0206824


Erastin
0.26429415
−0.0214976
0.3025221
−0.0339298
0.20770296


Camptothecin
0.00400806
0.00283709
0.12877867
0.14416065
0.01719276


Irinotecan
0.01845108
0.00045945
0.09054955
0.08432789
0.01452359


Daunorubicin
0.00989487
0.00826483
0.15149319
0.13983003
−0.002722


Doxorubicin
0.00992997
0.01512156
0.20435466
0.19812467
−0.0141349


Etoposide
−0.0062763
0.00392981
0.14284045
0.13112076
0.01328409


Mitoxantrone
0.04839288
−0.0039153
0.10012715
0.13212973
−0.0138833


Cycloheximide
0.0166659
0.00875763
0.20413327
0.045701
0.02126525


Dinitrophenol
0.09943976
−0.0099967
0.11141446
−0.0713337
0.03606919


NaN3
−0.0310041
0.00681043
0.07881732
0.0194835
−0.0013756


Vatinomycin
0.22919637
−0.015257
0.00414526
−0.1615377
0.04462648


CIL1
0.21597804
−0.0284574
0.0835898
−0.0038093
0.02477115


CIL2
0.0475732
−0.0268688
0.02962643
−0.0122222
−0.0309394


CIL4
0.02362326
0.00102644
−3.785E−05
−0.0238929
−0.0201009


CIL5
0.03602558
−0.0140327
−0.0102471
0.00553377
0.0124658


CIL6
0.12044039
−0.0173022
0.02051429
0.00960657
−0.0169697


CIL7
0.02896411
−0.0174047
−0.0507117
0.00386935
−0.0063211


CIL9
0.05157132
−0.0065015
0.05678115
−0.0198389
−0.0359906


CIL10
0.10336708
−0.0120139
−0.0043187
0.01443156
−0.006674


CIL11
0.07926238
−0.0182738
0.02780207
−0.0137624
−0.0211487


CIL13
0.10290408
0.0100602
0.05411474
0.03512182
−0.0397506


CIL15
0.12236417
−0.0039057
0.04236842
−0.0123817
−0.0295595


CIL16
0.10252396
−0.0132248
0.05919129
−0.0188439
−0.015239


CIL17
0.1450374
−0.005211
0.02893647
−0.0031893
0.00459499


CIL18
0.12951956
−0.0001406
−0.0099506
−0.0041984
−0.0120546


CIL20
0.02920257
0.00861751
0.03912145
0.01850849
0.03002217


CIL22
0.04912063
−0.0117414
0.06506518
0.05675427
−0.0049209


CIL23
0.03662668
−0.0011222
0.15124603
0.03673254
0.0044018


CIL25
0.04405336
0.00908228
0.07762606
−0.0071491
−0.025622


CIL26
0.04649457
−0.0334855
−0.1033088
−0.0364118
−0.0326987


CIL27
0.04361958
−0.0116561
0.14407851
0.0012616
−0.0594296


CIL28
0.10678443
−0.0334142
−0.0431247
−0.0071044
−0.0302473


CIL33
0.04094235
−0.0165102
0.10561421
−0.034127
−0.0507868


CIL34
0.08124871
0.00132076
0.11145397
−0.0103167
−0.0299319


CIL36
0.08941918
−0.0052761
0.06451547
−0.0355653
0.00242516


CIL40
0.12315101
−0.0178454
−0.0811097
−0.0353912
−0.0276823


CIL41
0.31598831
−0.0218245
0.17951591
−0.0925466
0.16096292


CIL44
0.04386167
−0.0201165
0.10411951
0.01991823
−0.0210643


CIL46
0.04736192
−0.01026
0.03466786
−0.0467148
−0.0144967


CIL47
0.04666403
−0.0018324
0.01073579
0.03488622
−0.0382838


CIL48
0.09021444
−0.0084499
0.12681006
−0.015135
−0.0261174


CIL49
0.02137465
−0.0260633
0.07453051
0.00164748
−0.0152272


CIL50
0.06547382
0.01960927
−0.0408397
0.02124336
−0.0156542


CIL51
0.05134539
0.03796139
0.00406195
−0.0382123
−0.012566


CIL52
0.07145737
0.06783834
0.23249093
−0.005506
−0.0394203


CIL55
0.20744323
0.00129686
0.11081063
−0.0490999
0.14692164


CIL56
0.24364778
0.05422681
0.12694489
−0.1401832
0.14210439


CIL58
0.03304649
0.08558235
0.08912049
0.02289948
−0.0479106


CIL60
0.08708149
0.04978843
0.07701682
−0.036018
−0.0265017


CIL62
0.08235099
0.02726492
0.10607881
−0.0244838
−0.0312336


CIL63
0.07268938
0.06439766
0.08988671
0.02370705
0.03221966


CIL64
−0.0624863
0.16540942
0.12701332
0.03480285
−0.0807861


CIL66
0.05045432
0.03186052
0.12989118
−0.0545586
−0.0333379


CIL67
0.05005779
0.04656953
0.11763528
−0.0217528
−0.0568708


CIL69
0.34563046
−0.0166997
0.30053665
−0.0261029
0.26312402


CIL70
0.27332319
−0.0017074
0.2561347
−0.1365882
0.12049926


CIL71
0.03732862
−0.0128102
−0.0036536
−0.0104564
−0.013114


CIL72
−0.0211095
−0.0123259
0.19715488
−0.0141282
−0.0359053


CIL74
0.07412252
−0.0203197
−0.0089452
−0.0258559
−0.0236027


CIL75
0.3815567
−0.0187117
0.40089859
−0.0369968
0.3451772


CIL76
0.00326543
−0.0071296
0.13758731
0.01031687
−0.0270673


CIL77
0.02076032
−0.0050775
0.09144811
−0.0140507
−0.0106377


CIL79
0.44882849
0.00567531
0.54943789
−0.0705
0.42246924


CIL80
0.0048774
0.00387361
0.16715785
0.06492902
−0.0195351


CIL87
0.05929111
0.01926934
0.07034052
0.04582203
−0.0318765


CIL88
0.07161912
0.02317928
0.06160293
0.05526244
−0.0157674


CIL89
0.06709686
0.01857298
0.04586863
0.01145507
−0.0214735






HT.zVAD









MS275
0.02219614






Scriptaid
0.02988417






TrichostatinA
0.0401151






Echinomycin
0.07367089






Colchicine
0.13398874






NPC25
0.18370661






NPC4
0.13439139






NPC7
0.09516486






Podophyllotoxin
0.09401676






Rotenone
0.01972194






Vinbiastine
0.08923891






Vincristine
0.11717599






DPI3
−0.0520186






DPI4
−0.0445076






DPI6
−0.0403944






RSL3
−0.0159859






Bortezomib
0.01074076






MG132
−0.0992034






MG262
−0.0583711






Erastin
−0.0720745






Camptothecin
0.03249301






Irinotecan
0.03379276






Daunorubicin
0.04455104






Doxorubicin
0.05137839






Etoposide
0.03817009






Mitoxantrone
0.03446406






Cycloheximide
0.03579612






Dinitrophenol
0.04232695






NaN3
0.01031129






Vatinomycin
−0.0575268






CIL1
−0.0551038






CIL2
−0.028281






CIL4
0.01460239






CIL5
−0.0137832






CIL6
−0.0167612






CIL7
−0.0377272






CIL9
−0.0016603






CIL10
−0.0128631






CIL11
−0.0024945






CIL13
0.0116389






CIL15
−0.0468174






CIL16
0.00548187






CIL17
0.00853559






CIL18
0.0100969






CIL20
−0.0207892






CIL22
0.02835078






CIL23
0.02042705






CIL25
0.03342493






CIL26
−0.0122635






CIL27
−0.0150776






CIL28
−0.0090028






CIL33
0.03456928






CIL34
0.00714776






CIL36
−0.0035883






CIL40
0.01716741






CIL41
−0.016697






CIL44
0.01751267






CIL46
0.0172399






CIL47
−0.0058941






CIL48
−0.0050534






CIL49
0.00987936






CIL50
0.02906893






CIL51
0.00779885






CIL52
0.02338762






CIL55
0.02659525






CIL56
0.03666347






CIL58
0.0245897






CIL60
−0.0099218






CIL62
−0.0179367






CIL63
0.0184627






CIL64
0.05556764






CIL66
0.01628958






CIL67
0.00499615






CIL69
−0.0004734






CIL70
0.00227166






CIL71
0.01755953






CIL72
0.05313729






CIL74
0.01022255






CIL75
0.00011911






CIL76
0.03199583






CIL77
0.02553717






CIL79
0.00368164






CIL80
0.03135047






CIL87
0.0225789






CIL88
0.04370741






CIL89
0.03057583









Table 9 provides the raw data used to generate the hierarchical clustering in FIG. 1B. In Table 9, rows are lethal compounds; columns are modulators; values are AUCs.


Example 3
Probe Optimization Led to a Specific Ferroptosis Inducer

CIL56 induced iron-dependent ROS (FIG. 2B). Antioxidants and iron chelators suppressed the lethality of low concentrations of CIL56 (FIG. 2C). It was hypothesized that CIL56 was capable of engaging two independent death pathways: ferroptosis at low concentrations, and necrotic, non-suppressible cell death at higher concentrations. To identify a more selective analog of CIL56 that retained the ability to induce ferroptosis but lacked the ability to induce the other form of death, structure—activity relationship analysis of the CIL56 scaffold was carried out, which determined that the oxime moiety was crucial for inducing ferroptosis, and that hydrophobicity of the piperidine moieties of CIL56 correlated with potency (FIG. 11). Ultimately, an analog (SRS7-34) that has cyclohexylamine moieties in place of the piperidine moieties of CIL56 was discovered (FIG. 2D). This new compound, termed FIN56 (2) (ferroptosis inducer derived from CIL56), exhibited greater potency as well as greater oncogenic RAS selectivity in the BJ cell line series compared to CIL56 (FIG. 2E); FIN56 was also fully suppressed by the ferroptosis inhibitors deferoxamine and α-tocopherol (FIG. 2F), indicating that it engages only ferroptosis and does not have the ability to engage the second death mechanism activated by CIL56.


Example 4
FIN56-Induced Ferroptosis Involves Decreased GPX4 Abundance

Previously reported ferroptosis inducers either deplete glutathione (by inhibiting cystine uptake) or are covalent GPX4 inhibitors. In the NCI60 cell line panel (Shoemaker et al. 2006), it was found that GPX4 inhibitors were more cell-line selective than compounds inducing glutathione depletion (Shimada et al. 2016) (FIG. 12A). In this regard, FIN56 was more similar to other GPX4 inhibitors than to GSH-depleting compounds. Supporting this, it was found that FIN56 did not deplete glutathione, which suggested that it also did not block cystine import; however, FIN56 did cause the loss of GPX4 activity in cell lysates (FIG. 3A and FIG. 12B). Intriguingly, compared to the covalent GPX4 inhibitor (1S, 3R)-RSL3, FIN56 was slower to induce the accumulation of ROS (FIG. 3B), which suggested that it did not cause a loss of GPX4 activity via direct inhibition of enzymatic activity. Indeed, it was determined that the abundance of GPX4, but not that of the related selenoprotein GPX1, was substantially decreased after FIN56 treatment (5 μM, 10 h) (FIG. 3C and FIGS. 12C-12D). Neither erastin nor (1S, 3R)-RSL3 affected the abundance of GPX4 to the same extent as FIN56, demonstrating that this effect was specific to FIN56. GPX4 knockdown enhanced FIN56 lethality (FIG. 3D), and FIN56-induced cell death was suppressed by GFP-GPX4 fusion-protein overexpression (FIGS. 3E-F and FIGS. 12E-12F), suggesting that the loss of GPX4 was critical for FIN56-induced ferroptosis. It was confirmed that selenite supplementation, which is known to upregulate the expression of selenoproteins, including GPX4 (Romanowska et al. 2007), also suppressed FIN56 lethality (FIGS. 12G-12H). The decrease in GPX4 protein abundance was not inhibited by α-tocopherol, indicating that it was not a downstream consequence of lipid ROS generation. Together, these results suggested that FIN56 triggers ferroptosis through a mechanism involving the downregulation of GPX4 protein abundance.


How FIN56 causes a decrease in the amount of GPX4 protein was further investigated. It was found that the GPX4 transcript level increased, rather than decreased, after FIN56 treatment (FIG. 12, panel (i)), which suggests that FIN56-induced depletion of GPX4 protein is not mediated by transcriptional changes in GPX4 mRNA. Observations that cells treated with cycloheximide, which inhibits ribosome function, did not show substantially decreased amounts of GPX4 protein compared to a housekeeping α-tubulin protein (FIG. 12J) and that knockdown of the gene tRNA isopentenyltransferase 1 (TRIT1) (Fradejas et al. 2013), which has been reported to be required for synthesis of selenoproteins, including of GPX4, did not dramatically affect GPX4 protein abundance (FIGS. 12K-12M) suggested that FIN56 did not inhibit GPX4 protein synthesis but rather induced post-translational GPX4 protein degradation. It is not clear, however, how GPX4 degradation is regulated, because a proteosome inhibitor (MG132) did not inhibit GPX4 degradation significantly or protect cells from FIN56 lethality. It is of note that GPX4 abundance was reported to decrease when the proto-oncogene serine/threonine-protein (Pim) kinases were inhibited (Song et al. 2015); however, this is not relevant to FIN56 lethality because pan-Pim kinase inhibitors did not induce ferroptosis, as these compounds were not suppressed by α-tocopherol (FIG. 12N).


Example 5
SQS Encoded by FDFT1 is a Target Protein of FIN56

To better understand the mechanism of action of FIN56, a search for proteins binding directly to FIN56 was conducted using a chemoproteomic approach. First, structural analogs of FIN56 were explored. This resulted in the creation of SRS11-31, an analog with a polyethylene glycol (PEG) moiety, which induces ferroptosis at tenfold higher EC50 than FIN56 (FIG. 4A and FIG. 13). In contrast, substitution of the cyclohexyl moiety in FIN56 with a 4-tetrahydropyran (SRS8-18 (3)) or its PEG conjugate (SRS11-66) (4) resulted in a complete loss of activity. Next, both SRS11-31 (5) (active, or A) and SRS11-66 (inactive, or I) were conjugated to Profinity epoxide resin via an epoxy ring-opening reaction and incubated the resins with HT-1080 whole-cell lysates. Mass spectrometry was used to identify and quantify the pulldown proteins found with each probe. Seventy proteins, excluding universally expressed proteins (actins, tubulins, and ribosome subunits), were found to be more abundant on the resin conjugated with the active probe.


It was then tested that whether these candidate target proteins were inhibited (loss of function) or activated (gain of function) by FIN56 to induce ferroptosis using RNA interference (RNAi). RNAi-mediated knockdown of the relevant target should either enhance or suppress FIN56 sensitivity, depending on FIN56's mechanism of action (FIG. 14A). Expression of many genes is affected by off-target effects of RNAi, and the resulting phenotypes may differ from cell line to cell line; however, on-target effects are more likely to be consistent among different cell lines. Therefore, the effects of up to five short hairpin RNA (shRNA) clones per gene encoding 70 candidate target proteins in four independent ferroptosis-susceptible cell lines: HT-1080, BJeLR, Calu-1 lung adenocarcinoma, and 143B osteosarcoma were examined (FIG. 4B and FIGS. 14B-14C). A search for candidate FIN56 targets responsible for ferroptosis was conducted using two criteria: (i) high selective abundance on the active versus inactive probe resins, and (ii) high proportion of consistently performing shRNAs in all four cell lines subjected to RNAi-mediated silencing of each gene (FIG. 4C and FIG. 14D). Among the 70 tested proteins, it was found that proteins, such as chaperones and nuclear transport proteins, for which knockdown not only enhanced FIN56 lethality but also independently induced toxicity. However, their lethality was not suppressed by α-tocopherol, unlike with GPX4 knockdown, indicating that these cell death phenotypes were distinct from ferroptosis. The potency of FIN56 might be aided by modulation of these essential proteins, but they are not the primary targets of FIN56 (FIG. 4D).


Example 6
Validating the Functional Relevance of the Target

It was found that four of the five shRNAs against FDFT1 mRNA (which encodes SQS protein) suppressed FIN56 consistently in all four cell lines tested, indicating that FIN56 activates, rather than inhibits, SQS (a gain-of-function model). Therefore, how the FIN56-SQS interaction is relevant to FIN56's lethality was investigated. It was confirmed that not only shRNAs targeting FDFT1 but also small-molecule inhibitors of SQS activity (YM-53601 and zaragozic acid A) suppressed FIN56 lethality (FIG. 5A). SQS is an enzyme that acts downstream of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase in the mevalonate pathway. SQS couples two farnesyl pyrophosphate (FPP) molecules to form squalene. Inhibition of SQS consequently increases the pool of FPP. FPP is essential for several processes, including protein prenylation and metabolite synthesis (e.g., sterols, coenzyme Q10 (CoQ10), dolichol, and heme) (Tansey et al. 2000), some subset of which may be relevant to the modulatory effect on ferroptosis sensitivity. Supplementation with FPP indeed suppressed the lethality of FIN56 (FIG. 5B). SQS-FIN56 binding was also examined by confirming that SQS from HT-1080 whole-cell lysate bound selectively to active probes versus to an inactive probe (FIG. 5C). Moreover, bacterially expressed truncated human SQS protein (Liu et al. 2014) (with the lipophilic N and C termini removed) binding to an active affinity probe was efficiently suppressed by pre-incubation of purified SQS protein with FIN56, suggesting that SQS and FIN56 directly interact (FIG. 15).


The role of processes upstream of FPP in the mevalonate pathway was investigated. It was found that statins, chemical inhibitors of HMG-CoA reductase, enhanced FIN56 lethality (FIG. 16A). HMG-CoA reductase synthesizes mevalonic acid (MVA). Supplementation of MVA reversed the effect of cerivastatin, the most potent and selective statin tested (FIGS. 16B-16C), demonstrating that the HMG-CoA-reductase-inhibiting effect of cerivastatin is responsible for its enhancement of FIN56 lethality. However, these effects did not affect GPX4 abundance (FIG. 16D).


More extensive investigation of the mevalonate pathway showed that supplementation of FPP suppressed FIN56 more effectively than treatment with other isoprenoid pyrophosphates did, which suggests that FPP is functionally relevant to the regulation of sensitivity to FIN56 (FIG. 5D). It was found that an inhibitor of squalene monooxygenase, which is involved in a rate-limiting step downstream of SQS in cholesterol synthesis (Chugh et al. 2003), and an SQS inhibitor both suppressed FIN56 lethality, supporting the notion that nonsteroidogenic products of the mevalonate pathway contribute to the suppression of FIN56 (FIG. 5E). It was found that among the metabolites derived from FPP, idebenone, a hydrophilic analog of CoQ10, was the only suppressor of FIN56-induced ferroptosis (FIGS. 5F-5G), which suggests that CoQ10 mediates the connection between the mevalonate pathway and regulation of sensitivity to FIN56. Note that supplementation of CoQ10 itself is known to be ineffective because of its extreme hydrophobicity (Gueven et al. 2015). To further assess the specificity of the three mevalonate-pathway modulators (FPP, YM-53601, and idebenone), and of α-tocopherol, a modulatory profiling scheme was used in HT-1080 cells with lethal compounds, including ones that induce oxidative stress. It was found that both GPX4 inhibitors, FIN56 and (1S, 3R)-RSL3, were potently suppressed by all the compounds (FIG. 5H). Erastin, a GSH depletor, was also suppressed somewhat by the modulators; the rest of the compounds showed more distinct patterns. These results suggest that modulators of the mevalonate pathway are specific inhibitors of ferroptosis, rather than of lethality in general.


CoQ10 is an electron carrier in the mitochondrial respiratory chain and an endogenous antioxidant. However, whether it functions in the regulation of ferroptosis via either of these mechanisms remains to be elucidated. 143B cells with or without mitochondrial DNA (ρ+ or ρ0 cells, respectively) were both sensitive to FIN56-induced ferroptosis (FIGS. 17A-17B), which suggests that the respiratory chain is not involved in ferroptosis. The lipophilic antioxidant α-tocopherol suppressed staurosporine-induced cell death, but idebenone enhanced it, suggesting that idebenone has distinct antioxidant properties compared to α-tocopherol. In addition, although both α-tocopherol and idebenone inhibited lipid ROS generation after FIN56 treatment, idebenone treatment did not change the basal lipid ROS level (FIG. 17C). Thus, exactly how CoQ10 protects cells from FIN56 remains elusive, but the process may involve reprogramming of lipid metabolism in a way that is not conducive to the execution of ferroptosis, or a specific antioxidant action distinct from that of α-tocopherol.


Example 7
An Acc Inhibitor Prevents GPX4 Protein Degradation

Modulators of the mevalonate pathway, such as idebenone, are potent suppressors of ferroptosis, particularly of direct and indirect GPX4 inhibitors (FIN56 and (1S, 3R)-RSL3) (FIG. 5H); however, these compounds did not block the decrease in GPX4 protein abundance caused by FIN56 treatment or induce overexpression of the protein (FIG. 6A and FIG. 18), indicating that there may be an additional pathway that regulates GPX4 protein level in response to FIN56 treatment. It was discovered that 5-(tetradecyloxy)-2-furoic acid (TOFA), an inhibitor of acetyl-CoA carboxylase (ACC), inhibited the loss of GPX4. TOFA was also found to be a potent suppressor of FIN56 and suppressed lipid ROS generation after FIN56 treatment (FIGS. 6B-6C). ACC is an enzyme involved in fatty acid synthesis. It was previously observed through genetic screening that ACC's activity is involved in the mechanism of non-ferroptotic cell death induced by CIL56 (Dixon et al. 2015). However, ACC itself was not identified as a direct FIN56 target, and the detailed mechanism linking FIN56 to ACC remains to be understood.


In conclusion, characterization of FIN56 revealed that its mechanism involves two distinct pathways (FIG. 6D): degradation of GPX4, which requires the enzymatic activity of ACC; and activation of SQS, which leads to coenzyme Q10 depletion. Together these effects result in potent induction of ferroptosis.


Example 8
Total Synthesis and NMR Data of CIL56 and FIN56 and Analogs thereof

Structural analogs of CIL56 and FIN56 were synthesized based on the procedures described in international patent application no. PCT/US2008/006015 by Cholody, W. M. et al. (2008), which is incorporated herein by reference in its entirety. The total synthesis of CIL56 and FIN56 is depicted in Scheme 1.




embedded image



General Procedure A: Preparation of Ketone Compounds


9-oxo-9H-fluorene-2,7-disulfonyl dichloride (6) (1 equiv.) was dissolved in 50 mL of dichloromethane and the mixture was cooled to −50° C. To this mixture was added a primary or secondary amine (e.g. piperidine or cyclohexylamine) (2.6 equiv.) and diisopropylethylamine (2-3 equiv.). The reaction mixture was stirred at room temperature for 17 hrs. The solvent was evaporated and the residue was purified by flash-column chromatography on silica gel to provide the desired ketone compounds. The purity of the ketones were confirmed by different spectroscopic methods such as 1H NMR and mass spectrometry.


General Procedure B: Preparation of Oxime Compounds


A mixture of the ketone from the general procedure A (1.0 equiv.) and hydroxylamine hydrochloride (10 equiv.) were dissolved in pyridine (10 mL). The mixture was stirred at 95° C. for 36 hrs. The pyridine was evaporated and the residue was stirred with 1 N hydrogen chloride (HCl) (10 mL) for several mins. White product was collected by filtration, washed with water and dried. The crude material was either crystallized from ethanol or purified by flash-column chromatography on silica gel to provide the desired oxime compounds. The purity of the oximes was confirmed by different spectroscopic methods such as 1H NMR and mass spectrometry.


Synthesis of 2,7-bis(piperidin-1-ylsulfonyl)-9H-fluoren-9-one (SRS1-63)



embedded image


General procedure A was followed, using 9-oxo-9H-fluorene-2,7-disulfonyl dichloride (6) (400 mg, 1.060 mmol), piperidine (273 mL, 2.757 mmol) and diisopropylethylamine (270 mL, 2.120 mmol). The crude reaction mixture was purified by column chromatography (dichloromethane:methanol=40:1) to give the desired 2,7-bis(piperidin-1-ylsulfonyl)-9H-fluoren-9-one (SRS1-63) (7) (420 mg, 0.886 mmol, 84%). 1H NMR (400 MHz, CDCl3) δ 8.07 (s, 2H), 7.99 (d, J=7.4 Hz, 2H), 7.79 (d, J=7.8 Hz, 2H), 3.04 (d, J=4.8 Hz, 8H), 1.66 (s, 8H), 1.44 (s, 4H). MS (APCI+, M+1) 475.16.


Synthesis of N2,N7-dicyclohexyl-9-oxo-9H-fluorene-2,7-disulfonamide (SRS7-25)



embedded image


General procedure A was followed, with 9-oxo-9H-fluorene-2,7-disulfonyl dichloride (1) (500 mg, 1.326 mmol), cyclohexylamine (394 mL, 3.448 mmol) and diisopropylethylamine (692.9 mL, 3.978 mmol). The crude reaction mixture was purified by column chromatography (dichloromethane:methanol=20:1) to give the desired N2,N7-dicyclohexyl-9-oxo-9H-fluorene-2,7-disulfonamide (SRS7-25) (8) (532 mg, 1.059 mmol, 80%). 1H NMR (400 MHz, CDCl3) δ 8.19 (s, 2H), 8.13 (dd, J=7.8, 1.6 Hz, 2H), 7.77 (d, J=7.8 Hz, 2H), 4.57 (d, J=7.7 Hz, 2H), 3.22 (d, J=3.8 Hz, 2H), 1.81 (d, J=9.8 Hz, 4H), 1.65 (s, 4H), 1.32-1.13 (m, 12H); MS (APCI+, M+1) 503.01.


Synthesis of 2,7-bis(piperidin-1-ylsulfonyl)-9H-fluoren-9-one oxime (CIL56)



embedded image


General procedure B was followed, with 2,7-bis(piperidin-1-ylsulfonyl)-9H-fluoren-9-one (SRS1-63) (350 mg, 0.738 mmol), hydroxylamine hydrochloride (509 mg, 7.384 mmol) and pyridine (30 mL). The crude reaction mixture was precipitated as salt after 1 N HCl treatment and washed several times with cold water and ethanol to give the N2,N7-dicyclohexyl-9-oxo-9H-fluorene-2,7-disulfonamide (CIL56) (1) (306 mg, 0.627 mmol, 85%). 1H NMR (400 MHz, CDCl3) δ 2.50 (s, 1H), 8.78 (s, 1H), 8.13 (s, 1H), 7.88-7.77 (m, 3H), 7.75 (d, J=1.4 Hz, 1H), 2.96 (d, J=4.8 Hz, 8H), 1.58 (s, 8H), 1.35 (s, 4H); MS (APCI+, M+1) 490.16.


Synthesis of N2,N7-dicyclohexyl-9-(hydroxyimino)-9H-fluorene-2,7-disulfonamide

(FIN56)




embedded image


General procedure B was followed, with N2,N7-dicyclohexyl-9-oxo-9H-fluorene-2,7-disulfonamide (SRS7-25) (500 mg, 0.996 mmol), hydroxylamine hydrochloride (687.2 mg, 9.960 mmol) and pyridine (30 mL). The crude reaction mixture was precipitated as a salt after 1 N HCl treatment and washed several times with cold water and ethanol to give the N2,N7-dicyclohexyl-9-oxo-9H-fluorene-2,7-disulfonamide (FIN56) (2) (437 mg, 0.845 mmol, 85%). 1H NMR (400 MHz, DMSO) δ 13.28 (s, 1H), 8.82 (s, 1H), 8.25-8.14 (m, 3H), 8.03 (dd, J=8.0, 1.3 Hz, 1H), 7.95 (dd, J=8.0, 1.2 Hz, 1H), 7.85 (d, J=7.4 Hz, 1H), 7.81 (d, J=7.3 Hz, 1H), 3.00 (s, 2H), 1.59 (s, 8H), 1.44 (d, J=11.5 Hz, 2H), 1.22-0.98 (m, 10H); MS (APCI+, M+1) 517.92.


Synthesis 2,7-bis(4,4-difluoropiperidin-1-ylsulfonyl)-9H-fluoren-9-one oxime (SRS5-19)



embedded image


General procedure B was followed, with 2,7-bis(4,4-difluoropiperidin-1-ylsulfonyl)-9H-fluoren-9-one (25 mg, 0.0458 mmol), hydroxylamine hydrochloride (32.0 mg, 0.458 mmol) and pyridine (30 mL). The crude reaction mixture was precipitated as a salt after 1 N HCl treatment and washed several times with cold water and ethanol to give the desired 2,7-bis(4,4-difluoropiperidin-1-ylsulfonyl)-9H-fluoren-9-one oxime (SRS5-19) (10) (15 mg, 0.026 mmol, 58%). 1H NMR (400 MHz, DMSO) δ 13.42 (s, 1H), 8.68 (s, 1H), 8.34 (dd, J=17.8, 7.9 Hz, 2H), 8.09-7.87 (m, 13H), 3.15 (s, 8H), 2.08 (s, 8H); MS (APCI+, M+1) 561.95.


Synthesis of 9-(hydroxyimino)-N2,N7-diphenyl-9H-fluorene-2,7-disulfonamide (SRS5-55)



embedded image


General procedure B was followed, with 9-oxo-N2,N7-diphenyl-9H-fluorene-2,7-disulfonamide (25 mg, 0.051 mmol), hydroxylamine hydrochloride (35.2 mg, 0.510 mmol) and pyridine (30 mL). The crude reaction mixture was precipitated as a salt after 1 N HCl treatment and washed several times with cold water and ethanol to give the desired 2,7-bis(4,4-difluoropiperidin-1-ylsulfonyl)-9H-fluoren-9-one oxime (SRS5-55) (11) (23 mg, 0.045 mmol, 89%). 1H NMR (400 MHz, DMSO) δ 13.32 (s, 1H), 10.36 (d, J=25.6 Hz, 2H), 8.73 (s, 1H), 8.10 (dd, J=14.8, 8.0 Hz, 2H), 8.02 (s, 1H), 7.85 (dd, J=26.4, 8.0 Hz, 2H), 7.29-6.98 (m, 10H); MS (APCI+, M+1) 505.98.


Synthesis of 2,7-bis(4-methylpiperidin-1-ylsulfonyl)-9H-fluoren-9-one oxime (SRS5-57)



embedded image


General procedure B was followed, with 2,7-bis(4-methylpiperidin-1-ylsulfonyl)-9H-fluoren-9-one (25 mg, 0.050 mmol), hydroxylamine hydrochloride (35 mg, 0.5 mmol) and pyridine (30 mL). The crude reaction mixture was precipitated as a salt after 1 N HCl treatment and washed several times with cold water and ethanol to give the desired 2,7-bis(4-methylpiperidin-1-ylsulfonyl)-9H-fluoren-9-one oxime (SRS5-57) (12) (23 mg, 0.044 mmol, 89%). 1H NMR (400 MHz, DMSO) δ 13.36 (s, 1H), 8.65 (s, 1H), 8.31 (dd, J=17.4, 7.9 Hz, 2H), 7.92 (dd, J=35.8, 10.2 Hz, 3H), 3.66 (s, 4H), 3.48 (s, 4H), 2.28 (t, J=11.1 Hz, 3H), 1.66 (d, J=11.7 Hz, 3H), 1.23-1.09 (m, 4H), 0.84 (d, J=5.6 Hz, 6H); MS (APCI+, M+1) 518.08.


Synthesis of 2,7-bis(4-methoxypiperidin-1-ylsulfonyl)-9H-fluoren-9-one oxime (SRS6-13)



embedded image


General procedure B was followed, with 2,7-bis(4-methoxypiperidin-1-ylsulfonyl)-9H-fluoren-9-one (25 mg, 0.047 mmol), hydroxylamine hydrochloride (32 mg, 0.47 mmol) and pyridine (30 mL). The crude reaction mixture was precipitated as a salt after 1 N HCl treatment and washed several times with cold water and ethanol to give the desired 2,7-bis(4-methoxypiperidin-1-ylsulfonyl)-9H-fluoren-9-one oxime (SRS6-13) (13) (19 mg, 0.034 mmol, 74%). 1H NMR (400 MHz, DMSO) δ 13.37 (s, 1H), 8.65 (s, 1H), 8.40-8.24 (m, 2H), 7.94 (d, J=34.2 Hz, 3H), 3.23 (s, 2H), 3.12 (s, 10H), 2.88 (s, 4H), 1.83 (s, 4H), 1.54 (s, 4H); MS (APCI+, M+1) 550.01.


Synthesis of 2,7-bis(3,5-dimethylpiperidin-1-ylsulfonyl)-9H-fluoren-9-one oxime (SRS6-15)



embedded image


General procedure B was followed, with 2,7-bis(3,5-dimethylpiperidin-1-ylsulfonyl)-9H-fluoren-9-one (25 mg, 0.047 mmol), hydroxylamine hydrochloride (32.5 mg, 0.47 mmol) and pyridine (30 mL). The crude reaction mixture was precipitated as a salt after 1 N HCl treatment and washed several times with cold water and ethanol to give the desired 2,7-bis(3,5 dimethyl-piperidin-1-ylsulfonyl)-9H-fluoren-9-one oxime (SRS6-15) (14) (21 mg, 0.038 mmol, 82%). 1H NMR (400 MHz, DMSO) δ 13.37 (s, 1H), 8.64 (d, J=6.2 Hz, 1H), 8.30 (dd, J=17.5, 8.1 Hz, 2H), 8.01-7.83 (m, 3H), 2.18 (t, J=7.2 Hz, 4H), 1.77 (d, J=5.2 Hz, 4H), 1.66 (d, J=7.4 Hz, 4H), 1.47 (s, 4H), 1.23-1.20 (m, 12H); MS (APCI+, M+1) 546.09.


Synthesis of 2,7-bis(4-hydroxypiperidin-1-ylsulfonyl)-9H-fluoren-9-one oxime (SRS6-23)



embedded image


General procedure B was followed with 2,7-bis(4-hydroxypiperidin-1-ylsulfonyl)-9H-fluoren-9-one (25 mg, 0.049 mmol), hydroxylamine hydrochloride (34.1 mg, 0.49 mmol) and pyridine (30 mL). The crude reaction mixture was precipitated as a salt after 1 N HCl treatment and washed several times with cold water and ethanol to give the desired 2,7-bis(4-hydroxypiperidin-1-ylsulfonyl)-9H-fluoren-9-one oxime (SRS6-23) (15) (14 mg, 0.027 mmol, 55%). 1H NMR (400 MHz, DMSO) δ 13.36 (s, 1H), 8.65 (s, 1H), 8.31 (dd, J=17.6, 8.1 Hz, 2H), 7.93 (dd, J=34.3, 8.6 Hz, 2H), 3.50 (s, 45H), 3.22 (s, 95H), 2.79 (s, 92H), 1.74 (s, 92H), 1.45 (s, 87H); MS (APCI+, M+1) 522.03.


Synthesis of 2,7-bis(octahydroisoquinolin-2(1H)-ylsulfonyl)-9H-fluoren-9-one oxime (SRS6-25)



embedded image


General procedure B was followed, with 2,7-bis(octahydroisoquinolin-2(1H)-ylsulfonyl)-9H-fluoren-9-one (25 mg, 0.043 mmol), hydroxylamine hydrochloride (29.6 mg, 0.43 mmol) and pyridine (30 mL). The crude reaction mixture was precipitated as a salt after 1 N HCl treatment and washed several times with cold water and ethanol to give the desired 2,7-bis(octahydroisoquinolin-2(1H)-ylsulfonyl)-9H-fluoren-9-one oxime (SRS6-25) (16) (17 mg, 0.028 mmol, 66%). 1H NMR (400 MHz, DMSO) δ 13.35 (s, 1H), 8.64 (s, 1H), 8.30 (d, J=9.4 Hz, 2H), 7.92 (d, J=34.3 Hz, 3H), 3.72 (s, 2H), 3.57 (s, 2H), 2.26 (s, 4H), 1.91 (s, 4H), 1.59 (d, J=23.0 Hz, 12H), 1.19 (s, 12H); MS (APCI+, M+1) 598.13.


Synthesis of 2,7-bis(3,3-difluoropiperidin-1-ylsulfonyl)-9H-fluoren-9-one oxime (SRS6-27)



embedded image


General procedure B was followed, with 2,7-bis(3,3-difluoropiperidin-1-ylsulfonyl)-9H-fluoren-9-one (25 mg, 0.045 mmol), hydroxylamine hydrochloride (31.5 mg, 0.457 mmol) and pyridine (30 mL). The crude reaction mixture was precipitated as a salt after 1 N HCl treatment and washed several times with cold water and ethanol to give the desired 2,7-bis(3,3-difluoropiperidin-1-ylsulfonyl)-9H-fluoren-9-one oxime (SRS6-27) (17) (14 mg, 0.025 mmol, 55%). 1H NMR (400 MHz, DMSO) δ 13.41 (s, 1H), 8.68 (s, 1H), 8.35 (dd, J=18.0, 8.0 Hz, 2H), 8.00 (dd, J=34.5, 8.1 Hz, 3H), 3.32-3.29 (m, 4H), 3.12 (s, 4H), 1.95 (s, 4H), 1.71 (s, 4H); MS (APCI+, M+1) 562.01.


Synthesis of 9-(hydroxyimino)-N2,N7-bis(tetrahydro-2H-pyran-4-yl)-9H-fluorene-2,7-disulfonamide (SRS8-18)



embedded image


General procedure B was followed with 9-oxo-N2,N7-bis(tetrahydro-2H-pyran-4-yl)-9H-fluorene-2,7-disulfonamide (9) (25 mg, 0.049 mmol), hydroxylamine hydrochloride (34 mg, 0.494 mmol) and pyridine (30 mL). The crude reaction mixture was precipitated as a salt after 1 N HCl treatment and washed several times with cold water and ethanol to give the desired 9-(hydroxyimino)-N2,N7-bis(tetrahydro-2H-pyran-4-yl)-9H-fluorene-2,7-disulfonamide (SRS8-18) (3) (17 mg, 0.033 mmol, 66%). 1H NMR (400 MHz, DMSO) δ 8.77 (s, 1H), 8.22-8.10 (m, 3H), 7.96 (dd, J=33.0, 8.0 Hz, 4H), 3.66 (d, J=11.5 Hz, 4H), 3.17 (dd, J=21.0, 9.6 Hz, 10H), 2.46 (d, J=1.5 Hz, 3H), 1.49 (d, J=11.9 Hz, 5H), 1.41-1.25 (m, 6H); MS (APCI+, M+1) 522.03.


Synthesis of 9-(hydroxyimino)-N2,N7-dimethyl-N2,N7-diphenyl-9H-fluorene-2,7-disulfonamide (SRS6-51)



embedded image


General procedure B was followed with N2,N7-dimethyl-9-oxo-N2,N7-diphenyl-9H-fluorene-2,7-disulfonamide (25 mg, 0.048 mmol), hydroxylamine hydrochloride (33 mg, 0.48 mmol) and pyridine (30 mL). The crude reaction mixture was precipitated as a salt after 1 N HCl treatment and washed several times with cold water and ethanol to give the desired 9-(hydroxyimino)-N2,N7-dimethyl-N2,N7-diphenyl-9H-fluorene-2,7-disulfonamide (SRS6-51) (18) (21 mg, 0.039 mmol, 82%). 1H NMR (400 MHz, DMSO) δ 13.17 (s, 5H), 8.46 (s, 4H), 8.21 (dd, J=14.4, 8.1 Hz, 10H), 7.73-7.62 (m, 16H), 7.34 (d, J=7.6 Hz, 32H), 7.16 (d, J=7.0 Hz, 23H), 3.19 (s, 40H); MS (APCI+, M+1) 534.01.


Synthesis 2,7-bis(morpholinosulfonyl)-9H-fluoren-9-one oxime (SRS2-95)



embedded image


General procedure B was followed with 2,7-bis(morpholinosulfonyl)-9H-fluoren-9-one (25 mg, 0.052 mmol), hydroxylamine hydrochloride (36 mg, 0.52 mmol) and pyridine (10 mL). The crude reaction mixture was precipitated as a salt after 1 N HCl treatment and washed several times with cold water and ethanol to give the 2,7-bis(morpholinosulfonyl)-9H-fluoren-9-one oxime (SRS2-95) (19) (18 mg, 0.037 mmol, 70%). 1H NMR (400 MHz, CDCI3) δ 7.93 (d, J=7.4 Hz, 1H), 7.88-7.77 (m, 2H), 3.12-2.96 (m, 4H), 1.69 (d, J=5.0 Hz, 4H); MS (APCI+, M+1) 494.09.


Synthesis of SRS11-31 and SRS11-66 for Target Identification Study

The total synthesis of SRS11-31 and SRS11-66 is depicted in Scheme 2.




embedded image


The N2,N7-dicyclohexyl-9-oxo-9H-fluorene-2,7-disulfonamide (FIN56) or 9-(hydroxyimino)-N2,N7-bis(tetrahydro-2H-pyran-4-yl)-9H-fluorene-2,7-disulfonamide (SRS8-18) were reacted with 2-(2-(2-chloroethoxy)ethoxy)ethyl benzoate (1 equiv) in the presence of sodium iodide (0.5 equiv) and potassium carbonate (3 equiv.) in DMF. The mixture was heated at 50° C. for 17 h then aqueous ammonium chloride was added. The compounds were extracted with ethyl acetate, dried with magnesium sulfate and the organic solvent was evaporated under vacuum. The mass of the desired ester intermediates were confirmed by LC/MS and used without further purification. To the ester intermediates were added sodium hydroxide (NaOH; 5 equiv.) in THF (2 ml). The mixture were stirred for 17 h at room temperature then acidified to pH=5. The compounds were extracted with ethyl acetate, dried with magnesium sulfate and the solvent was evaporated under vacuum. The crud material was either crystallized from ethanol or purified by flash-column chromatography on silica gel to provide the desired oxime compounds. The purity of the oxime alcohol linkers SRS11-31 (4) and SRS11-66 (5) were confirmed by 1H NMR and Mass.


N2,N7-dicyclohexyl-9-(2-(2-(2-hydroxyethoxy)ethoxy)ethoxyimino)-9H-fluorene-2,7-disulfonamide (SRS11-31) (5)




embedded image



1H NMR (400 MHz, CDCl3) δ 8.79 (d, J=12.2 Hz, 1H), 8.29 (d, J=7.8 Hz, 1H), 8.07-8.00 (m, 1H), 8.00-7.92 (m, 1H), 7.80 (dd, J=12.1, 5.8 Hz, 2H), 5.90 (t, J=11.3 Hz, 1H), 5.10 (dd, J=13.9, 9.9 Hz, 1H), 4.63 (s, 2H), 3.74 (d, J=3.9 Hz, 8H), 3.65 (d, J=3.9 Hz, 2H), 3.16 (s, 2H), 1.82 (s, 8H), 1.62 (s, 4H), 1.51 (d, J=10.4 Hz, 2H), 1.21 (s, 6H). MS (APCI+, M+1) 650.19.


9-(2-(2-(2-hydroxyethoxy)ethoxy)ethoxyimino)-N2,N7-bis(tetrahydro-2H-pyran-4-yl)-9H-fluorene-2,7-disulfonamide (SRS11-66) (4)



embedded image



1H NMR (400 MHz, CDCl3) δ 8.83 (s, 1H), 8.31 (s, 1H), 8.06 (d, J=7.3 Hz, 1H), 7.97 (d, J=7.7 Hz, 1H), 7.82 (t, J=7.6 Hz, 2H), 6.22 (d, J=7.2 Hz, 1H), 4.92 (d, J=7.3 Hz, 1H), 4.66 (s, 2H), 3.96 (s, 4H), 3.76 (d, J=9.2 Hz, 2H), 3.70 (s, 2H), 3.34-3.30 (m, 4H), 1.69 (s, 4H), 1.53 (d, J=6.2 Hz, 2H), 1.25 (dd, J=35.5, 16.2 Hz, 10H); MS (APCI+, M+1) 654.01.


Example 9
Enforcing Cell-Based Phenotypic Screening with Polypharmacology to Interpret Compound Mechanisms of Action

Cell-based phenotypic high-throughput screening using chemical libraries has been a successful approach for modern drug discovery for decades. Many small molecules with novel biological activities have been discovered in this manner. However, characterizing the mechanism of action of hit compounds discovered through this approach is often a challenging problem. We proposed a novel computational framework to approach this problem using ligand-based pharmacophore target prediction program, known as Similarity Ensemble Approach (SEA). Integrating phenotypic screening with prediction of target proteins, we sought to understand the multi-modal mechanisms of cell death induced by a lethal compound, CIL56. Since it is computationally inexpensive, this polypharmacology approach will serve as a compensatory approach to interpret the phenotypic screening results.


High-throughput screening of small molecules has been essential tool not only in drug discovery but also in basic chemical biology problems for decades. Two major screening strategies are target-based and phenotype-based: target-based screening seeks compounds that bind to and perturb proteins of interest in vitro and is used when a particular target is already implicated in disease. However, in many more cases, disease-modifying targets are unknown. In these cases, phenotypic screening of small molecules for activity in cellular models of disease is powerful both to investigate disease mechanisms and to discover potential drugs.


However, even after compounds yielding desired effects (e.g., synthetic lethality in specific cancer genetic backgrounds) are discovered through the phenotypic screening, it is non-trivial to identify the mechanism of action of compounds. Most small molecules, particularly those with higher lipophilicity and lower molecular weight, bind to more than one protein, and yet targets and downstream mechanisms of most compounds remain largely elusive (Hu et al. 2015).


Ideally, one would like to systematically define in a screening experiment which cellular mechanisms are responsible for the effect. However, this requires that relevant targets and mechanisms be defined for all screening compounds, which is not practical currently. However, to compensate for this limitation, chemoinformatic efforts to predict binding targets of small molecules have been made. Similarity Ensemble Approach (SEA) is one such algorithm that compares the structure of a query compound with diverse collection of small molecule agonists and antagonists of each protein in a database, assembled from previous assays n (Keiser et al. 2007). The analysis computes a likelihood of the query molecule targeting each protein. While it is still at its earlier stage and there is a room for improvement, SEA has been utilized to investigate clinically available drugs, and multiple functionally relevant targets of them were successfully identified previously (Keiser et al. 2007; Keiser et al. 2009; Gregori-Puigjané et al. 2012).


In this study, we aimed to test whether high-throughput screening of a compound library could be used to define the molecular mechanism of a probe compound; we propose an analytic framework termed ‘target enrichment analysis (TEA). TEA integrates phenotypic screening of a chemical library with ligand-based target predictions using SEA (FIG. 21). The goal of TEA is to improve the predictions of SEA based on experimental data. TEA attempts to find the common target(s) of compounds that are discovered to be effective in chemical library screening. TEA prioritizes ligand-target relationships initially given by SEA to suggest which ligand-target predictions are more likely to be true-positives that explain the signaling event inside the phenomenon of interest. Therefore, predictions given by TEA are more likely to be functionally meaningful than those given by SEA.


The TEA workflow consists of four steps: (i) performing cell-based high-throughput screening using a chemical library, (ii) predicting target proteins using SEA for each of the screening compounds, and (iii) integrating data acquired in the previous steps, and identifying protein targets over-represented among hit compounds from the first step that induce the desired phenotype using one sample Kolmogorov-Smirnov (KS) test. This is analogous to assess whether a predefined set of genes are over-/under-expressed among a data set in Gene Set Enrichment Analysis (GSEA) (Subramanian et al. 2005); the algorithm used here assesses whether a predefined set of compounds assigned to a target are overrepresented among hit compounds of a phenotypic assay. However, this does not immediately give us proteins targeted by the hit compounds because similar sets of compounds often bind to multiple proteins, e.g., ones in the same family. The algorithm cannot distinguish proteins that are more likely to be responsible for the phenomena without further experimental perturbation. However, at least to highlight such redundancy, (iv) the algorithm tests for significant overlap of between compound sets targeting each proteins. The third and fourth steps of TEA algorithm should pick ligand-target predictions more reliable and functionally relevant to the phenotype of interest, among ones predicted by SEA that are generally highly false positive.


As a proof of principle, we applied TEA to seek for the mechanism of action of CIL56, a multimodal lethal compound, inducing both ferroptosis and non-apoptotic non-ferroptotic cell death (Shimada et al. 2016). Ferroptosis occurs in cells when glutathione peroxidase 4 (GPX4) is inhibited and lipid peroxides accumulate to lethal levels in cells. Lipophilic antioxidants and iron chelators suppress ferroptotic cell death (Shimada et al. 2016). CIL56 was initially identified as a ferroptosis inducer, because antioxidants and iron chelators suppress its lethality up to 10-fold in ferroptosis-sensitive HT-1080 cells. However, the same ferroptosis suppressors inhibit other ferroptosis inducers completely, suggesting that CIL56 induces ferroptosis as well as a mechanistically distinct cell death at higher concentrations. We termed this secondary cell death necrosis for the sake of convenience, because CIL56 was found not to induce apoptosis but completely uncharacterized otherwise (Shimada et al. 2016). We later created a structurally similar molecule FIN56 that induces only ferroptosis, but not necrosis, supporting the notion that CIL56 induces both ferroptosis and necrosis (Shimada et al. 2016) (FIG. 22).


A molecule inducing more than one phenotype is generally challenging to study, because it is potentially confusing as to which phenotype is relevant to detected molecular changes. While our exploration of structural analogs of CIL56 eventually discovered a selective ferroptosis inducer FIN56, this selective molecule was not available at the time the screening for this study was performed. Therefore, we studied CIL56-induced cell death (Dixon et al. 2015; Viswanathan et al. 2017) using TEA.


To distinguish ferroptosis from necrosis induced by CIL56, we treated HT-1080 cells under two different conditions: “CIL56 only (C)” or “concomitant treatment of CIL56 and α-tocopherol (C+A)”. The former induces both ferroptosis at lower concentrations and necrosis at higher concentrations while the latter induces only necrosis because a lipophilic antioxidant α-tocopherol completely suppresses ferroptosis, according to the previous study of FIN56 (Shimada et al. 2016).


In the first step of TEA, we screened for enhancers and suppressors of CIL56-induced cell death. We seeded 1,000 HT-1080 cells in 384 well plates, and immediately treated them with a lethal stimulus (either C or C+A) and one of 2,000 death modulators from the MicroSource bioactive compound library. Cells were incubated for 48 h before adding AlamarBlue (Invitrogen) and fluorescence (488/535) was measured on a Victor3 plate reader (PerkinElmer).


Next, we computationally predicted target proteins of each of 2,000 death modulators using SEA. Reference compound sets were extracted from chemical library screening experiments deposited in ChEMBL (Gaulton et al. 2012). First, all compounds with binding activity data for any better than or smaller to 10 μM were extracted and a compound set was associated with each target. Second, starting with the same compound sets, all ligands from orthologous eukaryotic targets were merged and clustered using Ward's clustering method (Ward 1963) implementation by ChemAxon. SEA was used as a ligand-based method to calculate the similarity between the query compounds and the different reference sets of ligands (Keiser et al. 2007). This method uses SciTegic's topological extended connectivity fingerprints (ECFP_4) (Rogers and Hahn 2010) to mathematically describe each molecule. To compare two sets of compounds, all pairwise Tanimoto similarities were calculated for the molecules in each set and those above a predetermined similarity threshold are summed in a raw score. This raw score was transformed into a Z-score based on the standard deviation calculated on a randomized background distribution. An expectation value (E-value) is calculated for this Z-score using a statistical technique analogous to the one used by the basic local alignment search tool (BLAST) (Altschul et al. 1990), calculating the probability of observing a given score by random chance alone. Predictions whose E-values were less than 1 were used for predicted ligand-target relationships in TEA. Of 2,000 death modulators, 1,482 were predicted to target 512 proteins (data not shown).


Third, we sorted the 1,482 death modulator treatments based on their effects on each lethal stimulus (i.e., suppressing most to enhancing most), and performed a one-sample KS test to assess overrepresentation of each compound set among enhancers or suppressors per protein target. We identified proteins that may (a) enhance ferroptosis, (b) suppress ferroptosis, (c) enhance necrosis, or (d) suppress necrosis (FIG. 22).


Lastly, we computed overlaps of compounds among the predicted protein targets. This revealed sets of proteins, such that the same compounds were predicted to perturb each set of proteins. Through TEA, we predicted that three non-overlapping compound sets targets nine potential target proteins to perturb CIL56-induced ferroptosis or necrosis (Table 10).









TABLE 10







Three distinct sets of CIL56 enhancer/suppressors and their predicted targets.



















Expected


Compound
Predicted



OI within
effect on


set
target
Protein name
Ferroptosis
Necrosis
the set
ferroptosis





1
MMP1
Matrix
Not
Enhancer
0.9-1
Not




Metalloproteinase 1
available.*1


available.*1



NR3C1
Glucocorticoid








Receptor







NR3C2
Mineralocorticoid








Receptor







PGR
Progesterone








Receptor






2
ADRA1B
Adrenoreceptor a1B
Suppressor
Suppressor
1
Modulation



ADRA1D
Adrenoreceptor a1D



of Ca2+



CALM3
Calmodulin 3



signaling


3
ALOX12
Arachidonate
Suppressor
No effect
0.6
ALOXs




Lipoxygenase 12



produce



ALOX15
Arachidonate



lipid ROS




Lipoxygenase 15





*1Necrotic cell death was enhaced by cmpd set #1, and became more potent than ferroptotic cell death.






The first compound set we examined was enhancers of necrosis. Because necrosis was more potentiated than ferroptosis, we could not tell if ferroptosis was affected from this analysis. These compounds were predicted to bind to matrix metalloprotease 1 (MMP1) and/or nuclear receptors (NR3C1, MR3C2, PGR).


The second compound set examined was suppressors of ferroptosis that have no effect on necrosis; these were predicted to target two proteins: arachidonate lipoxygenases (ALOX), ALOX12 and ALOX15. ALOXs peroxidize poly-unsaturated fatty acids such as arachidonic acids, whose products are lipid signaling molecules mediating inflammatory signaling. While there are five ALOXs in humans (ALOX5, ALOX12, ALOX12B, ALOX15 and ALOX15B), only three were used in SEA's ligand-target predictions (ALOX5, ALOX12, and ALOX15). Whichever ALOX species may be genuinely relevant to CIL56-induced ferroptosis, ALOX proteins are suggested to be involved in some form of ferroptosis (Yang et al. 2016; Shintoku et al. 2017).


The third compound set examined was suppressors of both ferroptosis and necrosis. These were predicted to target three proteins: alpha-1 (α1) adrenergic receptors 1B and 1D and calmodulin 3. Activation of α1 activates phospholipase C and increases Ca(II). Calmoduline 3 is a calcium-binding protein. Since all three proteins are relevant to calcium signaling, we hypothesized that calcium mediates FIN56-induced ferroptosis. Flow cytometry with a Fluo-4 Ca(II) indicator showed that 2 μM FIN56 treatment increased intracellular Ca(II) levels in HT-1080 cells over the course of 9 h. Moreover, intracellular calcium induction with treatment with two ionophores (1 μM ionomycin, 0.25 μM A23187) or a sodium-potassium pump inhibitor (0.5 μM digoxin) enhanced FIN56 lethality, while Ca(II) chelators (2 μM EGTA or 10 μM BAPTA) suppressed it. These data indicated that Ca(II) plays a critical role in FIN56-induced ferroptosis (FIGS. 23A-23B).


Involvement of Ca(II) and ALOX in cell death mechanism points to glutamate-induced toxicity in neuronal cells, known as oxytosis (Tobaben et al. 2011; Henke et al. 2013), High concentrations of glutamate inhibit the function of the cystine-glutamate antiporter (system xc), reduces the enzymatic activity of GPX4 and consequently induces massive lipophilic reactive oxygen species generation. FIN56 does not inhibit system xc (Shimada et al. 2016).


In summary, TEA predicted some aspects of ferroptosis, that is the involvement of ALOX and Ca(II) for progression of ferroptotic cell death. Whether the predicted individual genes are truly involved in the mechanism of action further require single or combination of functional gene deletion. On the other hand, CIL56-induced necrosis is still much elusive. While a few enhancer molecules and predicted targets against the necrosis were discovered, it was not so beneficial towards understanding the mechanism as suppressor molecules, since synergism of lethality can occur more unexpectedly. There were only a few studies that explored the mechanism of action of CIL56-induced necrosis phenotype (Dixon et al. 2015), however, one of them performed a haploid cell screening system may be relevant; they discovered several genes whose deletion rescued cells from CIL56-induced lethality (ZDHHCS, TECR, ACACA, NADK) (Dixon et al. 2015). Because deletion of the genes made cells resistant to the concentration that induces both necrosis and ferroptosis in multiple cell lines (5.5 μM), these genes' functions may be involved in progression of both cell death phenotypes.


TEA can serve as a compensatory approach to other technologies. In most phenotypic screening experiments, only a handful of hit compounds were further studied but the rest of the information is discarded. By integrating with ligand-target prediction such as SEA, we could identify the mechanisms of action of the hit molecules simultaneously. However, one needs to be cautious using such predictions; no ligand-based target prediction algorithms including SEA is a self-contained technology yet, and experimental validation needs to follow. But by collecting reference datasets (ChEMBL) and improvement prediction algorithms (SEA), phentotypic screening of compound library should better serve as a routine to understand the molecular mechanisms.


The embodiments described in this disclosure can be combined in various ways. Any aspect or feature that is described for one embodiment can be incorporated into any other embodiment mentioned in this disclosure. While various novel features of the inventive principles have been shown, described and pointed out as applied to particular embodiments thereof, it should be understood that various omissions and substitutions and changes may be made by those skilled in the art without departing from the spirit of this disclosure. Those skilled in the art will appreciate that the inventive principles can be practiced in other than the described embodiments, which are presented for purposes of illustration and not limitation.


DOCUMENTS CITED



  • Alegre-Aguarón, E. et al. Growth factor priming differentially modulates components of the extracellular matrix proteome in chondrocytes and synovium-derived stem cells. PLoS One 9, e88053 (2014).

  • Altschul, S. F.; Gish, W.; Miller, W.; Myers, E. W.; Lipman, D. J. Basic Local Alignment Search Tool. J. Mol. Biol. 1990, 215 (3), 403-410.http://doi.org/10.1016/S0022-2836(05)80360-2.

  • Aravind, L., Dixit, V. M. & Koonin, E. V. The domains of death: evolution of the apoptosis machinery. Trends Biochem. Sci. 24, 47-53 (1999).

  • Backman, T. W. H., Cao, Y. & Girke, T. ChemMine tools: an online service for analyzing and clustering small molecules. Nucleic Acids Res. 39, W486-W491 (2011).

  • Berghe T V, Linkermann A, Jouan-Lanhouet S, Walczak H, Vandenabeele P. Regulated necrosis: the expanding network of non-apoptotic cell death pathways. Nat. Rev. Mol. Cell Biol. 2014; 15:135-147.

  • Cholody, W. M. et al. Derivatives of fluorene, anthracene, xanthene, dibenzosuberone and acridine and uses thereof. US patent application PCT/US2008/006015 (2008).

  • Chugh, A., Ray, A. & Gupta, J. B. Squalene epoxidase as hypocholesterolemic drug target revisited. Prog. Lipid Res. 42, 37-50 (2003).

  • Degterev, A. et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat. Chem. Biol. 1, 112-119 (2005).

  • Degterev, A. et al. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat. Chem. Biol. 4, 313-321 (2008).

  • Dixon S J, et al. Ferroptosis: An Iron-Dependent Form of Nonapoptotic Cell Death. Cell. 2012; 149:1060-1072.

  • Dixon S J, et al. Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis. eLife. 2014; 3:e02523.

  • Dixon, S. J. et al. Human haploid cell genetics reveals roles for lipid metabolism genes in nonapoptotic cell death. ACS Chem. Biol. 10, 1604-1609 (2015).

  • Fradejas, N. et al. Mammalian Trit1 is a tRNA([Ser]Sec)-isopentenyl transferase required for full selenoprotein expression. Biochem. J. 450, 427-432 (2013).

  • Fuchs, Y. & Steller, H. Programmed cell death in animal development and disease. Cell 147, 742-758 (2011).

  • Gaulton, A.; Bellis, L. J.; Bento, A. P.; Chambers, J.; Davies, M.; Hersey, A.; Light, Y.; McGlinchey, S.; Michalovich, D.; Al-Lazikani, B.; et al. ChEMBL: A Large-Scale Bioactivity Database for Drug Discovery. Nucleic Acids Res. 2012, 40 (D1), D1100-D1107. http://doi.org/10.1093/nar/gkr777.

  • Gregori-Puigjané, E.; Setola, V.; Hert, J.; Crews, B. A.; Irwin, J. J.; Lounkine, E.; Marnett, L.; Roth, B. L.; Shoichet, B. K. Identifying Mechanism-of-Action Targets for Drugs and Probes. Proc. Natl. Acad. Sci. 2012, 109 (28), 11178-11183. https://doi.org/10.1073/pnas.1204524109.

  • Gueven, N., Woolley, K. & Smith, J. Border between natural product and drug: comparison of the related benzoquinones idebenone and coenzyme Q10. Redox Biol. 4, 289-295 (2015).

  • Hahn, W. C. et al. Creation of human tumour cells with defined genetic elements. Nature 400, 464-468 (1999).

  • Hayano, M., Yang, W. S., Corn, C. K., Pagano, N. C. & Stockwell, B. R. Loss of cysteinyl-tRNA synthetase (CARS) induces the transsulfuration pathway and inhibits ferroptosis induced by cystine deprivation. Cell Death Differ. 23, 270-278 (2016).

  • Henke, N.; Albrecht, P.; Bouchachia, I.; Ryazantseva, M.; Knoll, K.; Lewerenz, J.; Kaznacheyeva, E.; Maher, P.; Methner, A. The Plasma Membrane Channel ORAI1 Mediates Detrimental Calcium Influx Caused by Endogenous Oxidative Stress. Cell Death Dis. 2013, 4 (1), e470. https://doi.org/10.1038/cddis.2012.216.

  • Hirsch, H. A. et al. A transcriptional signature and common gene networks link cancer with lipid metabolism and diverse human diseases. Cancer Cell 17, 348-361 (2010).

  • Hitomi, J. et al. Identification of a molecular signaling network that regulates a cellular necrotic cell death pathway. Cell 135, 1311-1323 (2008).

  • Hu, Y.; Jasial, S.; Bajorath, J. Promiscuity Progression of Bioactive Compounds over Time. F1000Research 2015. https://doi.org/10.12688/f1000research.6473.2.

  • Imai, H. & Nakagawa, Y. Biological significance of phospholipid hydroperoxide glutathione peroxidase (PHGPx, GPx4) in mammalian cells. Free Radic. Biol. Med. 34, 145-169 (2003).

  • Kaczmarek, A., Vandenabeele, P. & Krysko, D. V. Necroptosis: the release of damage-associated molecular patterns and its physiological relevance. Immunity 38, 209-223 (2013).

  • Kamphorst, J. J., Fan, J., Lu, W., White, E. & Rabinowitz, J. D. Liquid chromatography-high resolution mass spectrometry analysis of fatty acid metabolism. Anal. Chem. 83, 9114-9122 (2011).

  • Keiser, M. J.; Roth, B. L.; Armbruster, B. N.; Ernsberger, P.; Irwin, J. J.; Shoichet, B. K. Relating Protein Pharmacology by Ligand Chemistry. Nat. Biotechnol. 2007, 25 (2), 197-206. https://doi.org/10.1038/nbt1284.

  • Keiser, M. J.; Setola, V.; Irwin, J. J.; Laggner, C.; Abbas, A. I.; Hufeisen, S. J.; Jensen, N. H.; Kuijer, M. B.; Matos, R. C.; Tran, T. B.; et al. Predicting New Molecular Targets for Known Drugs. Nature 2009, 462 (7270), 175-181. https://doi.org/10.1038/nature08506.



Kono, H. & Rock, K. L. How dying cells alert the immune system to danger. Nat. Rev. Immunol. 8, 279-289 (2008).

  • Linkermann, A. et al. Synchronized renal tubular cell death involves ferroptosis. Proc. Natl. Acad. Sci. USA 111, 16836-16841 (2014).
  • Linkermann, A., Stockwell, B. R., Krautwald, S. & Anders, H.-J. Regulated cell death and inflammation: an auto-amplification loop causes organ failure. Nat. Rev. Immunol. 14, 759-767 (2014).
  • Liu, C.-I. et al. Structural insights into the catalytic mechanism of human squalene synthase. Acta Crystallogr. D Biol. Crystallogr. 70, 231-241 (2014).
  • Rogers, D.; Hahn, M. Extended-Connectivity Fingerprints. J. Chem. Inf. Model. 2010, 50 (5), 742-754. http://doi.org/10.1021/ci100050t.
  • Romanowska, M. et al. Effects of selenium supplementation on expression of glutathione peroxidase isoforms in cultured human lung adenocarcinoma cell lines. Lung Cancer 55, 35-42 (2007).
  • Santos, C. R. & Schulze, A. Lipid metabolism in cancer. FEBS J. 279, 2610-2623 (2012).
  • Shimada, K. et al. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat. Chem. Biol. 2016 July; 12(7):497-503.
  • Shimada, K., Hayano, M., Pagano, N. C. & Stockwell, B. R. Cell-line selectivity improves the predictive power of pharmacogenomic analyses and helps identify NADPH as biomarker for ferroptosis sensitivity. Cell Chem. Biol. 23, 225-235 (2016).
  • Shintoku, R.; Takigawa, Y.; Yamada, K.; Kubota, C.; Yoshimoto, Y.; Takeuchi, T.; Koshiishi, I.; Torii, S. Lipoxygenase-mediated Generation of Lipid Peroxides Enhances Ferroptosis Induced by Erastin and RSL3. Cancer Sci. 2017, 108 (11), 2187-2194. https://doi.org/10.1111/cas.13380.
  • Shoemaker, R. H. The NCI60 human tumour cell line anticancer drug screen. Nat. Rev. Cancer 6, 813-823 (2006).
  • Skouta, R. et al. Ferrostatins inhibit oxidative lipid damage and cell death in diverse disease models. J. Am. Chem. Soc. 136, 4551-4556 (2014).
  • Song, J. H. et al. Deletion of Pim kinases elevates the cellular levels of reactive oxygen species and sensitizes to K-Ras-induced cell killing. Oncogene 34, 3728-3736 (2015).
  • Subramanian, A.; Tamayo, P.; Mootha, V. K.; Mukherjee, S.; Ebert, B. L.; Gillette, M. A.; Paulovich, A.; Pomeroy, S. L.; Golub, T. R.; Lander, E. S.; et al. Gene Set Enrichment Analysis: A Knowledge-Based Approach for Interpreting Genome-Wide Expression Profiles. Proc. Natl. Acad. Sci. U.S.A. 2005, 102 (43), 15545-15550.
  • Takahashi, N. et al. Necrostatin-1 analogues: critical issues on the specificity, activity and in vivo use in experimental disease models. Cell Death Dis. 3, e437 (2012).
  • Tansey, T. R. & Shechter, I. Structure and regulation of mammalian squalene synthase. Biochim. Biophys. Acta 1529, 49-62 (2000).
  • Tobaben, S.; Grohm, J.; Seiler, A.; Conrad, M.; Plesnila, N.; Culmsee, C. Bid-Mediated Mitochondrial Damage Is a Key Mechanism in Glutamate-Induced Oxidative Stress and AIF-Dependent Cell Death in Immortalized HT-22 Hippocampal Neurons. Cell Death Differ. 2011, 18 (2), 282-292. httos://doi.org/10.1038/cdd.2010.92.
  • Vanden Berghe, T., Linkermann, A., Jouan-Lanhouet, S., Walczak, H. & Vandenabeele, P. Regulated necrosis: the expanding network of non-apoptotic cell death pathways. Nat. Rev. Mol. Cell Biol. 15, 135-147 (2014).
  • Viswanathan, V. S.; Ryan, M. J.; Dhruv, H. D.; Gill, S.; Eichhoff, O. M.; Seashore-Ludlow, B.; Kaffenberger, S. D.; Eaton, J. K.; Shimada, K.; Aguirre, A. J.; et al. Dependency of a Therapy-Resistant State of Cancer Cells on a Lipid Peroxidase Pathway. Nature 2017, 547 (7664), 453. https://doi.org/10.1038/nature23007.
  • Ward, J. H. Hierarchical Grouping to Optimize an Objective Function. J. Am. Stat. Assoc. 1963, 58 (301), 236-244. https://doi.org/10.1080/01621459.1963.10500845.
  • Wolpaw, A. J. et al. Modulatory profiling identifies mechanisms of small molecule-induced cell death. Proc. Natl. Acad. Sci. USA 108, E771-E780 (2011).
  • Yagoda, N. et al. RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature 447, 864-868 (2007).
  • Yang, W. S.; Kim, K. J.; Gaschler, M. M.; Patel, M.; Shchepinov, M. S.; Stockwell, B. R. Peroxidation of Polyunsaturated Fatty Acids by Lipoxygenases Drives Ferroptosis. Proc. Natl. Acad. Sci. 2016, 113 (34), E4966-E4975. https://doi.org/10.1073/pnas.1603244113.
  • Yang, W. S. & Stockwell, B. R. Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RASharboring cancer cells. Chem. Biol. 15, 234-245 (2008).
  • Yang, W. S. et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317-331 (2014).

Claims
  • 1. A compound having the structure of formula (I):
  • 2. A compound having the structure of formula (II):
  • 3. A compound according to claim 2, which is selected from the group consisting of:
  • 4. A composition comprising a compound according to any one of claims 1-3, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • 5. A pharmaceutically acceptable salt of a compound according to any one of claims 1-3.
  • 6. A composition comprising a pharmaceutically acceptable salt of a compound according to any one of claims 1-3 and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • 7. A method for inducing ferroptosis in a cell comprising contacting the cell with an effective amount of a compound having the structure of formula (I):
  • 8. A method for inducing ferroptosis in a cell comprising contacting the cell with an effective amount of a compound having the structure of formula (II):
  • 9. A method for inducing ferroptosis in a cell comprising contacting the cell with an effective amount of a compound selected from the group consisting of:
  • 10. The method according to claim 7, wherein the compound is FIN56:
  • 11. The method according to any one of claims 7-10, wherein the cell is a mammalian cell.
  • 12. The method according to claim 11, wherein the cell is a human cell.
  • 13. The method according to any one of claims 7-10, wherein the cell is from a laboratory animal.
  • 14. The method according to any one of claims 7-10, which is carried out in vitro, or ex vivo.
  • 15. A method for decreasing GPX4 in a cell, comprising contacting the cell with an effective amount of a compound having the structure of formula (I):
  • 16. A method for decreasing GPX4 in a cell, comprising contacting the cell with an effective amount of a compound having the structure of formula (II):
  • 17. A method for decreasing GPX4 in a cell, comprising contacting the cell with an effective amount of a compound selected from the group consisting of:
  • 18. The method according to claim 15, wherein the compound is FIN56:
  • 19. The method according to any one of claims 15-18, wherein the cell is a mammalian cell.
CROSS-REFERENCE TO RELATED APPLICATIONS

The present application is a continuation in part of PCT international application No. PCT/US2017/066822, filed Dec. 15, 2017, which claims priority to U.S. Provisional Patent Application No. 62/436,064, filed on Dec. 19, 2016. The entire contents of the aforementioned applications are incorporated by reference as if recited in full herein.

GOVERNMENT FUNDING

This invention was made with government support under grant no. CA097061, awarded by the National Institutes of Health. The government has certain rights in the invention.

US Referenced Citations (1)
Number Name Date Kind
20100137354 Cholody et al. Jun 2010 A1
Foreign Referenced Citations (2)
Number Date Country
2008140792 Sep 2008 WO
2010082912 Jul 2010 WO
Non-Patent Literature Citations (54)
Entry
Ward, J. H. “Hierarchical Grouping to Optimize an Objective Function,” J. Am. Stat. Assoc. 1963, 58 (301), 236-244. https://doi.org/10.1080/01621459.1963.10500845.
Wolpaw, et al. “Modulatory profiling identifies mechanisms of small molecule-induced cell death,” Proc. Natl. Acad. Sci. USA 108, E771-E780 (2011).
Yagoda, et al. “RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels,” Nature 447, 864-868 (2007).
Yang, et al. “Peroxidation of Polyunsaturated Fatty Acids by Lipoxygenases Drives Ferroptosis,” Proc. Natl. Acad. Sci. 2016, 113 (34), E4966-E4975. https://doi.org/10.1073/pnas.1603244113.
Shimada, et al. “Global Survey of Cell Death Mechanisms Reveals Metabolic Regulation of Ferroptosis,” Nat Chem Biol. Jul. 2016;12(7):497-503.
Pubchem, Compound Summary for SID 173334080, National Center for Biotechnology Information. PubChem Database. REGID_for_CID_3097937, Source=Meiler Lab, Vanderbilt University, SID=173334080, https://pubchem.ncbi.nlm.nih.gov/substance7173334080 (accessed on Jan. 24, 2020).
International Search Report for PCT/US2017/066822 dated Apr. 13, 2018.
Alegre-Aguaron, E. et al. “Growth factor priming differentially modulates components of the extracellular matrix proteome in chondrocytes and synovium-derived stem cells.” PLoS One 9, e88053 (2014).
Altschul, et al. “Basic Local Alignment Search Tool,” J. Mol. Biol. 1990, 215 (3), 403-410. httpildoi.org/10.1016/S0022-2836(05)80360-2.
Aravind, et a. “The domains of death: evolution of the apoptosis machinery,” Trends Biochem. Sci. 24, 47-53 (1999).
Backman, et al. “ChemMine tools: an online service for analyzing and clustering small molecules,” Nucleic Acids Res. 39, W486—W491 (2011).
Berghe, et al. “Regulated necrosis: the expanding network of non-apoptotic cell death pathways,” Nat. Rev. Mol. Cell Biol. 2014;15:135-147.
Yang, et al. “Synthetic lethal screening identities compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells,” Chem. Biol. 15, 234-245 (2008).
Dhugh, et al. “Squalene epoxidase as hypocholesterolemic drug target revisited,” Prog. Lipid Res. 42, 37-50 (2003).
Degterev, et al. “Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury,” Nat. Chem. Biol. 1, 112-119 (2005).
Degterev, et al. “Identification of RIP1 kinase as a specific cellular target of necrostatins,” Nat. Chem. Biol. 4, 313-321 (2008).
Dixon, et al. “Ferroptosis: An Iron-Dependent Form of Nonapoptotic Cell Death,” Cell. 2012;149:1060-1072.
Dixon, et al. “Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis,” eLife. 2014;3:e02523.
Dixon, et al. “Human haploid cell genetics reveals roles for lipid metabolism genes in nonapoptotic cell death,” ACS Chem. Biol. 10, 1604-1609 (2015).
Fradejas, et al. “Mammalian Trit1 is a tRNA([Ser]Sec)-isopentenyl transferase required for full selenoprotein expression,” Biochem. J. 450, 427-432 (2013).
Fuchs, et al. “Programmed cell death in animal development and disease,” Cell 147, 742-758 (2011).
Gaulton, et al. “ChEMBL: A Large-Scale Bioactivity Database for Drug Discovery,” Nucleic Acids Res. 2012, 40 (D1), D1100-D1107. http://doi.org/10.1093/nar/gkr777.
Gregori-Puigjane, et a. “Identifying Mechanism-of-Action Targets for Drugs and Probes,” Proc. Natl. Acad. Sci. 2012, 109 (28), 11178-11183. https://doi.org/10.1073/pnas.1204524109.
Gueven, et al. “Border between natural product and drug: comparison of the related benzoquinones idebenone and coenzyme Q10,” Redox Biol. 4, 289-295 (2015).
Hahn, et al. “Creation of human tumour cells with defined genetic elements,” Nature 400, 464-468 (1999).
Hayano, et al. “Loss of cysteinyl-tRNA synthetase (CARS) induces the transsulfuration pathway and inhibits ferroptosis induced by cystine deprivation,” Cell Death Differ 23, 270-278 (2016).
Henke, et al. “The Plasma Membrane Channel ORAl1 Mediates Detrimental Calcium Influx Caused by Endogenous Oxidative Stress,” Cell Death Dis. 2013, 4 (1), e470. https://doi.org/10.1038/cddis.2012.216.
Hirsch, et al. “A transcriptional signature and common gene networks link cancer with lipid metabolism and diverse human diseases,” Cancer Cell 17, 348-361 (2010).
Hitomi, et al. “Identification of a molecular signaling network that regulates a cellular necrotic cell death pathway,” Cell 135, 1311-1323 (2008).
Hu, et al. “Promiscuity Progression of Bioactive Compounds over Time,” F1000Research 2015 https://doi.org/10.12688/f1000research.6473.2.
Imai, et al. “Biological significance of phospholipid hydroperoxide glutathione peroxidase (PHGPx, GPx4) in mammalian cells,” Free Radic. Biol. Med. 34, 145-169 (2003).
Kaczmarek, et al. “Necroptosis: the release of damage-associated molecular patterns and its physiological relevance,” Immunity 38, 209-223 (2013).
Kamphorst, et al. “Liquid chromatography-high resolution mass spectrometry analysis of fatty acid metabolism,” Anal. Chem. 83, 9114-9122 (2011).
Keiser, et al. “Relating Protein Pharmacology by Ligand Chemistry,” Nat. Biotechnol. 2007, 25 (2), 197-206. https://doi.org/10.1038/nbt1284.
Keiser, et al. “Predicting New Molecular Targets for Known Drugs,” Nature 2009, 462 (7270), 175-181.
Kono, et al. “How dying cells alert the immune system to danger,” Nat. Rev. Immunol. 8, 279-289 (2008).
Linkermann, et al. “Synchronized renal tubular cell death involves ferroptosis,” Proc. Natl. Acad. Sci. USA 111, 16836-16841 (2014).
Linkermann, et al. “Regulated cell death and inflammation: an auto-amplification loop causes organ failure,” Nat. Rev. Immunol. 14, 759-767 (2014).
Liu, et al. “Structural insights into the catalytic mechanism of human squalene synthase,” Acta Crystallogr. D Biol. Crystallogr. 70, 231-241 (2014).
Rogers, et al. “Extended-Connectivity Fingerprints,” J. Chem. Inf. Model. 2010, 50 (5), 742-754. https://doi.org/10.1021/ci100050t.
Romanowska, et al. “Effects of selenium supplementation on expression of glutathione peroxidase isoforms in cultured human lung adenocarcinoma cell lines,” Lung Cancer 55, 35-42 (2007).
Santos, et al. “Lipid metabolism in cancer,” FEBS J. 279, 2610-2623 (2012).
Yang, et al. “Regulation of ferroptotic cancer cell death by GPX4,” Cell 156, 317-331 (2014).
Shimada, et al. “Cell-line selectivity improves the predictive power of pharmacogenomic analyses and helps identify NADPH as biomarker for ferroptosis sensitivity,” Cell Chem. Biol. 23, 225-235 (2016).
Shintoku et al. “Lipoxygenase-mediated Generation of Lipid Peroxides Enhances Ferroptosis Induced by Erastin and RSL3,” Cancer Sci. 2017, 108 (11), 2187-2194. https://doi.org/10.1111/cas.13380.
Shoemaker, R.N. “The NCI60 human tumour cell line anticancer drug screen,” Nat. Rev. Cancer 6, 813-823 (2006).
Skouta, et al. “Ferrostatins inhibit oxidative lipid damage and cell death in diverse disease models,” J. Am. Chem. Soc. 136, 4551-4556 (2014).
Song, et al. “Deletion of Pim kinases elevates the cellular levels of reactive oxygen species and sensitizes to K-Ras-induced cell killing,” Oncogene 34, 3728-3736 (2015).
Subramanian, et al. “Gene Set Enrichment Analysis: A Knowledge-Based Approach for Interpreting Genome-Wide Expression Profiles,” Proc. Natl. Acad. Sci. U. S. A. 2005, 102 (43), 15545-15550. https://doi.org/10.1073/pnas.0506580102.
Takahashi, et al. “Necrostatin-1 analogues: critical issues on the specificity, activity and in vivo use in experimental lisease models,” Cell Death Dis. 3, e437 (2012).
Tansey, et al. “Structure and regulation of mammalian squalene synthase,” Biochim. Biophys. Acta 1529, 49-62 (2000).
Tobaben, et al. “Bid-Mediated Mitochondrial Damage Is a Key Mechanism in Glutamate-Induced Oxidative Stress and AIF-Dependent Cell Death in Immortalized HT-22 Hippocampal Neurons, ” Cell Death Differ. 2011, 18 (2), 282-292. https://doi.org/10.1038/cdd.2010.92.
Vanden Berghe, et al. “Regulated necrosis: the expanding network of non-apoptotic cell death pathways,” Nat. Rev. Mol. Cell Biol. 15, 135-147 (2014).
Viswanathan, et al. “Dependency of a Therapy-Resistant State of Cancer Cells on a Lipid Peroxidase Pathway,” Nature 2017, 547 (7664), 453. https://doi.org/10.1038/nature23007.
Related Publications (1)
Number Date Country
20190315681 A1 Oct 2019 US
Provisional Applications (1)
Number Date Country
62436064 Dec 2016 US
Continuation in Parts (1)
Number Date Country
Parent PCT/US2017/066822 Dec 2017 US
Child 16445104 US