Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
AN EIF4A INHIBITOR WITH A NOVEL MECHANISM OF ACTION
Document Type and Number:
WIPO Patent Application WO/2023/034813
Kind Code:
A1
Abstract:
The present invention features inhibitors of the elF4A enzyme. These inhibitors have a novel mechanism for inhibiting the elF4A enzyme by occupying a binding pocket within the elF4A RNA-binding groove, thereby perturbing RNA binding, blocking ATP hydrolysis, and, consequently, inhibiting RNA helicase activity. Thus, the compounds of the present invention are RNA-competitive, ATP-uncompetitive elF4A inhibitors that directly bind elF4A and inhibit the protein.

Inventors:
CHAPMAN ELI (US)
ZERIO CHRISTOPHER (US)
Application Number:
PCT/US2022/075686
Publication Date:
March 09, 2023
Filing Date:
August 30, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV ARIZONA (US)
International Classes:
A61K31/4709; A61K31/47; A61K31/472; C07D215/02; C07D307/04; C12N9/14
Domestic Patent References:
WO2015106272A12015-07-16
WO2017106367A12017-06-22
Foreign References:
US20140275224A12014-09-18
US20120196854A12012-08-02
Other References:
BOTTLEY ANDREW, PHILLIPS NICOLA M., WEBB THOMAS E., WILLIS ANNE E., SPRIGGS KEITH A.: "eIF4A Inhibition Allows Translational Regulation of mRNAs Encoding Proteins Involved in Alzheimer's Disease", PLOS ONE, vol. 5, no. 9, 28 September 2010 (2010-09-28), pages e13030, XP093043765, DOI: 10.1371/journal.pone.0013030
BORDELEAU, ME ET AL.: "Functional characterization of IRESes by an inhibitor of the RNA helicase eIF4A", NATURE CHEMICAL BIOLOGY, vol. 2, no. 4, April 2006 (2006-04-01), XP003005918, DOI: 10.1038/nchembio776
DATABASE PUBCHEM SUBSTANCE ANONYMOUS : "MCULE-4868052373", XP093043767, retrieved from PUBCHEM
DATABASE PUBCHEM SUBSTANCE ANONYMOUS : "SID 290061748", XP093043807, retrieved from PUBCHEM
Attorney, Agent or Firm:
NGUYEN, Quan (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. An elF4A inhibitor having a formula according to Formula I: wherein R is H, alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, alkoxy, nitro, or halo, wherein the elF4A inhibitor occupies a binding pocket of an elF4A RNA-binding groove, thereby perturbing RNA binding to elF4A, blocking ATP hydrolysis, and inhibiting elF4A from unwinding RNA.

2. The elF4A inhibitor of claim 1 having a formula according to Formula II: Formula II.

3. An elF4A inhibitor having a formula according to Formula III: wherein A^ is aryl, substituted aryl, heteroaryl, or substituted heteroaryl, wherein the elF4A inhibitor occupies a binding pocket of an elF4A RNA-binding groove, thereby perturbing RNA binding to elF4A, blocking ATP hydrolysis, and inhibiting elF4A from unwinding RNA.

4. An elF4A inhibitor having a formula according to Formula IV:

54 wherein R4 is aryl, substituted aryl, heteroaryl, or substituted aryl, wherein the elF4A inhibitor occupies a binding pocket of an elF4A RNA-binding groove, thereby perturbing RNA binding to elF4A, blocking ATP hydrolysis, and inhibiting elF4A from unwinding RNA. The elF4A inhibitor of claim 4 having a formula according to Formula V: Formula V wherein R R2J and R3 are each independently H, alkyl, substituted alkyl, alkoxy, aryl, or amino. The elF4A inhibitor of claim 4 having a formula according to Formula VI: Formula VI wherein R and R2 are each independently H, alkyl, substituted alkyl, alkoxy, aryl, or amino. A method for inhibiting elF4A activity in a cell, the method comprising: a. contacting the cell with an elF4A inhibitor having a formula according to Formula I:

55

wherein R is H, alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, alkoxy, nitro, or halo, wherein the elF4A inhibitor occupies a binding pocket of an elF4A RNA-binding groove, thereby perturbing RNA binding to elF4A, blocking ATP hydrolysis, and inhibiting elF4A from unwinding RNA. The method of claim 7, wherein the elF4A inhibitor has a formula according to Formula II: Formula II. A method for inhibiting elF4A activity in a cell, the method comprising: a. contacting the cell with an elF4A inhibitor having a formula according to Formula III: wherein A^ is aryl, substituted aryl, heteroaryl, or substituted heteroaryl, wherein the elF4A inhibitor occupies a binding pocket of an elF4A RNA-binding groove, thereby perturbing RNA binding to elF4A, blocking ATP hydrolysis, and inhibiting elF4A from unwinding RNA. A method for inhibiting elF4A activity in a cell, the method comprising:

56 a. contacting the cell with an elF4A inhibitor having a formula according to Formula III: wherein R4 is aryl, substituted aryl, heteroaryl, or substituted aryl, wherein the elF4A inhibitor occupies a binding pocket of an elF4A RNA-binding groove, thereby perturbing RNA binding to elF4A, blocking ATP hydrolysis, and inhibiting elF4A from unwinding RNA. The method of claim 10, wherein the elF4A inhibitor has a formula according to Formula V: Formula V wherein R R2, and R3 are each independently H, alkyl, substituted alkyl, alkoxy, aryl, or amino. The method of claim 10, wherein the elF4A inhibitor has a formula according to Formula VI: Formula VI wherein R and R2 are each independently H, alkyl, substituted alkyl, alkoxy, aryl, or amino.

Description:
AN EIF4A INHIBITOR WITH A NOVEL MECHANISM OF ACTION

CROSS-REFERENCES TO RELATED APPLICATIONS

[0001]This application claims benefit of U.S. Provisional Application No. 63/238,456 filed August 30, 2021, the specification of which is incorporated herein in its entirety by reference.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT

[0002] This invention was made with government support under Grant No. T32 GM008804 awarded by National Institutes of Health. The government has certain rights in the invention.

FIELD OF THE INVENTION

[0003]The present invention relates to inhibitors of the elF4A enzyme. In some embodiments, the compounds of the present invention may be useful for the treatment of lymphoma.

BACKGROUND OF THE INVENTION

[0004] Protein synthesis through messenger RNA (mRNA) translation is a strictly controlled process. Dysregulation of this operation is a core feature of cancer, as many pathways that are crucial for oncogenesis are mediated by translational control. The most tightly regulated step of protein biosynthesis is the initiation of cap-dependent translation, in which initiation factors bind to the 5-prime (5’) 7-methylguanosine (m 7 G) cap of mature mRNA to launch translation of open reading frames. Many oncogenes, including those involved in tumor cell proliferation, growth, and angiogenesis depend directly on m 7 G-cap factors for their translation. Conversely, translation of many non-oncogenic genes is cap-independent. Targeting specific cap-dependent translation factors that are altered in expression or activity in human cancers and not involved in translation of non-oncogenic housekeeping genes offers great promise for the development of a new generation of cancer therapeutics.

[0005] Cap-dependent translation is driven by the heterotri meric eukaryotic initiation factor (elF)4F complex, which catalyzes ribosome recruitment to mRNA and comprises elF4G, elF4E, and elF4A. elF4G is a scaffolding protein that positions elF4F for heterotrimeric engagement and initiation factor-RNA binding, elF4E binds the m 7 G mRNA cap, and elF4A is the enzymatic driver of the complex. elF4A is an ATP-dependent DEAD-box RNA helicase that binds and unwinds secondary structure in the 5’-untranslated region (UTR) of mature mRNA, allowing the ribosome access to translate downstream genes. Mammalian cells have three elF4A isoforms. elF4AI (referred to as elF4A throughout) and elF4AII are 90% homologous, located in the cytosol, and can both incorporate into the elF4F complex. However, elF4AI is ten times more abundant than elF4AII in growing cells, and elF4AII is unable to rescue inhibition of translation and cellular proliferation upon elF4AI suppression, suggesting that elF4AI and elF4AII are functionally distinct in vivo. elF4AIII is a component of the exon-junction complex in the nucleus and is not involved in protein synthesis.

[0006]mRNAs are translated at different rates due to heterogeneity of secondary structure in their 5’-UTRs, and elF4A discriminates between mRNAs based on 5’-UTR complexity. mRNAs with increased structure show increased dependence on elF4A for translation, whereas initiation of simple, unstructured mRNAs is less reliant on elF4A. Overexpression of elF4A elicits small changes in overall protein synthesis rates, but enables a large, disproportionate, and specific increase in translation of a subset of mRNAs. mRNA identification studies reveal that the production of housekeeping proteins such as beta-actin and GAPDH is not altered by changes in elF4A levels. Conversely, the translation of mRNAs encoding proteins involved in sustaining the hallmarks of cancer such as MYC, MCL1, BCL-xL, and cyclin-D1 , which harbor long, highly-structured 5’-UTRs, is notably impacted by fluctuating elF4A levels. Despite the essential nature of elF4A in all cells, the increased dependency of oncoprotein production on elF4A along with increased protein synthesis demands in cancer cells creates a therapeutic window in which cancer cells are more dependent on elF4A than healthy cells. As a result, effects of elF4A inhibition are biased toward cancer cells, which makes elF4A an intriguing target for cancer therapy.

[0007] Many cancer drugs target signaling proteins such as kinases, and although these drugs have been useful in cancer treatment, they commonly have short-lived clinical responses. This is a result of cancer cells bypassing many of these proteins through parallel or redundant signaling. This leads to resistance, as inhibition of one point in a pathway can easily be circumvented by upregulation of other pro-oncogenic factors. elF4A, however, is a convergence point for numerous oncogenic pathways, including Ras-Raf-ERK, EGFR, and PI3K-TOR. Therapies that target this junction for numerous different oncogenic pathways may overcome problems that hamper current cancer treatments such as tumor heterogeneity and inhibitor resistance.

[0008] Past in vitro screening for inhibitors of cap-dependent translation initiation using Krebs-2 translation extracts yielded natural products that all work by interfering with elF4A. These compounds include hippuristanol, pateamine A (PatA), and silvestrol (a rocaglate or “flavagline”). Flavagline analogs are widely considered the most promising elF4A inhibitors in the field, as eFT226 (Zotatifin), a rocaglamide-inspired inhibitor, has entered clinical trials for solid tumors (NCT04092673) as well as COVID-19 (NCT04632381). Previously, our group has also discovered natural product elF4A inhibitors, including elatol, which is believed to bind elF4A with 2:1 stoichiometry and shows potent elF4A inhibition in cells. Common weaknesses of natural product inhibitors are they can be difficult to work with due to limited supply, and medicinal chemistry can be challenging due to synthetic inaccessibility.

[0009] Herein, the present invention features a novel elF4A inhibitor with a unique mechanism of action and a novel binding pocket. In addition, the elF4A inhibitor is a synthetically tractable small molecule allowing for further optimization.

BRIEF SUMMARY OF THE INVENTION

[0010] It is an objective of the present invention to provide compositions and methods that allow for the inhibition of the elF4A enzyme, as specified in the independent claims. Embodiments of the invention are given in the dependent claims. Embodiments of the present invention can be freely combined with each other if they are not mutually exclusive.

[0011] In one embodiment, the present invention features an elF4A inhibitor having a formula according to Formula I: Formula I

[0012] In another embodiment, the present invention features an elF4A inhibitor of claim 1 having a formula according to Formula II: Formula II

[0013] In yet another embodiment, the present invention features an elF4A inhibitor having a formula according to Formula III: Formula III

[0014] In other embodiments, the present invention features elF4A inhibitors having formulas according to Formula IV, Formula V, or Formula VI: Formula IV

[0015] One of the unique and inventive technical features of the present invention is the novel mechanism of inhibiting elF4A. Without wishing to limit the invention to any theory or mechanism, it is believed that the technical feature of the present invention advantageously provides for a novel mechanism for inhibiting the elF4A enzyme. It was surprisingly found that the compounds described herein inhibit elF4A with a novel mechanism, in which they perturb RNA binding, block the hydrolysis of ATP, and consequently, inhibit RNA helicase activity. Thus, the compounds of the present invention are RNA-competitive, ATP-uncompetitive elF4A inhibitors that directly bind elF4A and inhibit the protein.

[0016] None of the presently known prior references or work has the unique inventive technical feature of the present invention. For example, Rocaglates inhibit elF4A by increasing the affinity of elF4A for RNA and clamping elF4A shut on RNA. PatA acts similarly, also stabilizing the elF4A-RNA interaction. In addition, PatA stimulates elF4A ATPase and helicase activities and inhibits the association of elF4A and elF4G, sequestering elF4A from the elF4F complex. The mechanism of elatol is not explicitly known, but its interaction with K82 of elF4A suggests it may be ATP-competitive. Hippuristanol is the only other RNA-competitive inhibitor of elF4A, but unlike the compounds of the present invention, it locks elF4A closed and inhibits elF4A unwinding activity by completely preventing RNA binding.

[0017] In addition, inhibition of elF4A through this novel mechanism may result in downstream cellular consequences different from those resulting from clamping RNA onto elF4A. This may allow for elucidation of additional pathways that are reliant on elF4A for translation of protein. The compounds of the present invention may also occupy a novel binding pocket within the elF4A RNA-binding groove. Rocaglates bind a small cavity that is encompassed by the N-terminal domain of elF4A and polypurine RNA. Desmethyl pateamine A (DMPatA), a PatA analog, occupies largely the same binding site as rocaglates, forming major interactions with RNA and the N-terminal domain of elF4A. However, unlike rocaglates, DMPatA also extends across the RNA to form minor interactions with the C-terminal domain of elF4A, and is able to clamp onto both polypurine and polypyrimidine RNA. Elatol binds elF4A in a 2:1 stoichiometric fashion, with both elatol molecules predicted to reside near the elF4A ATP pocket. Hippuristanol, although it shares an RNA-competitive mechanism with the compounds of the present invention, binds exclusively to the C-terminal domain of elF4A.

[0018]Any feature or combination of features described herein are included within the scope of the present invention provided that the features included in any such combination are not mutually inconsistent as will be apparent from the context, this specification, and the knowledge of one of ordinary skill in the art. Additional advantages and aspects of the present invention are apparent in the following detailed description and claims.

BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS

[0019] The features and advantages of the present invention will become apparent from a consideration of the following detailed description presented in connection with the accompanying drawings in which:

[0020] FIG. 1 shows a table of chemical structures of elF4A inhibitors of the present invention and results from the RNA-stimulated malachite green ATPase assay.

[0021] FIG. 2 shows a table of chemical structures with different substitutions at the central ring of the inhibitor scaffold and their results from the RNA-stimulated malachite green ATPase assay.

[0022] FIG. 3 shows a table of chemical structures of elF4A inhibitors containing substituted phenyl groups and results from the RNA-stimulated malachite green ATPase assay.

[0023] FIG. 4 shows a table of additional chemical structures of elF4A inhibitors with substituted phenyl groups and results from the RNA-stimulated malachite green ATPase assay.

[0024] FIGs. 5A-5E show the discovery and initial biochemical testing of compound 1 .

[0025] FIG. 5A shows RNA-accelerated malachite green ATPase assay screening data. Each black dot represents one compound. Green line indicates 3 standard deviations above the mean, which was the threshold used for hit determination (18 compounds). Initial hit 1 is circled in red.

[0026] FIG. 5B shows the chemical structure of compound 1.

[0027] FIG. 5C shows the dose response of compound 1 against elF4A. IC50 = 26.6 ± 2.46 pM. n=4.

[0028] FIG. 5D shows the kinetic testing of compound 1 using the malachite green assay. ATP titration (left) used 250 pg/mL RNA. Data were fit using unbiased mixed-model inhibition in Prism 6.0. Alpha value for ATP is 0.424, indicating an ATP-uncompetitive inhibitor. K iapp for ATP is 10.3 pM. RNA titration (right) used 250 pM ATP. Alpha value for RNA is 4.53x10 13 indicating an RNA-competitive inhibitor. K iapp for RNA is 9.59 pM. Data shown are an average of 3 independent experiments.

[0029] FIG. 5E shows PDB: 5ZC9 on the left. RNA is shown as a red cartoon. AMP-PNP is shown as purple sticks. Compound 1 (cyan) docks to the RNA pocket of elF4A (center). Zoomed surface view of the binding pocket is shown on the right.

[0030] FIGs. 6A-6D show mutagenesis studies that support docking pose of compound 28 binding to elF4A RNA groove.

[0031] FIG. 6A shows a surface view of compound 28 in the RNA groove of elF4A. (PDB: 5ZC9).

[0032] FIG. 6B shows compound 28 docked to elF4A with close residues labeled and shown as pink sticks. Potential interactions between the quinoline side of compound 28 and the pocket are shown as orange dashes (right).

[0033] FIG. 6C shows a 2D view of the right panel of FIG. 6B. Distances indicated are Angstroms.

[0034] FIG. 6D shows potency of compound 28 against elF4A mutants in the malachite green ATPase assay. Amino acid substitution is indicated under each bar. * Indicates IC50 > 50 pM.

[0035] FIG. 7A shows that compounds 1 and 28, but not compound 29, decrease BJAB cell viability. Compounds 1, 28, and 29 were tested using a CellTiter-Glo Luminescent Cell Viability assay in a BJAB Burkitt lymphoma cell line, n=4.

[0036] FIG. 7B shows that compound 28 inhibits cap-dependent translation. Top: Schematic representation of pSP/(CAG) 33 /FF/HCV/Ren pA 51 bicistronic reporter. Bottom: Effect of compound 28 on cap-dependent and HCV IRES-mediated translation in rabbit reticulocyte lysate programmed with pSP/(CAG) 33 /FF/HCV/Ren pA 51 vector. Luciferase luminescence values are normalized to signal obtained in control experiments with 1% DMSO. Cycloheximide (CHX) dose was 600 pM. Silvestrol dose was 100 nM. * Indicates a statistically significant difference between Renilla and firefly luciferase signal in a t test, p < .002. n=3.

[0037] FIG. 7C shows that compound 28 engages elF4A in cells. CETSA dose-response stabilization of elF4A by compound 28 and silvestrol (100 nM) in A549 cells. Values represent western blot band intensities of elF4A measured using densitometry. Intensities are normalized to GAPDH and subtracted from intensity of DMSO-treated sample.

[0038] FIGs. 8A-8D show that compound 28 is an RNA-competitive, ATP-uncompetitive elF4A inhibitor.

[0039] FIG. 8A shows kinetic testing of compound 28 using the malachite green assay. ATP titration (left) was done with 250 pg/mL RNA. Data were fit using unbiased mixed-model inhibition in Prism 6.0. Alpha value for ATP is 0.514, indicating an ATP-uncompetitive inhibitor. K iapp for ATP = 4.79 pM. RNA titration (right) was done with 250 pM ATP. Alpha value for RNA is 4.52x10 21 indicating an RNA-competitive inhibitor. Kiapp for RNA = 4.27 pM. Data shown are an average of 3 independent experiments.

[0040] FIG. 8B shows the FP assay with MANT-ATP as the fluorophore. PEL = Polarized Excitation Light.

[0041] FIG 8C shows the FP assay with FAM-labeled RNA as the fluorophore (left), compound 28 IC50 = 58.3 ± 2.5 pM. Compound 1 IC50 = 161 ± 9.8 pM (center). The center experiment utilized FAM-A(CAA)5 RNA. The right experiment utilized two different RNAs.

[0042] FIG. 8D shows the thermal shift assay with compound 28 and elF4A. Error bars are too small to see, n=3.

[0043] FIG. 9 shows that compounds 1 and 28 inhibit elF4A duplex unwinding. Cartoon of duplex used in the unwinding assay (top-right) with Cy3-labeled RNA (orange) and DNA loading strand (black). Increasing concentrations of compounds 1 and 28 were added to unwinding reactions containing duplex, elF4A, and ATP (left). Products of unwinding reactions were resolved on native polyacrylamide gels. Cy3 signal on gels was quantified by band densitometry. Percent inhibition for each reaction was calculated using (Cy3 duplex signal / total Cy3 signal), n=3.

[0044] FIG. 10 shows a model of mechanism of elF4A inhibition by compound 28. Under normal conditions (top), elF4A can bind ATP (purple) and RNA (red), unwind the RNA, hydrolyze the ATP, and release its substrates. Upon introduction of compound 28 (bottom), elF4A can bind ATP and weakly bind RNA, but in a conformation that hinders its ability to hydrolyze ATP and unwind RNA.

[0045] FIG. 11A shows a cartoon of the malachite green assay used in these studies.

[0046] FIG. 11 B shows the presence of 250 pg/mL Yeast RNA in the malachite green assay accelerates elF4A ATPase activity without increasing background.

[0047] FIG. 11C shows the Z-Factor of each 384 well plate used for singlicate screening, triplicate testing, and initial dose responses. Average Z Factor of every plate was 0.74.

[0048] FIG. 12 shows a table of DSF of elF4A and mutants.

[0049] FIG. 13 shows that compounds 1 and 28 inhibit elF4A duplex unwinding. The products of unwinding reactions were analyzed by electrophoresis on 20% native polyacrylamide (37.5:1 acrylamide: bisacrylamide) gels for 2 h at 150 V at 4 °C in 1x TBE buffer. All gel lanes contained single-stranded Cy3-labeled RNA. The number on the left indicates the compound used. DS = double stranded Cy3-duplex. SS = single stranded Cy3RNA. One representative gel is shown for each compound. n=3.

[0050] FIG. 14 shows that the furan moiety of compound 28 may form a hydrogen bond with T109. Compound 28 docked to elF4A (PDB: 5ZC9). T109 is labeled and shown as pink sticks. Potential hydrogen bond between 28 and T109 is shown as orange dashes. Distance indicated is Angstroms.

[0051]FIG. 15 shows that decrease in BJAB cell viability tracks elF4A ATPase inhibition. Compounds 9, 24, 25, 46, and 47 were tested using a CellTiter-Glo Luminescent Cell Viability assay in a BJAB Burkitt lymphoma cell line. n=4.

[0052] FIG. 16 shows a western blot of isothermal dose-response of 28 in A549 cells. All dose response CETSAs were conducted using previously determined in-cell elF4A melting temperature of 55 °C. Silvestrol treatment was 100 nM. DETAILED DESCRIPTION OF THE INVENTION

[0053] Referring now to FIG. 1, the present invention features an elF4A inhibitor having a formula according to Formula I: Formula I

[0054] In some embodiments, R may be, but it not limited to, H, alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, amino, alkoxy, nitro, halo, or a combination thereof.

[0055] In another embodiment, the elF4A inhibitor may have a formula according to Formula II: Formula II

[0056] In yet another embodiment, the present invention features an elF4A inhibitor having a formula according to Formula III: Formula III

[0057] In some embodiments, An may be, but is not limited to, aryl, substituted aryl, heteroaryl, or substituted heteroaryl.

[0058] In other embodiments, the present invention features an elF4A inhibitor having a formula according to Formula IV: io Formula IV

[0059] In some embodiments, R4 may be, but is not limited to, aryl, substituted aryl, heteroaryl, or substituted aryl.

[0060] In further embodiments, the elF4A inhibitor may have a formula according to Formula V or Formula VI: I

[0061] In some embodiments, R R 2 , and R 3 may be each independently H, alkyl, substituted alkyl, alkoxy, aryl, or amino. Without wishing to limit the present invention to any mechanism or theory, each of the elF4A inhibitors described herein may occupy a binding pocket of an elF4A RNA-binding groove, thereby perturbing RNA binding to elF4A, blocking ATP hydrolysis, and inhibiting elF4A from unwinding RNA.

[0062] In further embodiments, the elF4A inhibitor may have another formula which allows the inhibitor to occupy the same binding pocket of elF4A as the inhibitor of Formula II. As a non-limiting example, the elF4A inhibitor may be configured to bind or interact with one or more of R110, T158, and R311 of elF4A. As another non-limiting example, the elF4A inhibitor may be configured to bind or interact with all three of R110, T158, and R311 of elF4A The elF4A inhibitor may be an ATP uncompetitive and RNA competitive inhibitor. The elF4A inhibitor may be configured to inhibit elF4A unwinding while incompletely inhibiting RNA binding.

[0063]The present invention features a method for inhibiting elF4A activity in a cell. The method may comprise contacting the cell with an elF4A inhibitor. The elF4A inhibitor may have a formula according to Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, or another formula. Without wishing to limit the present invention to any theory or mechanism, the elF4A inhibitor may occupy a binding pocket of an elF4A RNA-binding groove, thereby perturbing RNA binding to elF4A, blocking ATP hydrolysis, and inhibiting elF4A from unwinding RNA. As a non-limiting example, the elF4A inhibitor may be configured to occupy the same binding pocket of elF4A as the inhibitor of Formula II. As another non-limiting example, the elF4A inhibitor may be configured to bind or interact with one or more of R110, T158, and R311 of elF4A.

EXAMPLE

[0064]The following is a non-limiting example of the present invention. It is to be understood that said example is not intended to limit the present invention in any way. Equivalents or substitutes are within the scope of the present invention.

Results

[0065] Human elF4A protein was produced recombinantly and a greater than 100,000-compound screen for inhibitors was completed using a malachite green ATPase assay (FIG. 11 A). Surprisingly, the only hits identified were not potent and could not be validated in triplicate and dose response studies (data not shown). A stable mixture of yeast RNAs was added in an effort to improve this system and create a more biologically relevant elF4A assay. elF4A ATPase activity was accelerated without a statistically significant increase in the background signal in the presence of this RNA (FIG. 11 B), and the new conditions gave excellent statistical parameters in a 384-well plate format (FIG. 11C).

[0066] This unique RNA-stimulated malachite green ATPase assay was repeated in a pilot screen for inhibitors of elF4A. Eighteen hits were identified in this screen (FIG. 5A), the most potent being a 2-(5-(4-butylphenyl)furan-2-yl)quinoline-4-carboxylic acid, compound 1 (FIG. 5B), with an IC 50 of 26.59 ± 2.46 pM (FIG. 5C). This molecule passed Lipinski and PAINS filters with zero violations according to SwissADME, and was evaluated in kinetics assays to study its mechanism of inhibition. The expectation was that compound 1 would be an ATP-competitive inhibitor, due to its ability to inhibit the ATPase activity of elF4A. However, surprisingly, kinetic testing showed that compound 1 was uncompetitive with respect to ATP (K iapp = 10.3 pM), as Km and Vmax of elF4A both decreased upon addition of compound 1 (FIG. 5D, left). Instead, compound 1 was found to be RNA-competitive (K iapp = 9.59 pM), as the Km of el F4A decreased and Vmax was constant upon treatment with compound 1 (FIG. 5D, right).

[0067] Molecular docking was performed to predict the binding site of 1, and as expected for a RNA-competitive inhibitor, it docked to the RNA-binding groove of elF4A (FIG. 5E). This predicted binding pocket engages both RecA-like domains of elF4A and has never been accessed before by a small molecule inhibitor. Visualization of the probable binding mode of compound 1 suggested that modification of the inhibitor could increase its favorable molecular interactions with the binding pocket and enhance its potency (FIG. 5E, right). This observation, along with the uniqueness of the binding location and mechanism of inhibition of 1 , encouraged us to synthesize a small library of analogs to expand this class of inhibitors.

[0068] Scheme 1. Synthesis of substituted quinoline analogs.

[0069] Scheme 2. Esterification of substituted quinoline analogs.

[0070] Scheme 3. Synthesis of 5-aminoisatin and 5-acetamidoisatin.

[0071] Synthesis of compound 1 analogs (Scheme 1) began with bromination of 2-acetylfuran with N-bromosuccinimide to yield 1i. The brominated product was utilized in a Suzuki coupling with 4-butylphenylboronic acid to produce 1ii. This intermediate was stockpiled and employed in Pfitzinger reactions with substituted isatins to afford 2-(5-(4-butylphenyl)furan-2-yl)quinoline-4-carboxylic acids with varied quinoline substitutions in 21%-39% yield over three steps. The carboxylic acids could also easily be esterified upon addition of sulfuric acid in ethanol (Scheme 2). Each molecule was tested in the RNA-stimulated malachite green ATPase assay (FIG. 1). Compound 2, the 1 -ethyl ester, was about 5-fold less potent than compound 1, which suggested that the carboxylic acid played an important role in elF4A inhibition. This was expected, as our docking data indicated the acid moiety formed a salt bridge with R311 of elF4A (FIG. 6).

[0072] Compound 3, with no bromine, was also much less potent than compound 1, indicating that a quinoline substitution was necessary for inhibition of elF4A. Subsequent analogs explored the effect of various halogen substitutions on each position of the quinoline ring. The 5-position of the quinoline was immediately ruled out as 4 showed a large drop in potency. Functionalizing the 6-position of the quinoline with a chlorine (6) or iodine (7) atom revealed moderate losses of potency compared to the bromine substituent, possibly due to unfavorable atomic size. Analogs with fluorine (8), chlorine (9), and bromine (10) substitutions to the 7-position of the quinoline displayed moderate potency, with 9 performing the best in the ATPase assay of any inhibitor produced. At the 8-position of the quinoline, the chlorine substitution (11) was less potent than bromine (12). This group of molecules was unable to produce a lead with a potency lower than 20 pM in the ATPase assay, so a few di-substituted halogen analogs were synthesized in an attempt to increase potency of the series. However, 6,7 and 6,8 substituted analogs 13-15 did not afford increased potency.

[0073] Scheme 4. Synthesis of central ring analogs.

[0074] Scheme 5. Synthesis of 32iv for central pyrrole analog.

[0075] Scheme 6. Synthesis of substituted phenyl analogs.

[0076] Scheme 7. Deprotection of N-Boc amines.

[0077]A shift to production of hydrocarbon-substituted quinolines at position 6 with compounds 16-19 revealed that the size of the substitution may not play as prominent of a role in conferring potency of elF4A inhibition in this binding pocket as originally expected. Despite a drastic gain in size from compounds 16-19, potency stayed low and relatively constant. This trend was consistent with dimethyl-substituted 20. This suggested that potent interactions between the quinoline moiety and the elF4A binding pocket were driven by electronic interactions and not purely by the size of the substitution. The next molecules produced featured electron-donating groups at the 6 and 7-positions of the quinoline. 6-methoxy (21) was much more potent than 7-methoxy (22), but neither were more potent than earlier molecules in the series. In addition, 6-trifluoromethoxy (23) was much less potent than compound 21.

[0078] The next two analogs produced with electron-donating group substitutions on the quinoline were 6-amino (24) and 6-acetamido (25) (Scheme 3), which also showed major losses in potency. Failure to increase potency with electron-donating substitutions on the quinoline led to production of analogs with electron-withdrawing group substitutions. 7-trifluoromethyl (26) and 8-trifluoromethyl (27) performed better in the ATPase assay than molecules with electron-donating substitutions, and compound 27 faired about two-fold better than compound 26. Consequently, a strongly electron-withdrawing group was placed at the 6-position of the quinoline to produce compound 28, which had an IC 50 of 8.603 ± 0.944 pM in the elF4A ATPase assay. This was over two-fold more potent than any other molecule previously produced in the series. Immediately, compound 28 was esterified to compound 29, which lost all elF4A potency, further reinforcing the importance of the carboxylic acid moiety to elF4A ATPase inhibition by this class of molecules (FIG. 6). Finally, due to the relative success of compound 27 with an electron withdrawing group at position 8 on the quinoline, compound 30 was synthesized with a nitro group at this position. Compound 30 was over three-fold less potent than compound 28. compound 28, like compound 1, passed Lipinski and PAINS tests with no violations according to SwissADME, predicting that compound 28 has a favorable physicochemical profile.

[0079]Additional analogs of 28 were synthesized to probe structure-activity relationships between different moieties of 28 and the elF4A binding pocket. Initially, the nitroquinoline and butylphenyl groups were held constant, while the furan was substituted for different five and six-membered rings. To produce these analogs, brominated, acetylated homocyclic and heterocyclic rings were utilized in Suzuki couplings with 4-butylphenylboronic acid, and the resulting intermediates were employed in Pfitzinger reactions with 5-nitroquinoline to result in central ring analogs 31-37 (Scheme 4). The sequence of the synthesis was unchanged from the derivatization of the quinoline, as performing the Pfitzinger reaction as the last step produced pure compounds in high yields. The advanced pyrrole intermediate 32iv was produced differently than the other molecules, as the amine of a pyrrole must be protected before it is employed in a Suzuki coupling to prevent debromination of the starting material.30 2-acetylpyrrole was selectively brominated at the 5-position with sodium bromide and Oxone, and the pyrrole amine was protected with a tert-butoxycarbonyl (Boc) protecting group. The protected pyrrole was employed in a Suzuki coupling to attach the butylphenyl moiety and the pyrrole was deprotected through removal of Boc to produce 32iv (Scheme 5), which was utilized in a Pfitzinger reaction with 5-nitroquinoline to produce the pyrrole analog 32. Each of these analogs were tested in the elF4A ATPase assay (FIG. 2).

[0080] Replacement of furan with 5-membered heterocycles thiophene (31) and pyrrole (32) led to a large drop in potency in the ATPase assay. The next three molecules produced (33-35) were six-membered rings with the butylphenyl group positioned meta to the quinoline. Compounds 33 and 34 experienced a drop in potency in the ATPase assay and 35 lost potency altogether. Analogs 36 and 37, with the quinoline and butylphenyl groups positioned para to each other, were also much less potent than 28 in the ATPase assay.

[0081] Finally, analogs with changes to the butylphenyl moiety were produced leaving the nitroquinoline and furan rings unchanged. These compounds were produced with the same general synthetic scheme as the central ring analogs, and derivatization resulted from employing different boronic acid species in the Suzuki coupling (Scheme 6). Compounds 38-45 and 48-54 were produced in this manner, and 46 and 47 were the result of Boc deprotection of 44 and 45, respectively (Scheme 7). Each compound was tested against elF4A in the malachite green ATPase assay (FIG. 3).

[0082]The first substituted phenyl analog synthesized and tested was compound 38, which had no butyl tail. This molecule experienced a drastic loss in potency, indicating that substitution to the phenyl ring was important to elF4A inhibition by this class of molecules. Next, phenyl rings substituted with para-hydrocarbon chains of different lengths were tested. Compounds 39 and 40 featured propyl and pentyl chains, respectively, which are one carbon shorter and longer than the butyl chain of 28. Shortening the chain did not improve potency, as compound 39 was about 5-fold less potent than 28, and lengthening the chain also did not improve potency, as 40 was no more potent than 28. With optimal chain length established, different positions of the chain were investigated with propylether phenyl substitutions. Compounds 41-43 all experienced drops in potency in the ATPase assay. Boc substitutions to the phenyl ring were also investigated with compounds 44 and 45 before being deprotected to amines 46 and 47. All four of these compounds showed low potency in the ATPase assay.

[0083] Various ring species were synthesized to explore the necessity for a phenyl at this position. Compound 48, with the benzo[1,3]dioxole substitution to the furan, lost all potency in the ATPase assay, while the 1-naphthyl-substituted compound 49 was about four-fold less potent than compound 28. Finally, phenyl and phenoxy substitutions at varied positions of the phenyl moiety were explored. Phenyl-substituted compounds 50 and 51 were less potent than compound 28 in the ATPase assay, with para-substituted 50 more potent than meta-substituted 51. Phenoxy substituted compounds 52-54 were generally more potent than 50 and 51, but not as potent as compound 28. Compound 28 was the most potent inhibitor of elF4A ATPase activity, and it was carried forward for additional exploration into its mechanism of inhibition.

[0084] Molecular docking of compound 28 to elF4A was performed to investigate the potential molecular interactions driving the potency of compound 28 for elF4A. Unsurprisingly, compound 28 docked to the same pocket as compound 1 in the RNA-binding groove of elF4A (FIG. 6A). The docking predicts several close contacts between compound 28 and the amino acids that form the binding pocket, but the main residues that appear to drive binding are R110, T158, and R311 (FIG. 6B). R110 and R311 are part of a string of positively charged amino acids that line the RNA-binding groove of elF4A, allowing elF4A to bind the negatively charged sugar-phosphate backbone of RNA. The negatively charged oxygen atoms of the nitro and acid groups are predicted to form salt bridges with R110 and R311 , respectively, which would disrupt the interaction between elF4A and RNA (FIG. 6C).

[0085] Furthermore, T158 is predicted to form a hydrogen bond to the carbonyl group of the 6-nitro quinoline substitution of compound 28. The ability of the 6-nitro group to form two strong interactions with the binding pocket of elF4A is the most likely explanation for why this substitution afforded 28 such a large gain in potency for compound 28 compared to other analogs in the inhibitor series. Mutagenesis and recombinant production of mutant elF4A protein was performed to confirm the docking pose and predicted interactions between compound 28 and its elF4A binding pocket (FIG. 6D). All elF4A mutants produced were determined to be thermodynamically stable by differential scanning fluorimetry (DSF), as each mutant had a melting temperature indicative of a properly folded protein (FIG. 12). Compound 28 remained moderately potent for the R110K mutant, which retains its positive charge, while R110L, R110M, and R110E mutants lost all potency, supporting the presence of an ionic interaction between compound 28 and R110. Compound 28 also lost all potency for T158A and T158V elF4A mutants, which do not have a hydrogen bond donor to contribute an interaction with compound 28.

[0086]These results strongly support the presence of a hydrogen bond between T158 and compound 28. Furthermore, elF4A R311K and R311 E mutants were produced and tested with compound 28. As expected, compound 28 had no potency toward R311E, as the positively charged arginine is replaced with a negatively charged glutamic acid that repels the carboxylic acid of compound 28. However, compound 28 also had no potency for elF4A R311K, even though it still maintains its positive charge. This may be attributed to the inability of this lysine to simultaneously provide hydrogen bond donation and ionic interactions to compound 28 due to geometrical constraints. The molecular docking also predicted that the furan of 28 may form a hydrogen bond with T109, and the butyl group may lie within a small hydrophobic groove within the binding pocket (FIG. 6A). Nevertheless, mutations to R110, T158, and R311 causing drastic losses in potency of compound 28 provided strong evidence that this was the correct binding pocket and pose for compound 28. This site, wedged between each RecA-like domain of elF4A, is a novel binding pocket that has never been accessed by an elF4A inhibitor.

[0087] Compounds 1 and compound 28 were evaluated in a CellTiter-Glo (CTG) Luminescence cell viability assay in BJAB Burkitt lymphoma cells that are known to be sensitive to elF4A inhibition. This assay quantitates the number of living, metabolically active cells by measuring their ATP levels. Compound 1 decreased BJAB cell viability with an EC 50 of 2.13 ± 0.17 pM (FIG. 7). Compound 28 also decreased viability, and like in the ATPase assay, compound 28 was three to four times more potent than 1, with an EC 50 of 0.46 ± 0.07 pM. Compound 29, the 28-ethyl ester, is not potent in the ATPase assay, likely due to the removal of the interaction between the carboxylic acid and R311 of elF4A. Compound 29 was tested in the BJAB viability assay and did not inhibit cellular viability, showing that structure-activity relationships of 28 and 29 correlate between the ATPase and cellular assays.

[0088] Additional analogs with varying potencies in the ATPase assay (9, 24, 25, 46, 47) were tested in the cell viability assay to investigate if ATPase inhibition tracked BJAB CTG inhibition and to ensure that cell viability was not decreasing due to general toxicity of the inhibitor scaffold. The potency of compound 9 in the ATPase assay (IC 50 = 21.65 ± 3.39 pM) is similar to that of compound 1 (IC 50 = 26.59 ± 2.46 pM). Both compounds also inhibited BJAB viability with similar EC 50 values (9 EC 50 = 4.29 ± 0.23 pM, 1 EC 50 = 2.13 ± 0.17 pM) (Figure S3). Compounds 24 and 25 retain the furan and butylphenyl moieties of 28, but have different quinoline substitutions, causing them to lose potency in the ATPase assay. These molecules do not decrease BJAB cellular viability. Furthermore, compounds 46 and 47 retain the nitroquinoline and furan moieties of 28, but feature different phenyl substitutions, causing them to lose potency in the ATPase assay. As expected, these molecules also do not inhibit BJAB cell viability. Minor substitutions to compound 28 that reduce potency in the ATPase assay also reduce potency in the BJAB CTG assay. The elF4A ATPase assay inhibition data closely tracks the BJAB viability data, providing additional evidence that 28 decreases BJAB cell viability as a result of inhibiting elF4A.

[0089] In vitro translation studies were performed to confirm the ability of 28 to inhibit cap-dependent translation in an environment with complete cellular translation machinery and further link inhibition of cellular viability by 28 to inhibition of elF4A. A bicistronic luciferase reporter (pSP/(CAG) 33 /FF/HCV/Ren pA 51 ) (FIG. 7B, top) was programmed into rabbit reticulocyte lysates, cellular extracts capable of performing both cap-dependent and Hepatitis C virus (HCV) internal ribosomal entry site (IRES)-mediated translation. elF4A is the enzymatic driver of cap-dependent translation and is not involved in translation through HCV IRES. Compounds that inhibit elF4A should inhibit cap-dependent translation of firefly (FF) luciferase while leaving IRES-mediated translation of Renilla (Ren) luciferase relatively unaffected. The inhibitory activities of cycloheximide, silvestrol, and 28 were assessed in this in vitro translation assay (FIG. 7B, bottom). Cycloheximide, an inhibitor of translation elongation, inhibits both cap-dependent and IRES-mediated translation. Compound 28 and silvestrol both inhibit cap-dependent translation of firefly luciferase, but only show a modest effect on HCV IRES-dependent translation of Renilla luciferase. The ability of compound 28 to preferentially target cap-dependent translation in a similar manner as silvestrol provides promising evidence that 28 inhibits elF4A and not other proteins in the general translational machinery.

[0090]A cellular thermal shift assay (CETSA) was performed to test the ability of 28 to bind and stabilize elF4A in cells and connect its inhibition of cellular viability and cap-dependent translation to engagement of elF4A. In CETSA, thermal stability of a protein is assessed by heating cells and then separating insoluble, aggregated, or denatured proteins from soluble proteins through supernatant/pellet fractionation. Levels of the protein of interest (elF4A) remaining in the soluble fraction are then measured by western blot. The principle of CETSA is similar to differential scanning fluorimetry (DSF), in that thermal stability and solubility of a protein increases upon binding a ligand. Treatment of A549 cells with silvestrol increased thermal stability of elF4A (FIG. 7C). Similarly, treatment with compound 28 caused a dose-dependent increase in thermal stability of elF4A as more elF4A appeared in the soluble fraction (FIG. 16). Taken together, compound 28 inhibits BJAB cell viability, inhibits cap-dependent translation, and engages elF4A in cells, indicating that 28 confers its cellular potency through engagement and inhibition of elF4A.

[0091] Additional biochemical testing was performed on compound 28 to investigate its mechanism of elF4A inhibition. Initially, kinetics experiments were repeated with compound 28, and the results for compound 28 matched those of compound 1 (FIG. 5D, 4A). Upon addition of compound 28, Km and Vmax of elF4A decreased with respect to ATP. With respect to RNA, elF4A Km decreased and Vmax remained constant. Consequently, compound 28 is predicted to act as an RNA-competitive (Ki app = 4.27 pM), ATP-uncompetitive (Kj app = 4.79 pM) elF4A inhibitor that binds to ATP-bound elF4A in the RNA groove. Fluorescence polarization (FP) assays were utilized to confirm this mechanism of inhibition by testing the ability of compound 28 to compete fluorescent substrates off elF4A. First, an FP assay was performed with MANT-ATP, a fluorescent, non-hydrolysable ATP analog, as the substrate. ADP, but not compound 28, was able to compete MANT-ATP out of its binding pocket (FIG. 8B), confirming that compound 28 is not an ATP-competitive inhibitor.

[0092] Next, an FP assay was performed with a fluorescent, elF4A-binding RNA oligo (FAM-A(CAA) 5 ) as the fluorophore. At high concentrations, both compound 1 and compound 28 were able to compete the fluorescent RNA off of elF4A, indicating that compound 28 is an RNA-competitive inhibitor (FIG. 8C, middle). However, the potency of these molecules in this assay were surprisingly low, indicating that compound 1 and compound 28 do not fully compete RNA out of the RNA-binding groove at concentrations in which the compounds inhibit elF4A ATPase activity (discussed below). The IC 50 of compound 1 was 160.6 ± 9.8 pM, and the IC 50 of compound 28 was 58.3 ± 2.5 pM, meaning compound 28 was about three-fold more potent than 1 , which was consistent with the other assays performed. This same assay was used to investigate if changing the identity of the RNA fluorophore affected the ability of compound 28 to compete the RNA off elF4A. FAM-(AG) 8 was utilized alongside FAM-A(CAA) 5 and the IC 50 of compound 28 in this assay was consistent regardless of RNA identity (FIG. 8C, right). This is evidence that compound 28 inhibits the RNA-elF4A interaction by binding elF4A and not the RNA.

[0093] Further evidence of this was uncovered in a thermal shift assay (TSA) using DSF with elF4A and compound 28. Compound 28 increased elF4A stability, exhibited by a rise in elF4A melting temperature (FIG. 8D) upon heating of the protein. Larger doses of compound 28 caused larger melting temperature (Tm) shifts in this assay, which argues that compound 28 is directly binding elF4A. These data indicate that compound 28 is an RNA-competitive, ATP-uncompetitive elF4A inhibitor that directly binds elF4A and inhibits the protein through a novel mechanism.

[0094] A functional duplex unwinding assay with a duplex nucleic acid substrate was utilized to test if compound 1 and compound 28 could inhibit elF4A helicase activity. This unwinding assay features a Cy3-labeled RNA strand annealed to a DNA strand with a long overhang that elF4A can bind (FIG. 9, top right). When ATP is added to protein and substrate, elF4A unwinds the duplex, leaving an unlabeled DNA strand and a Cy3-labeled single stranded RNA. When visualized on a native gel, the larger intact fluorescent duplex substrate will run higher on the gel than the smaller unwound single-stranded fluorescent RNA. The ratio of these two Cy3 signals can be used to determine the helicase activity of elF4A in each condition.

[0095] Dose responses of compound 1 and compound 28 were conducted using this assay. With no elF4A or ATP, no unwinding takes place, and the Cy3 signal is present in the double-stranded state. Without compound, unwinding proceeds and the signal is present in the single-stranded state. Upon addition of increasing amounts of each compound, unwinding activity decreases, as shown by an increase in double-stranded signal and decrease in single-stranded signal. The intensity of each band in each gel lane was quantified by densitometry to obtain IC 50 values for compound 1 and compound 28 in this assay. Compound 28 was about four-fold more potent than compound 1, which was consistent with the other assays performed comparing these two compounds (FIG. 9). In addition, the IC 50 values for compound 1 and compound 28 in the helicase assay correlated very closely with both IC 50 values in the ATPase assay. This suggests that compound 1 and compound 28 inhibit elF4A duplex unwinding as a direct result of inhibiting the ATPase activity of the protein. Altogether, compound 28 is an RNA-competitive, ATP-uncompetitive elF4A inhibitor that binds a novel pocket in the RNA groove and inhibits elF4A with a novel mechanism, in which it perturbs RNA binding, blocks the hydrolysis of ATP, and consequently, inhibits RNA helicase activity.

Discussion

[0096] Compound 28 inhibits elF4A through an RNA-competitive, ATP-uncompetitive mechanism that has not been observed for other elF4A inhibitors. In the absence of an inhibitor, elF4A exists in an open state until it cooperatively binds ATP and RNA. ATP hydrolysis then triggers RNA unwinding, a transition from the closed to the open state, and release of substrates. As an ATP-uncompetitive inhibitor of elF4A, compound 28 prefers to bind elF4A in the ATP-bound state. This cooperative nature of ATP and compound 28 binding matches the cooperative nature that ATP and RNA have for elF4A binding. Once ATP and compound 28 are bound, RNA binds elF4A in a manner that is either incomplete, or in a conformation that is inadequate for enzymatic activity of elF4A (FIG. 10). With no ATP hydrolysis, elF4A is unable to unwind RNA, and the RNA can more easily exit without being unwound.

[0097] The FP assay with the RNA fluorophore does not indicate that compound 28 enhances the interaction between elF4A and RNA, but the potency of compound 28 in this assay is markedly lower than the other assays (FIG. 8C, middle). At high concentrations, compound 28 completely competes off. The reason for this is unknown, although it is possible that compound 28 may be able to occupy additional less-favorable binding sites within the RNA groove at high concentrations of inhibitor. At lower concentrations of compound 28, doses that inhibit ATPase activity and unwinding activity, RNA is still bound to elF4A. These collective data strongly argue that elF4A forms at least a pseudo-closed ATP-bound state with compound 28 and RNA engaging the RNA groove, and that RNA does not have to be completely competed off elF4A for 28 to inhibit unwinding activity. The presence of compound 28 may force RNA to bind elF4A incorrectly, with the entire RNA groove not occupied, placing the protein in a position incapable of ATP hydrolysis and RNA unwinding.

[0098]This mechanism of inhibition is unique among elF4A inhibitors. Rocaglates inhibit elF4A by increasing the affinity of elF4A for RNA and clamping elF4A shut on RNA. PatA acts similarly, also stabilizing the elF4A-RNA interaction. In addition, PatA stimulates elF4A ATPase and helicase activities and inhibits the association of elF4A and elF4G, sequestering elF4A from the elF4F complex. The mechanism of elatol is not explicitly known, but its interaction with K82 of elF4A suggests it may be ATP-competitive. Hippuristanol is the only other RNA-competitive inhibitor of elF4A, but unlike compound 28, it locks elF4A closed and inhibits elF4A unwinding activity by completely preventing RNA binding. Conversely, the reduced potency of compound 28 in the RNA FP assay, as discussed above, suggests that compound 28 inhibits elF4A unwinding while incompletely inhibiting RNA binding.

[0099] Compound 28 also occupies a novel binding pocket within the elF4A RNA-binding groove. Rocaglates bind a small cavity that is encompassed by the N-terminal domain of elF4A and polypurine RNA. Desmethyl pateamine A (DMPatA), a PatA analog, occupies largely the same binding site as rocaglates, forming major interactions with RNA and the N-terminal domain of elF4A. However, unlike rocaglates, DMPatA also extends across the RNA to form minor interactions with the C-terminal domain of elF4A, and is able to clamp onto both polypurine and polypyrimidine RNA. Elatol binds elF4A in a 2:1 stoichiometric fashion, with both elatol molecules predicted to reside near the elF4A ATP pocket. Hippuristanol, although it shares an RNA-competitive mechanism with compound 28, binds exclusively to the C-terminal domain of elF4A. The uncovering of a novel binding pocket provides additional molecular space that can be probed to produce additional novel elF4A inhibitors.

[00100] An elF4A inhibitor that is uncompetitive with respect to ATP has potential advantages in cells or in vivo. This uncompetitive mechanism means that compound 28 prefers to bind elF4A when the protein is in the ATP-bound state, which is advantageous, as binding of ATP to elF4A facilitates a closed state that forms a pocket or groove for either compound 28 or both compound 28 and RNA to bind. Intracellular ATP concentrations are typically anywhere from 1 to 10 mM, which poses a problem with ATP-competitive inhibitors in that it is difficult to successfully compete with such a high concentration of ATP for binding to the protein. However, for an ATP-uncompetitive inhibitor such as compound 28, the high concentration of ATP in the cell is a benefit for the molecule. elF4A has a high likelihood of being ATP-bound in the cell, and compound 28 prefers binding elF4A when ATP is present on the protein.

[00101] The EC 50 of compound 28 in BJAB Burkitt lymphoma cells is lower than the IC 50 values for the inhibitor in biochemical assays. It is possible that BJAB cells are extremely sensitive to elF4A inhibition, and only a minor amount of elF4A inhibition may be required to decrease cell viability. The BJAB cell viability tracking with biochemical inhibition of elF4A for multiple analogs in the series, the ability of compound 28 to preferentially inhibit cap-dependent translation in cellular extracts, and the ability of compound 28 to engage elF4A in cells provides strong evidence against major off-target effects leading to inhibition of BJAB cellular viability by compound 28. However, further work may be required to develop a deeper understanding of the intricacies and consequences of this mode of elF4A inhibition. Compound 28 is a unique elF4A inhibitor that operates through a novel mechanism and occupies a previously unreported elF4A binding pocket. This molecule may give insight into the mechanism and interplay of elF4A and its substrates, trigger biochemical probing into a new binding site that has potential pharmacological relevance, and uncover downstream cellular translational effects resulting from this novel mechanism of elF4A inhibition.

Experimental

[00102] Cloning and recombinant protein expression and purification. elF4A was cloned into the pSpeedET expression vector (N-terminal 6xHis tag and TEV protease site) using ligation independent PIPE cloning. BL21-CodonPlus cells were transformed with the desired plasmid and were used to inoculate 2xYT media containing 50 pg/ml kanamycin. This culture was grown to an OD 600 of 0.8 at 37 °C before being transferred to a 16 °C incubator. After 1 hour at 16 °C, arabinose was added to a final concentration of 2 mg/mL and the cells were grown overnight. The next morning, cells were harvested by centrifugation and then resuspended in lysis buffer (50 mM HEPES pH 7.4, 150 mM KCI, 1 mM MgCI2, 10% glycerol, and 2 mM BME, 1X Protease Inhibitor Tablet [Pierce]). This slurry was then passed through a microfluidizer (LM10, Microfluidics Corporation) at 12,000 PSI three times and clarified by centrifugation. The supernatant was then incubated with cobalt talon resin (GoldBio) for 1 hr before being applied to a gravity column. This resin was then washed with 10 CV of lysis buffer and 10 CV of lysis buffer containing 5 mM imidazole. The desired protein was then eluted in lysis buffer containing 100 mM imidazole. His 6 -TEV protease was then added to the His-tagged elF4A and it was dialyzed against 20 mM Tris (pH 7.5), 10% glycerol, 0.1 mM EDTA, 2 mM DTT for 16 h at 4 °C to remove the His-tag. Protein was then incubated with Ni-NTA Agarose Resin (Qiagen) that had been washed with 10 CV dialysis buffer to recapture His 6 -TEV and any uncut elF4A and resin was removed by gravity filtration. elF4A was aliquoted, flash frozen using liquid nitrogen, and stored at -80 °C.

[00103] ATPase assay. Recombinant WT or mutant elF4A was added to a clear bottom 384 well plate (Greiner) in ATPase buffer (20 mM Tris pH 7.4, 80 mM KCI, 2.5 mM MgCI 2 , 1 mM DTT, 1% glycerol). Compounds were added and the plate was incubated at 37 °C for 20 min. ATP/RNA solution was added to afford a 20 pL per well reaction volume with 750 nM elF4A, 250 pM ATP, and 0.25 mg/mL Yeast RNA (Sigma) for the initial screen and dose responses, and varying concentrations of ATP and RNA for the kinetic assays. The plate was incubated for 4 h at 37 °C for the initial screen and dose responses, and at seven time points (1 hour - 7 hours) for kinetic assays. 0.04% v/v (final concentration) Tween-20 was added to a Malachite Green solution, and the Malachite Green (40 pL per well) was added. The solution was incubated for 5 minutes at room temperature and Absorbance at 660 nm was read on a SpectraMax iD5 plate reader (Molecular Devices).

[00104] Molecular docking. AutoDockTools version 1.5.6 (Scripps) was used for the docking simulations. MM2 energy-minimized molecules (compound 1 , compound 28) were prepared in Chem3D version 16.0 (PerkinElmer). Lamarckian genetic algorithm was selected for ligand conformational searching, and rigid docking was performed on elF4A (PDB: 5ZC9) first with RNA, rocaglate, and AMPPNP removed, and then with only RNA and rocaglate removed. Each molecule was docked 20 times, and the output of each docking simulation was the result of 2,500,000 evaluations. The lowest energy outputs for each interaction are displayed.

[00105] MANT-ATP fluorescence polarization assay. Recombinant elF4A (15 pM) was added to a low-volume black 384 well plate (Greiner). Yeast RNA (0.25 mg/mL) was added along with compound (200 pM) or ADP (1 mM) and the plate was incubated for 20 min. MANT-ATP was added (100 nM) and the plate was incubated for 30 min before fluorescence polarization was read at 355 nm : 448 nm (Ex : Em) on a SpectraMax iD5 plate reader (Molecular Devices).

[00106] FAM-RNA fluorescence polarization assay. Oligos were purchased from Integrated DNA Technologies using RNase free preparations. Recombinant elF4A (2 pM) was incubated with 25 nM FAM-labelled RNA for 30 min in FP Buffer (15 mM HEPES-NaOH (pH 8), 100 m NaCI, 1 mM MgCI 2 , 15% glycerol, 2 mM DTT, 2 mM AMPPNP) at 22 °C in low-volume black 384 well plates (Greiner). elF4A was incubated with compound for 20 min prior to the addition of FAM-RNA. Plate was incubated for another 30 min before fluorescence polarization was read at 485 : 535 nm (Ex : Em) on a SpectraMax iD5 plate reader (Molecular Devices. IC 50 values were calculated using GraphPad Prism v6.0.

[00107] Thermal Shift Assay (TSA)/Difterential Scanning Fluorimetry (DSF). TSA/DSF experiments were performed on a Lightcycler 480 II (Roche Molecular Systems). elF4A (5 pM) in buffer (20 mM Tris (pH 7.5), 10% glycerol, 0.1 mM EDTA, 2 m DTT, 2 m AMPPNP, 4% DMSO) was added to 96-well PCR plates (USA Scientific, Inc.) with or without compound. After a 5 min incubation, SYPRO orange dye was added to a final concentration of 5x. Samples were melted over a gradient from 20 to 85 °C with 20 acquisitions per °C at a ramp rate of 0.03 °C/s.

[00108] Cell viability assay. BJAB lymphoma cells were plated in quadruplicate at 5,000 cells/well in serial dilutions of drug ranging two logs with the top concentration of compounds tested at 10 uM. Viability was measured after 72 h using Cell Titer Gio (Promega G7573) following the manufacturer's protocol. Luminescence was detected on the BioTek HT Synergy plate reader and values were normalized to vehicle-treated (DMSO) wells. Silvestrol was included as a positive control (not shown).

[00109] Cellular Thermal Shift Assay (CETSA). A549 cells were seeded into wells in 6-well plates and on the following day were exposed to 28 or silvestrol (100 nM) for 90 min. Cells were harvested, resuspended in 50 pL DMEM, and dispensed into thin-walled PCR tubes. Cells were subjected to 3 min of heat at 55 °C in a thermocycler (miniPCR). After heating, cells were left to cool for 3 min and then snap-frozen on dry ice. Cells were subjected to 3 rapid freeze thaws with 15 seconds of vortexing after each thaw. After the first thaw, 1 pL of a dissolved Pierce protease inhibitor tablet (ThermoFisher - A32963) was added to the cell lysate. Cell lysates were centrifuged at 20,000 x g for 20 min. Samples were prepared for gel-electrophoresis and immunoblot analysis by transferring 40 pL of supernatant to 40 pL of 2X Laemmli (62.5mM Tris-CI (pH 6.9), 3% SDS, 20% glycerol, 10% |3-mercaptoethanol, and 0.1 % bromophenol blue), sonicating for 30 seconds using a Fisher Scientific Sonic Dismembrator 100 (ThermoFisher), and boiling for 10 minutes. Cell lysates were then resolved by SDS-PAGE and subjected to immunoblot analysis using primary antibodies against elF4A (Cell Signaling; 2490S) and GAPDH (Santa Cruz; sc-32233), and HRP-conjugated goat anti-rabbit (A0545) and goat anti-mouse (A9044) secondary antibodies (Sigma). All immunoblot images were taken using the Azure 600 imaging system (Azure Biosystems) and analyzed using Imaged 1.51s (NIH). Gel band densitometry analysis was performed using Imaged 1.53k (NIH).

In vitro transcription and translation

[00110] The dual-luciferase reporter plasmid pSP/(CAG) 33 /FF/HCV/Ren pA 51 was linearized with BamHI and subsequently in vitro transcribed using the Promega mMessage mMachine SP6 transcription kit, according to manufacturer’s specifications. RNA was precipitated with LiCI and resuspended in H 2 O for use in in vitro translation experiments. Rabbit reticulocyte lysate (RRL) (nuclease-treated, Promega) was used to assess inhibitory effects of 28 on translation of the dual-reporter. Briefly, a reaction consisting of 50% RRL, 30 ng/pL reporter RNA, 10 pM Amino Acid Mixture Minus Methionine, 10 pM Amino Acid Mixture Minus Leucine, 100 pM KCI, 1 U/pL RNAseOut (Thermo), and drug (final DMSO concentration 1%) were incubated at 30 °C for 90 minutes. The reaction was quenched with 50 pL 1x Passive Lysis Buffer (Promega), 10 pL of the mix was transferred to a 96-well white plate (Cellstar), and the Dual-Reporter Luciferase Assay System (Promega) was used to generate luminescence according to the manufacturer’s instructions. Luminescence was detected using a plate reader (BioTek HT Synergy).

[00111] Helicase assay. Oligos were purchased from Integrated DNA Technologies using RNase free preparations. Unwinding reactions are performed in 50 ul volumes which contained 10 nM RNA/DNA duplex, 2.5 pM elF4A, 2 mM ATP, 20 mM Tris (pH 7.5), 50 mM KCI, 1 mM DTT, 2 mM MgCI 2 , and 0.1 mg/ml BSA. The sequence of the short RNA strand was 5’-GUGCUUUACGGUGCU-3’ and the 5’ end was amine modified for labeling with Cy3. The sequence of the DNA strand was: 5’-GGGAGAAAAACAAAACAAAACAAAACTAGCACCGTAAAGCACGC-3’ . Reactions were incubated at 37 °C for 1 h and terminated by the addition of 12.5 l of 50% glycerol, 2% SDS, 20 mM EDTA. For samples with no elF4A, Cy3-RNA and DNA were mixed at 1 pM, placed in a 90 °C heat block, and the block was allowed to cool to room temp over 90 min. The samples were then placed on ice for 10 min, diluted to 50 nM and termination buffer was added. All products were analyzed by electrophoresis on 20% native polyacrylamide (37.5:1 acrylamide:bisacrylamide) gels. Gels were run at 4 °C for 2 hours and Cy3 signal was detected on an Azure 600 Fluorescent Imaging System (Azure Biosystems) at 524 nm : 572 nm (Ex : Em). Cy3 signal was quantified by gel band densitometry performed using lmageJ32 (NIH). IC 50 values were calculated using GraphPad Prism v6.0.

Chemistry

[00112] All chemicals that were commercially available were used without further purification. Detailed synthesis procedures can be found below for reactions described in Schemes 1-7. All reaction intermediates were purified by flash column chromatography. 1 H and 13 C NMR spectra were recorded on a Bruker AVIII-400 or Bruker DRX-500 NMR spectrometer. Coupling constants (J) are reported in hertz (Hz). Chemical shifts (5) are reported in parts per million referenced with respect to residual solvent (DMSO-d 6 ) 2.50 ppm and (Chloroform-d) 7.24 ppm or from internal standard tetramethylsilane (TMS) 0.00 ppm. The following abbreviations were used in reporting spectra: s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; dd, doublet of doublets; td, triplet of doublets; ddd, doublet of doublet of doublets; p, pentet; h, hextet. HPLC-grade solvents were used for all reactions. NMR data were processed using MestReNova 14.2.0 (Mestrelab). Flash column chromatography was performed using silica gel (230—400 mesh, Agela). High-resolution mass spectrometry (HRMS) was performed using a Q Exactive Plus Orbitrap MS system (Thermo Fisher). The mass spectrometer was operated in electrospray positive ionization mode (ESI+). Acquisition was a scan from m/z 100 to 1000 with an accumulation time of 2 minutes. The purity was assessed using an Agilent 1200 Series HPLC with an Alltech Econosphere Silica 5u column (cat no. 70006), 250 x 4.6 mm, at a flow rate of 0.75 mL/min; A = 430 nm; mobile phase: 95% dichloromethane, 5% methanol. All compounds were characterized by HRMS, proton NMR, and carbon NMR unless low yield prevented it and were confirmed >95.0% pure by HPLC traces. Synthesis Procedures

1 -(5-bromofuran-2-yl)ethan-1 -one (1 i).

[00113] To a round bottom flask was added 2-acetylfuran (2.75 g, 25 mmol). A solution of N-bromosuccinimide (4.89 g, 27.5 mmol) in DMF (5 ml_) was added dropwise over a period of 15 minutes and the reaction stirred at room temperature overnight. Ethyl acetate was added, and the reaction was washed with brine, dried (MgSO 4 ), filtered, and concentrated in vacuo. The product was purified by silica gel column chromatography (95:5 Hexanes :EtOAc) to afford the final product (white powder, 2.5 g, 13.2 mmol, 53% yield). 1 H NMR (400 MHz, Chloroform-d) 5 7.14 (d, J = 3.6 Hz, 1 H), 6.51 (d, J = 3.6 Hz, 1H), 2.49 (s, 3H). 13 C NMR (101 MHz, Chloroform-d) 5 185.48, 154.55, 128.17, 118.90, 114.38, 25.75. HRMS (ESI) calcd for [M+H] + , 188.9473; found 188.9506.

General procedure for the production of advanced acetylated ring intermediates (Hi, 311, 331-451, 481-541).

[00114] To a stirred solution of a boronic acid-substituted ring (3 mmol), Cs s CO 3 (978 mg, 3 mmol), acetylated, bromo-substituted ring (1.5 mmol), and 9:1 toluene:water was added tetrakis(triphenylphosphine)palladium(0) (87 mg, 5 mol%,) and the reaction stirred overnight at 100 °C. The mixture was filtered through celite and rinsed with ethyl acetate, and the filtrate was concentrated in vacuo. The product was purified by silica gel column chromatography to afford the final product.

1 -(5-(4-butylphenyl)furan-2-yl)ethan-1 -one (1 ii)

[00115] 1 H NMR (400 MHz, Chloroform-d) 5 7.73 (d, 2H), 7.31 - 7.23 (m, 3H), 6.75 (d, J = 3.7 Hz, 1H), 2.67 (t, 2H), 1.65 (p, 2H), 1.40 (h, 2H), 0.97 (t, 3H). 13 C NMR (101 MHz, Chloroform-d) 5 186.34, 158.08, 151.67, 144.50, 128.95, 126.86, 124.97, 119.60, 106.82, 35.53, 33.42, 25.93, 22.32, 13.93. HRMS (ESI) calcd for [M+H] + , 243.1307; found 243.13798.

1 -(5-(4-butylphenyl)thiophen-2-yl)ethan-1 -one (311)

[00116] 1 H NMR (500 MHz, Chloroform-d) 5 7.60 (d, J = 3.9 Hz, 1H), 7.54 - 7.50 (m, 2H), 7.24 (d, J = 4.0 Hz, 1H), 7.20 - 7.16 (m, 2H), 2.59 (t, J = 7.7 Hz, 2H), 2.51 (s, 3H), 1.61 - 1.53 (m, 2H), 1.33 (h, 2H), 0.89 (t, J = 7.4 Hz, 3H). HRMS (ESI) calcd for C 16 H 19 OS [M+H] + , 259.1078; found 259.1093. 1 -(4"-buty l-[1 , 1 '-biphenyl]-3-yl)ethan-1 -one (33 i)

[00117] 1 H NMR (400 MHz, Chloroform-d) 5 8.20 (t, J = 1.8 Hz, 1H), 7.97 - 7.91 (m, 1H), 7.81 (ddd, J = 7.7, 1.9, 1.1 Hz, 1 H), 7.60 - 7.51 (m, 3H), 7.34 - 7.29 (m, 2H), 2.75 - 2.66 (m, 5H), 1.68 (h, 2H), 1.43 (h, J = 7.3 Hz, 2H), 0.99 (t, J = 7.3 Hz, 3H). HRMS (ESI) calcd for C 18 H 21 O [M+H] + , 253.1514; found 253.1563.

1 -(6-(4-butylphenyl)pyridin-2-yl)ethan-1 -one (34i)

[00118] 1 H NMR (500 MHz, Chloroform-d) 5 7.95 - 7.90 (m, 2H), 7.85 (dd, J = 7.3, 1.4 Hz, 1H), 7.82 - 7.73 (m, 2H), 7.26 - 7.20 (m, 2H), 2.74 (s, 3H), 2.60 (t, J = 7.7 Hz, 2H), 1.61 - 1.51 (m, 2H), 1.31 (h, J = 7.3 Hz, 2H), 0.87 (t, J = 7.3 Hz, 3H). HRMS (ESI) calcd for C 17 H 20 NO [M+H] + , 254.1467; found 254.1501.

1 -(5-(4-butylphenyl)pyridin-3-yl)ethan-1 -one (35i)

[00119] 1 H NMR (400 MHz, Chloroform-d) 5 9.13 (d, J = 2.1 Hz, 1H), 9.03 (d, J =

2.3 Hz, 1H), 8.46 (t, J = 2.2 Hz, 1H), 7.61 - 7.52 (m, 2H), 7.39 - 7.31 (m, 2H), 2.77 - 2.67 (m, 5H), 1.73 - 1.62 (m, 2H), 1.42 (h, J = 7.3 Hz, 2H), 0.98 (t, J = 7.3 Hz, 3H). HRMS (ESI) calcd for C 17 H 20 NO [M+H] + , 254.1467; found 254.1499.

1 -(4'-butyl-[1 ,1 '-biphenyl]-4-yl)ethan-1 -one (36 i)

[00120] 1 H NMR (500 MHz, Chloroform-d) 5 7.99 - 7.94 (m, 2H), 7.66 - 7.60 (m, 2H), 7.52 - 7.47 (m, 2H), 7.26 - 7.22 (m, 2H), 2.65 - 2.59 (m, 2H), 2.58 (s, 3H), 1.65 - 1 .55 (m, 2H), 1 .34 (h, J = 7.4 Hz, 2H), 0.90 (t, J = 7.4 Hz, 3H). HRMS (ESI) calcd for C 18 H 21 O [M+H] + , 253.1514; found 253.1542.

1 -(6-(4-butylphenyl)pyridin-3-yl)ethan-1 -one (37i)

[00121] 1 H NMR (500 MHz, Chloroform-d) 5 9.13 (dd, J = 2.4, 0.9 Hz, 1H), 8.23 (dd, J = 8.4, 2.3 Hz, 1 H), 7.94 - 7.88 (m, 2H), 7.75 (dd, J = 8.4, 0.8 Hz, 1 H), 7.26 - 7.20 (m, 2H), 2.61 - 2.54 (m, 5H), 1.59 - 1.49 (m, 2H), 1.28 (h, J = 7.4 Hz, 2H), 0.84 (t, J =

7.4 Hz, 3H). HRMS (ESI) calcd for C 17 H 20 NO [M+H] + , 254.1467; found 254.1485.

1 -(5-phenylfuran-2-yl)ethan-1 -one (38i)

[00122] 1 H NMR (500 MHz, Chloroform-d) 5 7.79 - 7.73 (m, 2H), 7.43 - 7.38 (m, 2H), 7.37 - 7.31 (m, 1 H), 7.24 (d, J = 3.7 Hz, 1 H), 6.75 (d, J = 3.7 Hz, 1 H), 2.51 (s, 3H). HRMS (ESI) calcd for C 12 H 11 O 2 [M+H] + , 187.0681; found 187.0712. 1 -(5-(4-propylphenyl)furan-2-yl)ethan-1 -one (39 i)

[00123] 1 H NMR (400 MHz, DMSO-d 6 ) 5 7.80 - 7.73 (m, 2H), 7.55 (d, J = 3.7 Hz, 1 H), 7.36 - 7.27 (m, 2H), 7.15 (d, J = 3.7 Hz, 1 H), 2.64 (t, J = 7.7 Hz, 2H), 2.46 (s, 3H), 1 .62 (h, 2H), 0.91 (t, J = 7.3 Hz, 3H). HRMS (ESI) calcd for C 15 H 17 O 2 [M+H] + , 229.1150; found 229.1188.

1 -(5-(4-pentylphenyl)furan-2-yl)ethan-1 -one (40 i)

[00124] 1 H NMR (400 MHz, DMSO-d 6 ) 5 7.81 - 7.73 (m, 2H), 7.55 (d, J = 3.7 Hz, 1 H), 7.36 - 7.28 (m, 2H), 7.14 (d, J = 3.7 Hz, 1 H), 2.64 (t, J = 7.7 Hz, 2H), 2.46 (s, 3H), 1.62 (h, 2H), 1.38 - 1.23 (m, 4H), 0.91 (t, J = 7.3 Hz, 3H). HRMS (ESI) calcd for C 17 H 20 O 2 [M+H] + , 257.1463; found 257.1490.

1 -(5-(4-propoxyphenyl)furan-2-yl)ethan-1 -one (41 i)

[00125] 1 H NMR (400 MHz, Chloroform-d) 5 7.77 - 7.70 (m, 2H), 7.30 - 7.25 (m, 1 H), 7.00 - 6.93 (m, 2H), 6.66 (d, J = 3.7 Hz, 1 H), 3.99 (t, J = 6.6 Hz, 2H), 2.54 (s, 3H), 1 .87 (h, 2H), 1.09 (t, J = 7.4 Hz, 3H). HRMS (ESI) calcd for C 15 H 17 O 3 [M+H] + , 245.1099; found 257.1126.

1 -(5-(3-propoxyphenyl)furan-2-yl)ethan-1 -one (42 i)

[00126] 1 H NMR (400 MHz, Chloroform-d) 5 7.43 - 7.32 (m, 3H), 7.28 (d, J = 3.7 Hz, 1 H), 6.94 (ddd, J = 7.6, 2.6, 1.6 Hz, 1 H), 6.79 (d, J = 3.7 Hz, 1 H), 4.01 (t, J = 6.5 Hz, 2H), 2.54 (s, 3H), 1.86 (h, J = 7.1 Hz, 2H), 1 .10 (t, J = 7.4 Hz, 3H). HRMS (ESI) calcd for C 15 H 17 O 3 [M+H] + , 245.1099; found 257.1129.

1 -(5-(2-propoxyphenyl)furan-2-yl)ethan-1 -one (43i)

[00127] 1 H NMR (400 MHz, Chloroform-d) 5 8.03 (dd, J = 7.8, 1.7 Hz, 1H), 7.34 (ddd, J = 8.4, 7.3, 1.8 Hz, 1H), 7.31 - 7.27 (m, 1H), 7.13 (d, J = 3.7 Hz, 1H), 7.06 (td, J = 7.6, 1.1 Hz, 1H), 6.99 (dd, J = 8.3, 1.1 Hz, 1H), 4.10 (t, J = 6.5 Hz, 2H), 2.56 (s, 3H), 1.97 (h, 2H), 1.14 (t, J = 7.4 Hz, 3H). HRMS (ESI) calcd for C 15 H 17 O 3 [M+H] + , 245.1099; found 257.1137. tert-butyl (4-(5-acetylfuran-2-yl)phenyl)carbamate (44i)

[00128] 1 H NMR (500 MHz, Chloroform-d) 5 7.73 - 7.68 (m, 1H), 7.44 (d, J = 8.4 Hz, 2H), 7.24 (d, J = 3.6 Hz, 1 H), 6.68 (d, J = 3.6 Hz, 2H), 2.51 (s, 3H), 1.53 (s, 9H). HRMS (ESI) calcd for C 17 H 20 NO 4 [M+H] + , 302.1314; found 302.1368. tert-butyl (3-(5-acetylfuran-2-yl)phenyl)carbamate (451)

[00129] 1 H NMR (500 MHz, Chloroform-d) 5 7.80 (t, J = 2.0 Hz, 1H), 7.45 (dt, J = 7.6, 1.5 Hz, 1H), 7.39 (d, J = 8.2 Hz, 1H), 7.33 (t, J = 7.9 Hz, 1H), 7.24 (d, J = 3.7 Hz, 1 H), 6.78 (d, J = 3.7 Hz, 1 H), 2.52 (s, 3H), 1 .53 (s, 9H). HRMS (ESI) calcd for C 17 H 20 NO 4 [M+H] + , 302.1314; found 302.1362.

1 -(5-(benzo[d][1 ,3]dioxol-5-yl)furan-2-yl)ethan-1 -one (48i)

[00130] 1 H NMR (400 MHz, DMSO-d 6 ) 5 7.52 (d, J = 3.7 Hz, 1H), 7.43 - 7.42 (m, 1 H), 7.39 (dd, J = 8.1, 1.8 Hz, 1H), 7.09 (d, J = 3.7 Hz, 1H), 7.05 (dd, J = 8.1, 0.4 Hz, 1 H), 6.11 (s, 2H), 2.45 (s, 3H).HRMS (ESI) calcd for C 13 H 11 O 4 [M+H] + , 231.0579; found 231.0602.

1 -(5-(naphthalen-1 -yl)furan-2-yl)ethan-1 -one (49 i)

[00131] 1 H NMR (500 MHz, Chloroform-d) 5 8.26 (dt, J = 8.6, 1.0 Hz, 1H), 7.81 - 7.75 (m, 2H), 7.72 - 7.68 (m, 1 H), 7.46 - 7.39 (m, 3H), 7.24 (d, J = 3.6 Hz, 1 H), 6.73 (d, J = 3.6 Hz, 1H), 2.44 (s, 3H). HRMS (ESI) calcd for C 16 H 13 O 2 [M+H] + , 237.0837; found 237.0854.

1 -(5-([1 ,1'-biphenyl]-4-yl)furan-2-yl)ethan-1 -one (50 i)

[00132] 1 H NMR (400 MHz, DMSO-d 6 ) 5 7.99 - 7.92 (m, 2H), 7.87 - 7.80 (m, 2H), 7.79 - 7.71 (m, 2H), 7.59 (d, J = 3.7 Hz, 1 H), 7.54 - 7.46 (m, 2H), 7.45 - 7.37 (m, 1 H), 7.28 (d, J = 3.6 Hz, 1H), 2.49 (s, 3H). HRMS (ESI) calcd for C 18 H 15 O 2 [M+H] + , 263.0994; found 263.1015.

1 -(5-([1 ,1'-biphenyl]-3-yl)furan-2-yl)ethan-1 -one (51 i)

[00133] 1 H NMR (400 MHz, DMSO-d 6 ) 58.11 (td, J = 1.8, 0.5 Hz, 1H), 7.86 (ddd, J = 7.7, 1.8, 1.1 Hz, 1 H), 7.79 - 7.73 (m, 2H), 7.72 (ddd, J = 7.8, 1 .8, 1.1 Hz, 1 H), 7.64 - 7.56 (m, 2H), 7.55 - 7.49 (m, 2H), 7.46 - 7.39 (m, 1 H), 7.36 (d, J = 3.7 Hz, 1 H), 2.49 (s, 3H). HRMS (ESI) calcd for C 18 H 15 O 2 [M+H] + , 263.0994; found 263.1037.

1 -(5-(4-phenoxyphenyl)furan-2-yl)ethan-1 -one (52 i)

[00134] 1 H NMR (400 MHz, Chloroform-d) 5 7.81 - 7.75 (m, 2H), 7.44 - 7.37 (m, 2H), 7.30 - 7.27 (m, 1H), 7.19 (ddt, J = 8.4, 7.6, 1.2 Hz, 1 H), 7.11 - 7.04 (m, 4H), 6.72 (d, J = 3.7 Hz, 1H), 2.55 (s, 3H). HRMS (ESI) calcd for C 18 H 15 O 3 [M+H] + , 279.0943; found 279.0980. 1 -(5-(3-phenoxyphenyl)furan-2-yl)ethan-1 -one (53I)

[00135] 1 H NMR (400 MHz, Chloroform-d) 5 7.59 - 7.53 (m, 1 H), 7.47 (dd, J = 2.5, 1 .6 Hz, 1 H), 7.43 - 7.35 (m, 3H), 7.27 (d, J = 3.7 Hz, 1 H), 7.16 (ddt, J = 7.5, 5.9, 1.1 Hz, 1 H), 7.09 - 7.04 (m, 2H), 7.01 (ddd, J = 8.1, 2.5, 1.0 Hz, 1 H), 6.77 (d, J = 3.7 Hz, 1H), 2.54 (s, 3H). HRMS (ESI) calcd for C 18 H 15 O 3 [M+H] + , 279.0943; found 279.0971.

1 -(5-(2-phenoxyphenyl)furan-2-yl)ethan-1 -one (54i)

[00136] 1 H NMR (400 MHz, Chloroform-d) 5 8.09 (dd, J = 7.8, 1.8 Hz, 1H), 7.40 - 7.33 (m, 2H), 7.33 - 7.26 (m, 1H), 7.25 - 7.19 (m, 2H), 7.14 (tt, J = 7.4, 1.1 Hz, 1H), 7.07 - 7.00 (m, 3H), 6.95 (dd, J = 8.2, 1.3 Hz, 1H), 2.52 (s, 3H). HRMS (ESI) calcd for C 18 H 15 O 3 [M+H] + , 279.0943; found 279.0984.

1 -(5-bromo-1 /7-pyrrol-2-yl)ethan-1 -one (32 i).

[00137] Oxone (7.15 g, 11.6 mmol) and NaBr (2.99 g, 29.1 mmol) were added to a solution of 2-acetylpyrrole (2.54 g, 23.3 mmol) in a mixture of methanol (50 ml_) and water (50 ml_). The reaction stirred for 8 hours at room temperature. The resulting mixture was filtered, and the filtrate was extracted with dichloromethane. The organic layer was dried (Na 2 SO 4 ) and filtered. The solvent was removed in vacuo and the residue was purified by silica gel chromatography (90:10 Hexanes :EtOAc) to afford the final product (white solid, 1.68 g, 8.91 mmol, 38% yield). 1 H NMR (400 MHz, Chloroform-d) 5 7.04 - 6.99 (m, 1H), 6.93 - 6.87 (m, 1H), 2.41 (s, 3H). HRMS (ESI) calcd for C 6 H 7 BrNO [M+H] + , 187.9633; found, 187.9657. tert-butyl 2-acetyl-5-bromo-1 H-pyrrole-1 -carboxylate (32II).

[00138] 1-(5-bromo-1H-pyrrol-2-yl)ethan-1-one (32I, 2.84 g, 15 mmol) was dissolved in triethylamine (2.1 ml_) and 4-dimethylaminopyridine (36.5 mg, 2 mol%) was added. Di-tert-butyl dicarbonate (3.27 g, 15 mmol) was dissolved in 15 mL THF and added to the reaction. This reaction stirred overnight at room temperature. The resulting mixture was diluted with 15 mL of saturated sodium bicarbonate, extracted with ether, dried (Na 2 SO 4 ), and filtered. The solvent was removed in vacuo and the residue was purified by silica gel chromatography (96:4 Hexanes :EtOAc) to afford the final product (white solid, 0.96 g, 0.33 mmol, 22% yield). 1 H NMR (400 MHz, Chloroform-d) 5 7.32 (d, J = 1.8 Hz, 1H), 6.81 (d, J = 1.8 Hz, 1H), 2.46 (s, 3H), 1.60 (s, 9H). HRMS (ESI) calcd for C 11 H 15 BrNO 3 [M+H] + , 288.0157; found 288.0210. tert-butyl 2-acetyl-5-(4-butylphenyl)-1 H-pyrrole-1 -carboxylate (32111).

[00139] To a stirred solution of 4-butylphenylboronic acid (356 mg, 2 mmol), Cs s CO 3 (652 mg, 2 mmol), tert-butyl 2 -acetyl-5-bromo-1 /-/-pyrrole-1 -carboxylate (32II, 285 mg, 1 mmol), 8 ml_ dioxane, and 2 ml_ water was added tetrakls(triphenylphosphine)palladium(0) (116 mg, 10 mol%,) and the reaction stirred overnight at 100 °C. The mixture was filtered through celite and rinsed with ethyl acetate, and the filtrate was concentrated in vacuo. The product was purified by silica gel column chromatography (95:5 Hexanes :EtOAc) to afford the final product (amber solid, 157 mg, 0.46 mmol, 46% yield). 1 H NMR (400 MHz, Chloroform-d) 5 7.57 (d, J = 1.8 Hz, 1H), 7.46 - 7.41 (m, 2H), 7.24 - 7.19 (m, 2H), 6.79 - 6.74 (m, 1H), 2.69 - 2.61 (m, 2H), 2.53 (s, 3H), 1.73 - 1.64 (m, 2H), 1.63 (s, 9H), 1 .39 (h, J = 7.3 Hz, 2H), 0.98 (t, J = 7.3 Hz, 3H). HRMS (ESI) calcd for C 21 H 27 NO 3 [M+H] + , 342.1991; found, 342.2017.

1 -(5-(4-butylphenyl)-1 /7-pyrrol-2-yl)ethan-1 -one (32iv).

[00140] tert-butyl 2-acetyl-5-(4-butylphenyl)-1 /-/-pyrrole-1 -carboxylate (32iii, 100 mg, 0.3 mmol) was dissolved in a 3 ml_ solution of 50:50 TFA:dichloromethane. The reaction stirred at room temperature and was monitored by TLC (80:20 Hexanes:EtOAc). When the reactant was consumed, saturated sodium bicarbonate (10 ml_) was added, the reaction was extracted with dichloromethane, washed with brine, dried (Na 2 SO 4 ), filtered, and purified via silica gel chromatography (85:15 Hexanes :EtOAc) to afford the product (white solid, 65 mg, 0.27 mmol, 90% yield). 1 H NMR (400 MHz, CDCI 3 ) 5 7.49 - 7.43 (m, 2H), 7.30 (dd, J = 3.0, 1.6 Hz, 1H), 7.24 - 7.20 (m, 2H), 7.17 (dd, J = 2.6, 1.6 Hz, 1H), 2.64 (t, J = 7.7 Hz, 2H), 2.51 (s, 3H), 1.71 - 1.60 (m, 2H), 1.41 (h, J = 7.3 Hz, 2H), 0.97 (t, J = 7.3 Hz, 3H). HRMS (ESI) calcd for C 16 H 20 NO [M+H] + , 242.1467; found, 242.1498.

5-aminoisatin (24i).

[00141] 5-nitroisatin (500 mg, 2.6 mmol) was dissolved in warm ethanol (10 ml_), and reduced iron powder was added. As the reaction stirred under argon, concentrated HCI (2 ml_) was added dropwise, and the reaction mixture stirred for 3 hours at 75 °C. The mixture was treated with saturated sodium bicarbonate and brine, and extracted with ethyl acetate. The organic layer was dried (MgSO 4 ), filtered, and concentrated in vacuo. The crude product was purified by silica gel chromatography (97:3 dichloromethane:methanol) to afford the final product (dark red solid, 170 mg, 1 mmol, 40% yield). 1 H NMR (400 MHz, DMSO-d 6 ) 5 10.58 (s, NH), 6.83 (dd, J = 2.5, 8.3 Hz, 1 H), 6.72 (d, J = 2.4 Hz, 1H), 6.64 (d, J = 8.2 Hz, 1H), 5.08 (s, 2NH). 13 C NMR (101 MHz, DMSO) 5 185.86, 159.84, 145.33, 141.33, 124.04, 118.57, 113.18, 109.56. HRMS (ESI) calcd for C 8 H 6 N 2 O 2 [M+H] + , 163.0429; found, 163.05020.

5-acetamidoisatin (25i).

[00142] 5-aminoisatin (40 mg, 0.25 mmol) was dissolved in pyridine (1.5 ml_) and stirred at 0 °C under argon. Acetic anhydride (28 pL, 0.25 mmol) was added dropwise, and the reaction stirred for 3 hr. A saturated solution of sodium bicarbonate was added (1.5 ml_), and the mixture was extracted with ethyl acetate, dried (MgSO 4 ) and concentrated in vacuo. The residue was purified by silica gel chromatography (50:50 Hexanes :EtOAc) to isolate the final product (orange solid, 11 mg, 0.054 mmol, 22% yield). 1 H NMR (400 MHz, Acetone-d 6 ) 5 9.93 (s, NH), 9.28 (s, NH), 7.95 (d, J = 2.2 Hz, 1 H), 7.78 (dd, J = 2.3, 8.4 Hz, 1H), 6.98 (d, J = 8.5 Hz, 1H), 2.08 (s, 3H). HRMS (ESI) calcd for C 10 H 8 N 2 O 3 [M+H] + , 204.0535; found, 204.05629.

General procedure for the production of carboxylic acid elF4A inhibitor analogs (1, 3-28, 30-45, 48-54).

[00143] To a stirred solution of an isatin (0.37 mmol) in ethanol (0.87 ml_), was added a solution of KOH (62.3 mg) in water (0.13 ml_). The reaction stirred for 20 minutes, an advanced acetylated ring intermediate (0.37 mmol) was added slowly, and the reaction stirred overnight at 90 °C. Water (1 ml_) was added, and glacial acetic acid was added in 20 pL increments to form a precipitate. The precipitate was filtered and washed with and dichloromethane and acetone to afford the product.

6-bromo-2-(5-(4-butylphenyl)furan-2-yl)quinoline-4-carbox ylic acid (1).

[00144] 1 H NMR (400 MHz, DMSO-d 6 ) 58.91 (d, J = 2.2 Hz, 1 H), 8.44 (s, 1 H), 8.04 (d, J = 9.0 Hz, 1H), 7.96 (dd, J = 9.0, 2.3 Hz, 1H), 7.82 (d, J = 8.2 Hz, 2H), 7.59 (d, J = 3.6 Hz, 1H), 7.34 (d, J = 8.2 Hz, 2H), 7.19 (d, J = 3.6 Hz, 1 H), 2.64 (t, J = 7.7 Hz, 2H), 1.60 (p, 2H), 1.35 (h, J = 7.3 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 167.43, 156.17, 151.83, 148.89, 147.71 , 143.50, 133.95, 131.88, 129.50, 129.32, 127.59, 125.00, 124.62, 121.23, 119.74, 114.83, 34.98, 33.29, 22.07, 14.09. HRMS (ESI) calcd for C 24 H 20 BrNO 3 [M+H] + , 450.0627; found, 450.0699. 2-(5-(4-butylphenyl)furan-2-yl)quinoline-4-carboxylic acid (3).

[00145] 1 H NMR (500 MHz, DMSO-d 6 ) 5 8.62 (dd, J = 1.4, 8.5 Hz, 1H), 8.38 (s, 1 H), 8.11 (dt, J = 1.2, 8.4 Hz, 1 H), 7.88 - 7.80 (m, 3H), 7.68 (ddd, J = 1.3, 6.8, 8.3 Hz, 1 H), 7.56 (d, J = 3.6 Hz, 1H), 7.35 (d, J = 8.3 Hz, 2H), 7.19 (d, J = 3.5 Hz, 1H), 2.65 (t, J = 7.7 Hz, 2H), 1.60 (p, 2H), 1.35 (h, 2H), 0.93 (t, J = 7.4 Hz, 3H). HRMS (ESI) calcd for C 24 H 22 NO 3 [M+H] + , 372.1521 ; found 372.15961.

2-(5-(4-butylphenyl)furan-2-yl)-5-chloroquinoline-4-carbo xylic acid (4).

[00146] 1 H NMR (400 MHz, DMSO-d 6 ) 5 8.12 (s, 1H), 8.07 (dd, J = 1.4, 8.2 Hz, 1 H), 7.89 - 7.83 (m, 2H), 7.83 - 7.72 (m, 2H), 7.61 (d, J = 3.6 Hz, 1H), 7.33 (d, J = 8.2 Hz, 2H), 7.19 (d, J = 3.6 Hz, 1H), 2.64 (t, J = 7.7 Hz, 2H), 1.60 (p, J = 7.5 Hz, 2H), 1.35 (h, J = 7.3 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (101 MHz, DMSO-d 6 ) 5 169.89, 157.20, 151.63, 151.51, 149.60, 146.58, 145.67, 143.42, 131.02, 129.39, 127.57, 124.71, 124.58, 120.76, 120.20, 108.70, 100.07, 35.11 , 33.44, 22.22, 14.25. HRMS (ESI) calcd for C 24 H 21 CINO 3 [M+H] + , 406.1132; found 406.12039.

2-(5-(4-butylphenyl)furan-2-yl)-6-fluoroquinoline-4-carbo xylic acid (5).

[00147] 1 H NMR (400 MHz, DMSO-d 6 ) 58.50 - 8.40 (m, 2H), 8.18 (dd, J = 5.8, 9.3 Hz, 1H), 7.85 - 7.73 (m, 3H), 7.55 (d, J = 3.6 Hz, 1H), 7.34 (d, J = 8.2 Hz, 2H), 7.18 (d, J = 3.6 Hz, 1H), 2.64 (t, J = 7.7 Hz, 2H), 1.60 (p, 2H), 1.35 (h, J = 7.4 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 167.53, 159.96, 156.22, 155.86, 151.90, 148.07, 146.37, 143.39, 135.63, 135.54, 132.66, 129.48, 127.66, 124.56, 119.70, 114.19, 108.71, 34.98, 33.30, 22.07, 14.09. HRMS (ESI) calcd for C 24 H 21 FNO 3 [M+H] + , 390.1427; found 390.15016.

2-(5-(4-butylphenyl)furan-2-yl)-6-chloroquinoline-4-carbo xylic acid (6).

[00148] 1 H NMR (400 MHz, DMSO-d 6 ) 5 8.75 (d, J = 2.4 Hz, 1H), 8.45 (s, 1H), 8.11 (d, J = 9.0 Hz, 1H), 7.85 (dd, J = 9.0, 2.4 Hz, 1H), 7.82 (d, J = 8.2 Hz, 2H), 7.58 (d, J = 3.6 Hz, 1H), 7.34 (d, J = 8.2 Hz, 2H), 7.19 (d, J = 3.6 Hz, 1 H), 2.64 (t, J = 7.7 Hz, 2H), 1.60 (p, J = 7.8, 6.5 Hz, 2H), 1.35 (h, J = 7.3 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 167.47, 158.56, 156.13, 151.84, 148.85, 148.82, 147.55, 147.22, 143.56, 136.86, 132.50, 132.30, 131.80, 131.39, 130.15, 129.51, 128.80, 127.60, 124.62, 108.82, 34.98, 33.30, 22.06, 14.09. HRMS (ESI) calcd for C 24 H 21 CINO 3 [M+H] + , 406.1132; found 406.12030. 2-(5-(4-butylphenyl)furan-2-yl)-6-iodoquinoline-4-carboxylic acid (7).

[00149] 1 H NMR (400 MHz, DMSO-d 6 ) 59.12 (d, J = 2.0 Hz, 1H), 8.29 (s, 1H), 8.03 (dd, J = 2.0, 8.8 Hz, 1 H), 7.85 - 7.77 (m, 3H), 7.52 (d, J = 3.6 Hz, 1 H), 7.37 - 7.30 (m, 2H), 7.17 (d, J = 3.6 Hz, 1 H), 2.64 (t, J = 7.7 Hz, 2H), 1.60 (p, J = 6.4, 7.9 Hz, 2H), 1.35 (h, J = 7.4 Hz, 2H), 0.93 (t, J = 7.4 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 169.98, 161.77, 159.26, 159.12, 157.73, 156.58, 155.91, 152.49, 150.82, 147.57, 137.77, 136.44, 135.21, 133.19, 129.56, 127.47, 126.30, 124.55, 102.95, 95.87, 35.05, 33.28, 22.12, 14.10. HRMS (ESI) calcd for C 24 H 21 INO 3 [M+H] + , 498.0488; found 498.05611.

2-(5-(4-butylphenyl)furan-2-yl)-7-fluoroquinoline-4-carbo xylic acid (8).

[00150] 1 H NMR (400 MHz, DMSO-d 6 ) 5 8.72 (dd, J = 6.4, 9.4 Hz, 1H), 8.37 (s, 1 H), 7.87 - 7.80 (m, 3H), 7.64 - 7.59 (m, 1H), 7.58 (d, J = 3.6 Hz, 1H), 7.37 - 7.33 (m, 2H), 7.20 (d, J = 3.6 Hz, 1 H), 2.65 (t, J = 7.7 Hz, 2H), 1.61 (p, 2H), 1.35 (h, J = 7.4, 14.6 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 170.45, 167.12, 162.98, 147.68, 142.28, 139.90, 133.70, 131.56, 129.53, 129.10, 126.09, 124.64, 120.97, 119.26, 117.32, 116.36, 114.52, 112.69, 111.73, 108.75, 33.35, 23.26, 22.11, 14.09. HRMS (ESI) calcd for C 24 H 21 FNO 3 [M+H] + , 390.1427; found 390.15007.

2-(5-(4-butylphenyl)furan-2-yl)-7-chloroquinoline-4-carbo xylic acid (9).

[00151] 1 H NMR (400 MHz, DMSO-d 6 ) 58.68 (d, J = 9.1 Hz, 1H), 8.36 (s, 1H), 8.12 (d, J = 2.2 Hz, 1H), 7.85 - 7.79 (m, 2H), 7.68 (dd, J = 2.2, 9.1 Hz, 1H), 7.57 (d, J = 3.6 Hz, 1H), 7.34 (d, J = 8.3 Hz, 2H), 7.19 (d, J = 3.6 Hz, 1H), 2.64 (t, J = 7.7 Hz, 2H), 1.60 (p, 2H), 1.35 (h, J = 7.3 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (101 MHz, DMSO-d 6 ) 5 171.77, 171.16, 165.59, 156.05, 152.01, 143.42, 141.87, 135.36, 133.98, 129.44, 125.57, 124.59, 124.24, 124.19, 122.56, 108.80, 35.11 , 33.43, 22.22, 14.25. HRMS (ESI) calcd for C 24 H 21 CINO 3 [M+H] + , 406.1132; found 406.12045.

7-bromo-2-(5-(4-butylphenyl)furan-2-yl)quinoline-4-carbox ylic acid (10).

[00152] 1 H NMR (400 MHz, DMSO-d 6 ) 58.60 (d, J = 9.1 Hz, 1H), 8.40 (s, 1H), 8.29 (d, J = 2.1 Hz, 1H), 7.85 - 7.77 (m, 3H), 7.58 (d, J = 3.6 Hz, 1H), 7.36 - 7.30 (m, 2H), 7.19 (d, J = 3.6 Hz, 1H), 2.64 (t, J = 7.7 Hz, 2H), 1.60 (p, 2H), 1.35 (h, J = 7.3 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 169.50, 166.72, 163.77, 156.23, 151.82, 149.55, 143.53, 133.96, 131.48, 130.85, 129.49, 127.59, 124.65, 124.31, 122.70, 118.95, 114.99, 112.56, 109.83, 108.86, 107.45, 22.09, 14.10. HRMS (ESI) calcd for C 24 H 21 BrNO 3 [M+H] + , 450.0627; found 450.07013.

2-(5-(4-butylphenyl)furan-2-yl)-8-chloroquinoline-4-carbo xylic acid (11).

[00153] 1 H NMR (400 MHz, DMSO-d 6 ) 5 8.56 (dd, J = 1.3, 8.5 Hz, 1H), 8.42 (s, 1 H), 8.02 (dd, J = 1.3, 7.5 Hz, 1 H), 7.87 - 7.80 (m, 2H), 7.66 - 7.55 (m, 2H), 7.38 - 7.31 (m, 2H), 7.21 (d, J = 3.6 Hz, 1H), 2.64 (t, J = 7.7 Hz, 2H), 1.60 (p, 2H), 1.35 (h, J = 7.3 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (101 MHz, DMSO-d 6 ) 5 167.65, 156.08, 151.94, 151.31, 148.72, 143.42, 133.44, 132.93, 130.96, 129.46, 127.72, 127.59, 125.48, 125.19, 124.58, 114.57, 108.83, 35.11, 33.41, 22.21 , 14.25. HRMS (ESI) calcd for C 24 H 21 CINO 3 [M+H] + , 406.1132; found 406.12057.

8-bromo-2-(5-(4-butylphenyl)furan-2-yl)quinoline-4-carbox ylic acid (12).

[00154] 1 H NMR (400 MHz, DMSO-d 6 ) 5 8.60 (dd, J = 1.3, 8.5 Hz, 1H), 8.43 (s, 1 H), 8.23 (dd, J = 1.2, 7.5 Hz, 1 H), 7.88 - 7.81 (m, 2H), 7.62 - 7.52 (m, 2H), 7.35 (d, J =

8.2 Hz, 2H), 7.22 (d, J = 3.6 Hz, 1H), 2.65 (t, J = 7.7 Hz, 2H), 1.60 (p, 2H), 1.35 (h, J =

7.3 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 166.44, 160.74, 158.08, 153.50, 141.93, 138.26, 132.86, 129.52, 127.63, 126.21, 125.37, 124.62, 123.11, 120.36, 119.11 , 111.78, 108.86, 35.02, 33.30, 24.87, 22.14, 16.74, 14.10. HRMS (ESI) calcd for C 24 H 21 BrNO 3 [M+H] + , 450.0627; found 450.06985.

2-(5-(4-butylphenyl)furan-2-yl)-6,7-difluoroquinoline-4-c arboxylic acid (13).

[00155] 1 H NMR (400 MHz, DMSO-d 6 ) 5 8.69 (dd, J = 9.1, 12.7 Hz, 1H), 8.44 (s, 1 H), 8.11 (dd, J = 8.1 , 11.6 Hz, 1H), 7.82 (d, 2H), 7.56 (d, J = 3.6 Hz, 1H), 7.35 (d, 2H), 7.19 (d, J = 3.6 Hz, 1H), 2.65 (t, J = 7.7 Hz, 2H), 1.60 (p, 2H), 1.35 (h, J = 7.3 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). HRMS (ESI) calcd for C 24 H 20 F 2 NO 3 [M+H] + , 408.1333; found 408.14047.

2-(5-(4-butylphenyl)furan-2-yl)-6,7-dichloroquinoline-4-c arboxylic acid (14).

[00156] 1 H NMR (400 MHz, DMSO-d 6 ) 5 8.99 (s, 1H), 8.38 (s, 1H), 8.30 (s, 1H), 7.85 - 7.76 (m, 2H), 7.56 (d, J = 3.6 Hz, 1 H), 7.35 - 7.29 (m, 2H), 7.18 (d, J = 3.6 Hz, 1 H), 2.64 (t, J = 7.7 Hz, 2H), 1.60 (p, 2H), 1.34 (h, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 156.32, 155.57, 154.21, 153.17, 151.81, 149.86, 148.04, 143.90, 143.54, 140.10, 134.76, 133.39, 130.12, 129.49, 127.53, 126.02, 124.64, 109.81, 38.91, 34.99, 33.29, 22.08, 14.09, 9.28. HRMS (ESI) calcd for C 24 H 20 CI 2 NO 3 [M+H] + , 440.0742; found 440.08140. 2-(5-(4-butylphenyl)furan-2-yl)-6,8-dichloroquinoline-4-carb oxylic acid (15).

[00157] 1 H NMR (400 MHz, DMSO-d 6 ) 58.71 (d, J = 2.3 Hz, 1H), 8.49 (s, 1H), 8.16 (d, J = 2.3 Hz, 1H), 7.83 (d, J = 8.2 Hz, 2H), 7.61 (d, J = 3.6 Hz, 1H), 7.36 - 7.31 (m, 2H), 7.21 (d, J = 3.6 Hz, 1 H), 2.64 (t, J = 7.7 Hz, 2H), 1.60 (p, J = 6.4, 7.8 Hz, 2H), 1.35 (h, J = 7.3 Hz, 2H), 0.93 (t, J = 7.4 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 174.34, 167.20, 156.43, 151.73, 147.06, 144.46, 140.42, 137.91, 136.29, 134.49, 133.78, 130.93, 129.54, 127.50, 125.50, 124.66, 124.46, 121.29, 109.80, 34.99, 33.29, 22.03, 14.06. HRMS (ESI) calcd for C 2 4H 20 CI 2 NO3[M+H] + , 440.0742; found 440.08151.

2-(5-(4-butylphenyl)furan-2-yl)-6-methylquinoline-4-carbo xylic acid (16).

[00158] 1 H NMR (400 MHz, DMSO-d 6 ) 5 8.39 (s, 1H), 8.32 (s, 1H), 8.00 (d, J = 8.6 Hz, 1H), 7.84 - 7.76 (m, 2H), 7.68 (dd, J = 2.0, 8.7 Hz, 1H), 7.50 (d, J = 3.6 Hz, 1H), 7.33 (d, J = 8.1 Hz, 2H), 7.16 (d, J = 3.6 Hz, 1H), 2.64 (t, J = 7.7 Hz, 2H), 2.55 (s, 3H), 1.60 (p, J = 6.4, 7.8 Hz, 2H), 1.35 (h, J = 7.3 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 170.31, 168.15, 160.76, 157.84, 155.58, 152.29, 147.64, 143.32, 130.25, 130.03, 129.48, 127.74, 126.83, 126.32, 125.41, 125.30, 125.21, 124.86, 124.50, 108.68, 34.98, 33.32, 22.07, 21.91, 14.10. HRMS (ESI) calcd for C 25 H 24 NO 3 [M+H] + , 386.1678; found 386.17500.

2-(5-(4-butylphenyl)furan-2-yl)-6-ethylquinoline-4-carbox ylic acid (17).

[00159] 1 H NMR (400 MHz, DMSO-d 6 ) 58.41 (d, J = 1.8 Hz, 1H), 8.32 (s, 1H), 8.02 (d, J = 8.6 Hz, 1H), 7.84 - 7.79 (m, 2H), 7.73 (dd, J = 2.0, 8.8 Hz, 1H), 7.50 (d, J = 3.6 Hz, 1H), 7.37 - 7.30 (m, 2H), 7.17 (d, J = 3.6 Hz, 1 H), 2.85 (q, J = 7.6 Hz, 2H), 2.64 (t, J = 7.7 Hz, 2H), 1.60 (p, 2H), 1.39 - 1.26 (m, 5H), 0.93 (t, J = 7.3 Hz, 3H). HRMS (ESI) calcd for C 26 H 26 NO 3 [M+H] + , 400.1834; found 400.19063.

2-(5-(4-butylphenyl)furan-2-yl)-6-isopropylquinoline-4-ca rboxylic acid (18).

[00160] 1 H NMR (400 MHz, DMSO-d 6 ) 58.44 (d, J = 1.9 Hz, 1 H), 8.33 (s, 1 H), 8.04 (d, J = 8.7 Hz, 1H), 7.84 - 7.76 (m, 3H), 7.50 (d, J = 3.6 Hz, 1H), 7.33 (d, J = 8.0 Hz, 2H), 7.17 (d, J = 3.6 Hz, 1 H), 3.13 (p, J = 6.8 Hz, 1 H), 2.64 (t, J = 7.7 Hz, 2H), 1.61 (p, 2H), 1.40 - 1.27 (m, 8H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 162.58, 155.55, 152.33, 150.68, 147.93, 147.72, 144.57, 143.24, 133.85, 129.72, 129.46, 127.76, 124.50, 121.40, 115.65, 113.60, 109.79, 108.63, 34.99, 34.13, 33.32, 23.99, 22.08, 14.10. HRMS (ESI) calcd for C 27 H 27 NO 3 [M+H] + , 414.1991; found 414.20633.

2-(5-(4-butylphenyl)furan-2-yl)-6-phenylquinoline-4-carbo xylic acid (19).

[00161] 1 H NMR (400 MHz, DMSO-d 6 ) 58.91 (d, J = 1.8 Hz, 1 H), 8.43 (s, 1 H), 8.21 - 8.12 (m, 2H), 7.82 (t, J = 7.9 Hz, 4H), 7.60 - 7.52 (m, 3H), 7.46 (t, J = 7.3 Hz, 1H), 7.35 (d, J = 8.0 Hz, 2H), 7.20 (d, J = 3.6 Hz, 1H), 2.65 (t, J = 7.7 Hz, 2H), 1.61 (p, J = 6.3, 7.9 Hz, 2H), 1.35 (h, J = 7.4 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 167.93, 155.86, 153.83, 152.16, 148.48, 145.50, 143.35, 141.66, 140.10, 140.05, 139.42, 135.58, 133.69, 131.06, 130.43, 130.40, 129.75, 129.50, 127.77,

127.62, 124.58, 124.14, 119.01, 117.68, 95.27, 35.00, 33.32, 22.09, 14.10. HRMS (ESI) calcd for C 30 H 26 NO 3 [M+H] + , 448.1834; found 448.19066.

2-(5-(4-butylphenyl)furan-2-yl)-6,8-dimethylquinoline-4-c arboxylic acid (20).

[00162] 1 H NMR (500 MHz, Chloroform-d) 5 8.29 (s, 1H), 8.19 (s, 1H), 7.84 - 7.79 (m, 2H), 7.55 (d, J = 1.8 Hz, 1H), 7.48 (d, J = 3.6 Hz, 1H), 7.36 - 7.31 (m, 2H), 7.17 (d, J = 3.5 Hz, 1H), 2.78 (s, 3H), 2.64 (t, J = 7.7 Hz, 2H), 2.49 (s, 3H), 1.60 (p, 2H), 1.34 (h, J = 7.4 Hz, 2H), 0.92 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, Chloroform-d) 5 163.04,

159.63, 158.80, 155.42, 140.38, 140.05, 139.61, 138.69, 137.46, 136.56, 130.26, 127.80, 127.33, 126.59, 126.27, 125.62, 124.47, 122.72, 122.10, 120.01, 109.76, 33.30, 22.08, 18.12, 14.10. HRMS (ESI) calcd for C 26 H 26 NO 3 [M+H] + , 400.1834; found 400.19099.

2-(5-(4-butylphenyl)furan-2-yl)-6-methoxyquinoline-4-carb oxylic acid (21).

[00163] 1 H NMR (500 MHz, DMSO-d 6 ) 5 8.37 (s, 1H), 8.14 (d, J = 2.8 Hz, 1H), 8.03 (d, J = 9.2 Hz, 1 H), 7.80 (d, J = 8.3 Hz, 2H), 7.51 (dd, J = 2.9, 9.2 Hz, 1 H), 7.44 (d, J = 3.6 Hz, 1H), 7.33 (d, J = 8.2 Hz, 2H), 7.15 (d, J = 3.5 Hz, 1H), 3.93 (s, 3H), 2.64 (t, J = 7.7 Hz, 2H), 1.59 (p, 2H), 1.34 (h, J = 7.3 Hz, 2H), 0.92 (t, J = 7.4 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 168.11 , 159.48, 159.01, 158.91, 158.13, 156.45, 155.29, 154.61, 153.27, 152.40, 149.48, 147.30, 143.17, 139.33, 134.22, 134.10, 132.87, 129.46, 127.03, 124.45, 120.90, 112.92, 109.81, 55.85, 22.08, 14.10. HRMS (ESI) calcd for C 25 H 24 NO 4 [M+H] + , 402.1627; found 402.17019.

2-(5-(4-butylphenyl)furan-2-yl)-7-methoxyquinoline-4-carb oxylic acid (22).

[00164] 1 H NMR (400 MHz, DMSO-d 6 ) 58.54 (d, J = 9.3 Hz, 1 H), 8.22 (s, 1 H), 7.82 (d, J = 8.3 Hz, 2H), 7.51 (d, J = 3.6 Hz, 1H), 7.48 (d, J = 2.7 Hz, 1H), 7.36 - 7.29 (m, 3H), 7.17 (d, J = 3.6 Hz, 1H), 3.98 (s, 3H), 2.65 (t, J = 7.7 Hz, 2H), 1.61 (p, 2H), 1.35 (h, J = 7.3 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 170.31, 168.15, 160.76, 157.84, 155.58, 152.29, 147.64, 143.32, 130.25, 130.03, 129.48, 127.74, 126.83, 126.32, 125.41, 125.30, 125.21, 124.86, 124.50, 108.68, 34.98, 33.32, 22.07, 21.91, 14.10. HRMS (ESI) calcd for C 25 H 24 NO4 [M+H] + , 402.1627; found 402.17014.

2-(5-(4-butylphenyl)furan-2-yl)-6-(trifluoromethoxy)quino line-4-carboxylic acid (23).

[00165] 1 H NMR (400 MHz, DMSO-d 6 ) 5 8.71 (dd, J = 1.3, 2.7 Hz, 1H), 8.49 (s, 1 H), 8.22 (d, J = 9.2 Hz, 1 H), 7.86 - 7.80 (m, 3H), 7.60 (d, J = 3.6 Hz, 1 H), 7.35 (d, J = 8.3 Hz, 2H), 7.20 (d, J = 3.6 Hz, 1H), 2.65 (t, J = 7.7 Hz, 2H), 1.61 (p, J = 7.4, 8.9 Hz, 2H), 1.35 (h, J = 7.3, 14.6 Hz, 2H), 0.93 (t, J = 7.4 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 167.44, 156.20, 151.78, 147.48, 146.29, 141.86, 140.47, 138.82, 138.20, 134.12, 133.00, 132.29, 130.55, 129.50, 127.66, 125.31, 124.63, 124.29, 119.50, 109.68, 108.80, 35.07, 33.31, 22.08, 14.09. HRMS (ESI) calcd for C 25 H 21 F 3 NO 4 [M+H] + , 456.1344; found 456.14191.

6-amino-2-(5-(4-butylphenyl)furan-2-yl)quinoline-4-carbox ylic acid (24).

[00166] 1 H NMR (400 MHz, DMSO-d 6 ) 58.19 (s, 1H), 7.81 - 7.74 (m, 3H), 7.65 (d, J = 2.5 Hz, 1H), 7.32 (d, J = 8.1 Hz, 2H), 7.27 (d, J = 3.5 Hz, 1H), 7.23 (dd, J = 2.5, 9.0 Hz, 1 H), 7.10 (d, J = 3.5 Hz, 1 H), 6.00 (s, 2H), 2.63 (t, J = 7.7 Hz, 2H), 1.60 (p, J = 6.4, 7.9 Hz, 2H), 1.35 (h, J = 7.4 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 168.60, 154.43, 146.07, 143.50, 130.82, 129.40, 127.89, 125.07, 124.20, 122.86, 122.12, 121.59, 121.28, 118.38, 116.90, 116.46, 106.38, 103.30, 72.65, 35.03, 33.34, 22.07, 14.10. HRMS (ESI) calcd for C 24 H 23 N 2 O 3 [M+H] + , 387.1630; found 387.17051.

6-acetamido-2-(5-(4-butylphenyl)furan-2-yl)quinoline-4-ca rboxylic acid (25).

[00167] 1 H NMR (500 MHz, DMSO-d 6 ) 5 10.39 (s, 1H), 8.87 (d, J = 2.3 Hz, 1H), 8.30 (s, 1H), 8.10 (dd, J = 2.3, 9.2 Hz, 1H), 8.03 (d, J = 9.1 Hz, 1H), 7.81 (d, J = 8.1 Hz, 2H), 7.47 (d, J = 3.6 Hz, 1H), 7.34 (d, J = 8.1 Hz, 2H), 7.16 (d, J = 3.6 Hz, 1H), 2.64 (t, J = 6.4, 8.8 Hz, 2H), 2.13 (s, 3H), 1.60 (p, J = 6.4, 7.8 Hz, 2H), 1.35 (h, J = 7.3 Hz, 2H), 0.93 (t, J = 7.4 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 163.41, 159.64, 150.89, 147.18, 140.92, 139.20, 138.91 , 135.76, 132.47, 131.65, 130.19, 124.49, 123.50,

121.72, 119.54, 117.80, 117.04, 111.02, 107.49, 105.46, 72.68, 33.18, 27.65, 22.12,

12.59, -3.71. HRMS (ESI) calcd for C 26 H 25 N 2 O 4 [M+H] + , 429.1736; found 429.18117.

2-(5-(4-butylphenyl)furan-2-yl)-7-(trifluoromethyl)quinol ine-4-carboxylic acid (26). [00168] 1 H NMR (400 MHz, DMSO-d 6 ) 5 8.90 - 8.84 (m, 1 H), 8.53 (s, 1 H), 8.41 (dt, J = 0.9, 1 .8 Hz, 1 H), 7.93 (dd, J = 2.0, 9.0 Hz, 1 H), 7.87 - 7.81 (m, 2H), 7.65 (d, J = 3.6 Hz, 1 H), 7.35 (d, J = 8.3 Hz, 2H), 7.22 (d, J = 3.6 Hz, 1 H), 2.65 (t, J = 7.7 Hz, 2H), 1.61 (p, J = 7.4, 8.9 Hz, 2H), 1 .35 (h, J = 7.3 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 167.43, 162.84, 160.79, 156.43, 151.58, 148.15, 145.79, 143.73, 137.52, 136.17, 134.04, 129.51 , 124.72, 118.21 , 116.99, 114.80, 111.57, 109.04, 60.72,

53.60, 35.00, 33.31 , 22.09, 14.10. HRMS (ESI) calcd for C 25 H 21 F 3 NO 3 [M+H] + , 440.1395; found 440.14691.

2-(5-(4-butylphenyl)furan-2-yl)-8-(trifluoromethyl)quinol ine-4-carboxylic acid (27).

[00169] 1 H NMR (400 MHz, DMSO-d 6 ) 5 8.89 (dd, J = 1.3, 8.7 Hz, 1 H), 8.49 (s, 1 H), 8.26 (d, J = 7.3 Hz, 1 H), 7.87 - 7.81 (m, 2H), 7.81 - 7.75 (m, 1 H), 7.55 (d, J = 3.6 Hz, 1 H), 7.38 - 7.32 (m, 2H), 7.22 (d, J = 3.6 Hz, 1 H), 2.65 (t, J = 7.7 Hz, 2H), 1.60 (p, 2H), 1.35 (h, J = 7.3 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 167.64, 156.48, 154.92, 151.84, 143.59, 141.98, 139.96, 138.59, 129.53, 127.62,

126.72, 125.83, 124.62, 124.22, 121.60, 115.22, 111.68, 109.81 , 108.88, 100.02, 34.99, 33.29, 22.06, 14.10. HRMS (ESI) calcd for C 25 H 21 F 3 NO 3 [M+H] + , 440.1395; found 440.14700.

2-(5-(4-butylphenyl)furan-2-yl)-6-nitroquinoline-4-carbox ylic acid (compound 28).

[00170] 1 H NMR (400 MHz, DMSO-d 6 ) 5 9.66 (d, J = 2.6 Hz, 1 H), 8.56 (s, 1 H), 8.51 (dd, J = 2.6, 9.3 Hz, 1 H), 8.25 (d, J = 9.2 Hz, 1 H), 7.84 (d, 2H), 7.74 (d, J = 3.7 Hz, 1 H), 7.36 (d, 2H), 7.25 (d, J = 3.7 Hz, 1 H), 2.65 (t, J = 7.7 Hz, 2H), 1 .61 (p, 2H), 1 .35 (h, J = 7.3 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 167.06, 157.10, 153.68, 151.55, 151.36, 145.69, 143.88, 139.76, 131.39, 129.53, 127.37, 124.83,

124.19, 122.82, 120.73, 101.53, 35.01 , 33.28, 22.08, 14.09. HRMS (ESI) calcd for C 24 H 21 N 2 O 5 [M+H] + , 417.1372; found 417.14449.

2-(5-(4-butylphenyl)furan-2-yl)-8-nitroquinoline-4-carbox ylic acid (30).

[00171] 1 H NMR (400 MHz, DMSO-d 6 ) 5 8.86 (dd, J = 1.3, 8.6 Hz, 1 H), 8.52 (s, 1 H), 8.32 (dd, J = 1.3, 7.6 Hz, 1 H), 7.86 - 7.76 (m, 3H), 7.56 (d, J = 3.6 Hz, 1 H), 7.35 (d, J = 8.1 Hz, 2H), 7.22 (d, J = 3.6 Hz, 1H), 2.65 (t, J = 7.6 Hz, 2H), 1.61 (p, 2H), 1.34 (h, J = 7.3 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 167.30, 163.02, 156.75, 153.84, 151.40, 149.92, 148.55, 144.16, 139.94, 138.64, 135.04, 129.55, 127.34, 124.75, 114.50, 109.29, 91.76, 77.75, 65.22, 35.00, 33.27, 22.06, 14.09, 2.71. HRMS (ESI) calcd for C 24 H 21 N 2 O 5 [M+H] + , 417.1372; found 417.14440.

2-(5-(4-butylphenyl)thiophen-2-yl)-6-nitroquinoline-4-car boxylic acid (31).

[00172] 1 H NMR (400 MHz, DMSO) 5 9.72 (d, J = 2.7 Hz, 1H), 8.43 (s, 1 H), 8.42 (dd, J = 9.2, 2.7 Hz, 1H), 8.17 (d, J = 4.0 Hz, 1H), 8.12 (d, J = 9.2 Hz, 1H), 7.72 (d, J = 8.0 Hz, 2H), 7.62 (d, J = 4.1 Hz, 1H), 7.30 (d, J = 8.0 Hz, 2H), 2.63 (t, J = 7.6 Hz, 2H), 1.59 (p, 2H), 1.34 (h, J = 7.4 Hz, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO) 5 167.29, 155.29, 150.91, 148.87, 145.80, 144.99, 143.57, 142.66, 131.23, 130.85, 130.63, 129.64, 126.09, 125.38, 124.40, 123.59, 118.95, 34.99, 33.40, 22.20, 14.25. HRMS (ESI) calcd for C 24 H 21 N 2 O 4 S [M+H] + 433.1144; found 433.1215.

2-(5-(4-butylphenyl)-1 /7-pyrrol-2-yl)-6-nitroquinoline-4-carboxylic acid (32).

[00173] 1 H NMR (400 MHz, DMSO) 5 12.08 (s, 1H), 9.64 (d, J = 2.6 Hz, 1H), 8.49 (s, 1H) 8.46 (dd, J = 9.2, 2.7 Hz, 1 H), 8.12 (d, J = 9.2 Hz, 1H), 7.76 - 7.45 (m, 4H), 7.17 (d, J = 8.0 Hz, 2H), 2.58 (t, J = 7.6 Hz, 2H), 1.57 (p, 2H), 1.34 (h, J = 7.3 Hz, 2H), 0.92 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO) 5 187.65, 172.54, 167.36, 153.40,

151.41, 144.71, 140.17, 132.64, 131.84, 130.47, 129.05, 126.74, 125.52, 123.87,

122.41, 121.90, 114.03, 110.52, 34.96, 33.64, 22.21 , 14.26. HRMS (ESI) calcd for C^H^NJ [M+H] + 416.1532; found 416.1605.

2-(4'-butyl-[1 ,1 *-biphenyl]-3-yl)-6-nitroquinoline-4-carboxylic acid (33).

[00174] 1 H NMR (400 MHz, DMSO) 59.84 (d, J = 2.7 Hz, 1 H), 8.56 - 8.51 (m, 2H), 8.47 (dd, J = 9.2, 2.7 Hz, 1H), 8.28 (dd, J = 8.9, 2.3 Hz, 2H), 7.84 (d, J = 8.0 Hz, 1H), 7.72 (d, J = 8.0 Hz, 2H), 7.67 (t, J = 7.7 Hz, 1 H), 7.35 (d, J = 8.0 Hz, 2H), 2.66 (t, J = 7.7 Hz, 2H), 1.61 (p, J = 7.6 Hz, 2H), 1.36 (h, J = 7.3 Hz, 2H), 0.94 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO) 5 167.77, 164.86, 159.84, 150.33, 145.50, 142.41, 141.44, 139.08, 137.88, 131.61, 129.65, 129.14, 127.84, 127.46, 124.74, 124.67, 124.14, 123.53, 120.21, 35.13, 33.85, 22.30, 14.67. HRMS (ESI) calcd for C 26 H 23 N 2 O 4 [M+H] + 427.1580; found 427.1653. 2-(6-(4-butylphenyl)pyridin-2-yl)-6-nitroquinoline-4-carboxy lic acid (34).

[00175] 1 H NMR (400 MHz, DMSO) 59.88 (d, J = 2.7 Hz, 1H), 9.14 (s, 1 H), 8.59 (t, J = 4.4 Hz, 1H), 8.51 (dd, J = 9.2, 2.7 Hz, 1H), 8.34 (d, J = 9.2 Hz, 1H), 8.17 (d, J = 8.1 Hz, 2H), 8.15 - 8.12 (m, 2H), 7.41 (d, J = 8.1 Hz, 2H), 2.68 (t, J = 7.7 Hz, 2H), 1.64 (p, J = 7.6 Hz, 2H), 1.37 (h, J = 7.4 Hz, 2H), 0.94 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO) 5 167.53, 159.18, 156.47, 154.34, 150.63, 149.19, 145.88, 144.42, 139.15, 136.21, 134.77, 131.85, 129.39, 127.09, 124.44, 124.38, 123.40, 122.06, 120.53, 116.81, 107.38, 35.07, 33.45, 22.24, 14.27. HRMS (ESI) calcd for C 25 H 22 N 3 O4 [M+H] + 428.1532; found 428.1603.

2-(5-(4-butylphenyl)pyridin-3-yl)-6-nitroquinoline-4-carb oxylic acid (35).

[00176] 1 H NMR (400 MHz, DMSO) 5 9.75 (d, J = 2.6 Hz, 1 H), 9.45 (d, J = 2.1 Hz, 1 H), 9.04 (d, J = 2.2 Hz, 1 H), 8.86 (t, J = 2.2 Hz, 1 H), 8.71 (s, 1 H), 8.52 (dd, J = 9.3, 2.7 Hz, 1H), 8.35 (d, J = 9.3 Hz, 1H), 7.82 (d, J = 8.0 Hz, 2H), 7.38 (d, J = 8.1 Hz, 2H), 2.68 (t, J = 7.6 Hz, 2H), 1.63 (p, J = 7.6 Hz, 2H), 1.36 (h, J = 7.4 Hz, 2H), 0.94 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO) 5 167.47, 157.65, 150.94, 149.43, 147.61, 146.10, 143.39, 139.69, 132.97, 129.65, 127.46, 124.14, 123.83, 120.51, 113.49, 101.56, 61.93, 40.57, 39.36, 35.13, 33.85, 22.00, 14.37. HRMS (ESI) calcd for C 25 H 22 N 3 O 4 [M+H] + 428.1532; found 428.1604.

2-(4'-butyl-[1 ,1 *-biphenyl]-4-yl)-6-nitroquinoline-4-carboxylic acid (36).

[00177] 1 H NMR (400 MHz, DMSO) 59.98 (d, J = 2.7 Hz, 1 H), 8.43 - 8.35 (m, 4H), 8.19 (d, J = 9.2 Hz, 1H), 7.88 (d, J = 8.0 Hz, 2H), 7.71 (d, J = 8.0 Hz, 2H), 7.34 (d, J = 8.1 Hz, 2H), 2.66 (t, J = 7.7 Hz, 2H), 1.62 (p, J = 7.6 Hz, 2H), 1.36 (h, J = 7.4 Hz, 2H), 0.94 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO) 5 167.48, 165.47, 159.27, 152.29, 150.98, 144.70, 142.74, 142.22, 137.33, 131.14, 129.49, 127.43, 127.09, 125.78, 124.66, 122.68, 118.67, 34.94, 33.53, 22.24, 14.27. HRMS (ESI) calcd for C 26 H 23 N 2 O 4 [M+H] + 427.1580; found 427.1652.

2-(6-(4-butylphenyl)pyridin-3-yl)-6-nitroquinoline-4-carb oxylic acid (37).

[00178] 1 H NMR (400 MHz, DMSO) 5 9.89 (d, J = 2.7 Hz, 1 H), 9.54 (d, J = 2.4 Hz, 1 H), 8.74 (dd, J = 8.4, 2.4 Hz, 1 H), 8.45 (dd, J = 9.2, 2.8 Hz, 1 H), 8.44 (s, 1H), 8.25 (d, J = 9.3 Hz, 1H), 8.18 - 8.09 (m, 3H), 7.37 (d, J = 8.1 Hz, 2H), 2.68 (t, J = 7.7 Hz, 2H), 1 .63 (p, J = 7.5 Hz, 2H), 1.36 (h, J = 7.4 Hz, 2H), 0.94 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO) 5 167.47, 161.35, 157.96, 150.94, 149.12, 145.50, 136.67, 136.06, 132.16, 131.76, 129.35, 126.55, 125.04, 123.23, 120.51, 119.45, 119.23, 115.30, 35.13, 33.55, 22.60, 14.07. HRMS (ESI) calcd for C 25 H 22 N 3 O4 [M+H] + 428.1532; found 428.1604.

6-nitro-2-(5-phenylfuran-2-yl)quinoline-4-carboxylic acid (38).

[00179] 1 H NMR (400 MHz, DMSO-d 6 ) 59.71 (d, J = 2.7 Hz, 1 H), 8.49 (s, 1 H), 8.47 (dd, J = 9.2, 2.4 Hz, 1 H), 8.22 (d, J = 9.4 Hz, 1 H), 7.93 (d, J = 8.0 Hz, 2H), 7.70 (d, J =

3.6 Hz, 1H), 7.53 (t, J = 7.6 Hz, 2H), 7.41 (t, J = 7.4 Hz, 1H), 7.30 (d, J = 3.7 Hz, 1H). 13 C NMR (126 MHz, DMSO) 5 167.17, 156.45, 152.22, 151.62, 150.94, 145.50, 145.20, 131.46, 129.95, 129.35, 125.04, 124.14, 123.53, 120.21, 119.83, 116.21, 110.10. HRMS (ESI) calcd for C 20 H 13 N 2 O 5 [M+H] + 361.0746; found 361.0819.

6-nitro-2-(5-(4-propylphenyl)furan-2-yl)quinoline-4-carbo xylic acid (39).

[00180] 1 H NMR (400 MHz, DMSO-d 6 ) 59.70 (d, J = 2.6 Hz, 1 H), 8.50 (s, 1 H), 8.48 (dd, J = 9.2, 2.4 Hz, 1 H), 8.22 (d, J = 9.3 Hz, 1 H), 7.87 - 7.82 (m, 2H), 7.70 (d, J = 3.7 Hz, 1H), 7.35 (d, J = 8.0 Hz 2H), 7.24 (d, J = 3.6 Hz, 1H), 2.63 (t, J = 7.5 Hz, 2H), 1.65 (h, 2H), 0.94 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO) 5 167.47, 156.75, 151.62, 151.21, 145.50, 143.99, 131.22, 129.65, 127.46, 124.74, 120.51, 116.81, 116.51, 109.19, 40.27, 39.96, 37.85, 37.53, 24.41, 14.07. HRMS (ESI) calcd for C 23 H 19 N 2 O 5 [M+H] + 403.1216; found 403.1288.

6-nitro-2-(5-(4-pentylphenyl)furan-2-yl)quinoline-4-carbo xylic acid (40).

[00181] 1 H NMR (400 MHz, DMSO-d 6 ) 5 9.74 (d, J = 2.6 Hz, 1 H), 8.47 (dd, J = 9.2, 2.4 Hz, 1H), 8.45 (s, 1H), 8.21 (d, J = 9.3 Hz, 1H), 7.84 (d, J = 8.0 Hz, 2H), 7.69 (d, J =

3.6 Hz, 1H), 7.35 (d, J = 8.0 Hz, 2H), 7.23 (d, J = 3.6 Hz, 1 H), 2.64 (t, J = 7.7 Hz, 2H), 1.63 (p, J = 7.5 Hz, 2H), 1.41 - 1.26 (m, 5H), 0.89 (t, J = 6.8 Hz, 3H). 13 C NMR (126 MHz, DMSO) 5 167.10, 163.55, 156.74, 151.86, 151.39, 145.26, 143.73, 135.10, 131.10, 129.51, 127.46, 123.78, 116.25, 109.14, 106.77, 40.58, 38.15, 35.41, 31.36, 30.93, 22.42, 14.41. HRMS (ESI) calcd for C 25 H 23 N 2 O 5 [M+H] + 431.1529; found 431.1601.

6-nitro-2-(5-(4-propoxyphenyl)furan-2-yl)quinoline-4-carb oxylic acid (41).

[00182] 1 H NMR (400 MHz, DMSO) 5 9.72 (d, J = 2.7 Hz, 1H), 8.46 (dd, J = 9.3,

2.7 Hz, 1H), 8.44 (s, 1H), 8.19 (d, J = 9.3 Hz, 1H), 7.88 - 7.81 (m, 2H), 7.66 (d, J = 3.6 Hz, 1H), 7.14 (d, J = 3.7 Hz, 1H), 7.08 (d, J = 8.0 Hz, 2H), 4.01 (t, J = 6.5 Hz, 2H), 1.78 (h, J = 7.1 Hz, 2H), 1.02 (t, J = 7.4 Hz, 3H). 13 C NMR (126 MHz, DMSO) 5 167.16, 159.65, 156.82, 151.41, 145.19, 141.80, 132.67, 131.04, 126.39, 123.91, 123.21, 122.51, 119.70, 119.53, 116.47, 115.52, 108.12, 69.59, 22.49, 10.87. HRMS (ESI) calcd for C 23 H 19 N 2 O 6 [M+H] + 419.1165; found 419.1236.

6-nitro-2-(5-(3-propoxyphenyl)furan-2-yl)quinoline-4-carb oxylic acid (42).

[00183] 1 H NMR (400 MHz, DMSO) 5 9.86 (d, J = 2.7 Hz, 1H), 8.39 (dd, J = 9.2, 2.7 Hz, 1H), 8.27 (s, 1H), 8.13 (d, J = 9.3 Hz, 1H), 7.60 (d, J = 3.6 Hz, 1H), 7.50 (d, J = 8.0, 1H), 7.46 - 7.39 (m, 2H), 7.30 (d, J = 3.7 Hz, 1H), 6.97 (dd, J = 8.3, 2.5, Hz, 1H), 4.04 (t, J = 6.5 Hz, 2H), 1.80 (h, J = 7.1 Hz, 2H), 1.04 (t, J = 7.4, 3H). 13 C NMR (126 MHz, DMSO) 5 167.41, 159.68, 155.81, 152.64, 151.42, 151.11, 144.67, 131.26, 130.79, 130.69, 125.28, 124.19, 123.14, 117.86, 116.92, 115.28, 115.07, 110.52, 109.94, 69.59, 22.55, 10.94. HRMS (ESI) calcd for C 23 H 19 N 2 O 6 [M+H] + 419.1165; found 419.1237.

6-nitro-2-(5-(2-propoxyphenyl)furan-2-yl)quinoline-4-carb oxylic acid (43)

[00184] 1 H NMR (400 MHz, DMSO) 5 9.82 (d, J = 2.7 Hz, 1H), 8.41 (dd, J = 9.3, 2.7 Hz, 1H), 8.34 (s, 1H), 8.14 (d, J = 9.2 Hz, 1H), 8.02 (dd, J = 7.8, 1.7 Hz, 1 H), 7.62 (d, J = 3.6 Hz, 1H), 7.38 (ddd, J = 8.6, 7.3, 1.7 Hz, 1H), 7.22 - 7.10 (m, 3H), 4.14 (t, J = 6.4 Hz, 2H), 1.92 (h, J = 7.0 Hz, 2H), 1.10 (t, J = 7.4 Hz, 3H). 13 C NMR (126 MHz, DMSO) 5 167.67, 155.55, 152.87, 151.45, 151.13, 144.75, 136.79, 130.71, 130.20, 126.35, 125.34, 124.93, 123.92, 123.30, 121.25, 118.40, 115.49, 113.35, 112.98, 70.16, 22.56, 11.27. HRMS (ESI) calcd for C 23 H 19 N 2 O 6 [M+H] + 419.1165; found 419.1237.

2-(5-(3-((tert-butoxycarbonyl)amino)phenyl)furan-2-yl)-6- nitroquinoline-4-carboxyli c acid (44).

[00185] 1 H NMR (400 MHz, DMSO) 5 9.65 (d, J = 2.7 Hz, 1H), 9.60 (s, 1 H), 8.54 (s, 1 H), 8.51 (dd, J = 9.2, 2.6 Hz, 1 H), 8.24 (d, J = 9.2 Hz, 1 H), 7.83 (d, J = 8.6 Hz, 2H), 7.73 (d, J = 3.8 Hz, 1 H), 7.62 (d, J = 8.5 Hz, 2H), 7.17 (d, J = 3.7 Hz, 1 H), 1.52 (s, 9H). 13 C NMR (126 MHz, DMSO) 5 167.17, 157.05, 153.11, 152.82, 151.31, 145.80, 140.59, 138.78, 131.46, 125.65, 123.83, 123.53, 122.93, 122.63, 121.12, 118.93, 117.12, 108.89, 80.20, 28.72. HRMS (ESI) calcd for C 25 H 22 N 3 O 7 [M+H] + 476.1380; found 476.1452. 2-(5-(4-((tert-butoxycarbonyl)amino)phenyl)furan-2-yl)-6-nit roquinoline-4-carboxyli c acid (45).

[00186] 1 H NMR (400 MHz, DMSO) 5 9.68 (d, J = 2.6 Hz, 1H), 9.56 (s, 1 H), 8.54 (s, 1 H), 8.52 (dd, J = 9.2, 2.4, 1 H), 8.25 (d, J = 9.3 Hz, 1 H), 8.02 (t, J = 2.0 Hz, 1 H), 7.74 (d, J = 3.7 Hz, 1H), 7.52 (t, J = 7.2 Hz 2H), 7.40 (t, J = 7.9 Hz, 1H), 7.20 (d, J = 3.7 Hz, 1 H), 1.53 (s, 9H). 13 C NMR (126 MHz, DMSO) 5 166.97, 156.75, 153.30, 151.79, 151.19, 145.69, 140.75, 139.07, 131.36, 130.12, 124.21, 123.26, 122.91, 120.67, 118.88, 116.71, 114.07, 109.87, 79.80, 31.43, 28.72. HRMS (ESI) calcd for C 25 H 22 N 3 O 7 [M+H] + 476.1380; found 476.1452.

2-(5-(benzo[cf][1 ,3]dioxol-5-yl)furan-2-yl)-6-nitroquinoline-4-carboxylic acid (48).

[00187] 1 H NMR (400 MHz, DMSO-d 6 ) 5 9.81 (d, J = 2.7 Hz, 1 H), 8.40 (dd, J = 9.3, 2.7 Hz, 1H), 8.28 (s, 1H), 8.12 (d, J = 9.3 Hz, 1H), 7.58 (d, J = 3.6 Hz, 1H), 7.52 - 7.42 (m, 2H), 7.16 (d, J = 3.6 Hz, 1H), 7.07 (d, J = 8.1 Hz, 1 H), 6.12 (s, 2H). 13 C NMR (126 MHz, DMSO) 5 167.27, 156.02, 152.05, 151.17, 148.50, 148.22, 144.65, 130.67, 125.04, 124.44, 123.19, 118.89, 118.02, 115.54, 109.44, 108.61, 105.13, 101.94, 91.01, 68.65. HRMS (ESI) calcd for C 2 IH 13 N 2 O 7 [M+H] + 405.0645; found 405.0716.

2-(5-(naphthalen-1 -yl)furan-2-yl)-6-nitroquinoline-4-carboxylic acid (49).

[00188] 1 H NMR (400 MHz, DMSO) 5 9.69 (d, J = 2.6 Hz, 1 H), 8.56 (s, 1 H), 8.55 - 8.46 (m, 2H), 8.26 (d, J = 9.3 Hz, 1H), 8.07 (d, J = 8.0 Hz, 2H), 8.00 (dd, J = 7.2, 1.2 Hz, 1 H), 7.84 (d, J = 3.6 Hz, 1H), 7.74 - 7.60 (m, 3H), 7.29 (d, J = 3.6 Hz, 1H). 13 C NMR (126 MHz, DMSO) 5 166.86, 156.45, 152.22, 151.31, 145.50, 139.39, 134.18, 131.46, 130.86, 130.18, 129.65, 129.31, 127.92, 126.85, 126.55, 125.65, 124.44, 123.83, 123.23, 120.21, 116.51, 113.49. HRMS (ESI) calcd for C 24 H 15 N 2 O 5 [M+H] + 411.0903; found 411.0975.

2-(5-([1 ,T-biphenyl]-4-yl)furan-2-yl)-6-nitroquinoline-4-carboxylic acid (50).

[00189] 1 H NMR (400 MHz, DMSO-d 6 ) 5 9.77 (d, J = 2.7 Hz, 1 H), 8.44 (dd, J = 9.2, 2.8, 1H), 8.43 (s, 1 H), 8.19 (d, J = 9.2 Hz, 1 H), 8.01 (d, J = 8.8 Hz, 2H), 7.83 (d, J = 8.8 Hz, 2H), 7.75 (dd, J = 8.4, 1.6, 2H), 7.68 (d, J = 3.6 Hz, 1H), 7.50 (t, J = 7.2, 2H), 7.41 (t, J = 7.2, 1H), 7.34 (d, J = 3.7 Hz, 1H). 13 C NMR (126 MHz, DMSO) 5 167.25, 156.02, 152.39, 151.17, 145.12, 140.50, 139.77, 131.01, 129.51, 127.75, 127.05, 125.26, 124.33, 123.60, 119.21, 117.73, 116.04, 109.98. HRMS (ESI) calcd for C 26 H 17 N 2 O 5 [M+H] + 437.1059; found 437.1131.

2-(5-([1,T-biphenyl]-3-yl)furan-2-yl)-6-nitroquinoline-4- carboxylic acid (51).

[00190] 1 H NMR (400 MHz, DMSO-d 6 ) 59.68 (d, J = 2.6 Hz, 1 H), 8.58 (s, 1 H), 8.51 (dd, J = 9.3, 2.7 Hz, 1 H), 8.27 (d, J = 9.4 Hz, 1 H), 8.18 (t, J = 2.0 Hz, 1 H), 7.95 (dt, J = 7.6, 1.4 Hz, 1H), 7.82 - 7.76 (m, 3H), 7.71 (dt, J = 7.8, 1.4 Hz, 1H), 7.64 (t, J = 7.8 Hz, 1 H), 7.54 (t, J = 7.2 Hz, 2H), 7.49 - 7.41 (m, 2H). 13 C NMR (126 MHz, DMSO) 5 167.02, 164.96, 156.57, 152.04, 151.31, 145.64, 143.73, 141.80, 139.99, 132.37, 130.47, 129.49, 127.44, 123.36, 123.02, 122.93, 120.58, 120.51, 116.69, 111.00, 110.39. HRMS (ESI) calcd for C 26 H 17 N 2 O 5 [M+H] + 437.1059; found 437.1131 .

6-nitro-2-(5-(4-phenoxyphenyl)furan-2-yl)quinoline-4-carb oxylic acid (52).

[00191] 1 H NMR (400 MHz, DMSO) 5 9.73 (d, J = 2.6 Hz, 1H), 8.46 (dd, J = 9.2,

2.6 Hz, 1H), 8.44 (s, 1H), 8.19 (d, J = 9.2 Hz, 1H), 7.95 (d, J = 8.7 Hz, 2H), 7.69 (d, J =

3.6 Hz, 1H), 7.45 (dd, J = 8.5, 7.3 Hz, 2H), 7.24 - 7.18 (m, 2H), 7.17 - 7.09 (m, 4H). 13 C NMR (126 MHz, DMSO) 5 198.27, 167.30, 161.96, 157.68, 156.47, 151.78, 151.28, 145.26, 143.01, 130.70, 126.74, 125.10, 124.50, 123.85, 123.23, 119.55, 116.41, 109.08, 40.34, 37.55. HRMS (ESI) calcd for C 26 H 17 N 2 O 6 [M+H] + 453.1008; found 453.1081.

6-nitro-2-(5-(3-phenoxyphenyl)furan-2-yl)quinoline-4-carb oxylic acid (53).

[00192] 1 H NMR (400 MHz, DMSO) 5 9.87 (d, J = 2.7 Hz, 1H), 8.39 (dd, J = 9.2,

2.7 Hz, 1H), 8.24 (s, 1H), 8.11 (d, J = 9.2 Hz, 1 H), 7.72 (d, J = 7.8 Hz, 1 H), 7.60 (t, J =

2.6 Hz, 2H), 7.54 (t, J = 8.0 Hz, 1H), 7.48 - 7.39 (m, 2H), 7.32 (d, J = 3.7 Hz, 1H), 7.19 (t, J = 7.4 Hz, 1H), 7.14 - 7.05 (m, 2H), 7.00 (dd, J = 8.2, 2.5 Hz, 1H). 13 C NMR (126 MHz, DMSO) 5 167.47, 157.96, 157.05, 154.94, 153.43, 152.71, 151.29, 150.94, 146.41, 145.20, 143.20, 131.76, 130.86, 124.44, 124.21, 123.53, 123.30, 118.93, 117.79, 115.29, 114.70, 110.40, 39.96. HRMS (ESI) calcd for C 26 H 17 N 2 O 6 [M+H] + 453.1008; found 453.1082.

6-nitro-2-(5-(2-phenoxyphenyl)furan-2-yl)quinoline-4-carb oxylic acid (54).

[00193] 1 H NMR (400 MHz, DMSO) 5 9.85 (d, J = 2.7 Hz, 1H), 8.40 (dd, J = 9.3,

2.7 Hz, 1H), 8.31 (s, 1H), 8.19 - 8.11 (m, 2H), 7.57 (d, J = 3.6 Hz, 1 H), 7.47 - 7.36 (m, 4H), 7.16 (t, J = 7.4 Hz, 1H), 7.11 - 7.01 (m, 4H). 13 C NMR (126 MHz, DMSO) 5 167.22, 156.75, 152.71, 152.19, 151.69, 151.32, 150.94, 144.79, 130.55, 130.35, 127.46, 127.16, 125.12, 123.99, 123.08, 121.72, 119.98, 118.32, 118.16, 114.93, 113.69, 87.67, 54.99. HRMS (ESI) calcd for C 26 H 17 N 2 O 6 [M+H] + 453.1008; found 453.1081.

General procedure for the production of ethyl 2-(5-(4-butylphenyl)furan-2-yl)quinoline-4-carboxylates (2, 29).

[00194] A 2-(5-(4-butylphenyl)furan-2-yl)quinoline-4-carboxylic acid (50 pmol) was dissolved in ethanol (2 ml_), stirred on ice, and H 2 SO 4 (120 pL) was added dropwise. The reaction was warmed to 75 °C and stirred overnight (Scheme 2). The reaction was cooled to room temperature, and a sodium carbonate solution was added until the mixture was basic. The mixture was extracted with ethyl acetate, dried (MgSO 4 ), filtered, and concentrated in vacuo to afford the product. ethyl 6-bromo-2-(5-(4-butylphenyl)furan-2-yl)quinoline-4-carboxyla te (2).

[00195] 1 H NMR (400 MHz, DMSO-d 6 ) 58.77 (d, J = 2.2 Hz, 1 H), 8.42 (s, 1 H), 8.06 - 7.95 (m, 2H), 7.82 (dd, J = 4.7, 8.2 Hz, 2H), 7.60 (d, J = 3.6 Hz, 1 H), 7.34 (dd, J = 3.7, 8.2 Hz, 2H), 7.20 (d, J = 3.6 Hz, 1H), 4.53 (q, J = 7.1 Hz, 2H), 2.65 (t, J = 7.7 Hz, 2H), 1.60 (p, J = 7.0, 8.5, 9.9 Hz, 2H), 1.46 (t, J = 7.1 Hz, 3H), 1.35 (h, 2H), 0.93 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, DMSO-d 6 ) 5 188.57, 184.37, 169.27, 168.65, 168.16, 167.92, 151.73, 135.76, 134.16, 132.66, 132.46, 132.30, 132.19, 131.92, 129.51, 127.55, 124.65, 121.40, 119.58, 109.79, 62.55, 37.27, 34.98, 33.25, 22.08, 14.09. HRMS (ESI) calcd for C 26 H 25 BrNO 3 [M+H] + , 478.0940; found 478.10111. ethyl 2-(5-(4-butylphenyl)furan-2-yl)-6-nitroquinoline-4-carboxyla te (29).

[00196] 1 H NMR (400 MHz, Chloroform-d) 5 9.74 (d, J = 2.5 Hz, 1H), 8.57 (s, 1H), 8.53 (dd, J = 2.5, 9.2 Hz, 1H), 8.29 (d, J = 9.5 Hz, 1H), 7.79 (d, J = 8.0 Hz, 2H), 7.63 (s, 1 H), 7.32 (d, J = 8.0 Hz, 2H), 6.91 (d, J = 3.6 Hz, 1H), 4.66 (q, J = 7.1 Hz, 2H), 2.70 (t, J = 7.7 Hz, 2H), 1.68 (p, J = 7.6 Hz, 2H), 1.60 (t, J = 7.1 Hz, 3H), 1.48 - 1.35 (m, 2H), 0.99 (t, J = 7.3 Hz, 3H). 13 C NMR (126 MHz, Chloroform-d) 5 172.17, 164.60, 159.70, 155.08, 150.83, 148.46, 147.46, 146.05, 144.28, 138.79, 137.91, 129.42, 127.13, 125.83, 122.96, 122.47, 119.95, 112.37, 104.91 , 98.48, 88.49, 58.49, 35.57, 33.21, 22.19, 13.77. HRMS (ESI) calcd for C 26 H 25 N 2 O 5 [M+H] + , 445.1685; found 445.17596.

General procedure for the production of 2-(5-(aminophenyl)furan-2-yl)-6-nitroquinoline-4-carboxylic acids (46, 47).

[00197] A 2-(5-(4-((fer -butoxycarbonyl)amino)phenyl)furan-2-yl)-6-nitroquinoline-4- carboxylic acid (50 pmol) was dissolved in a 1 ml_ solution of 50:50 TFA:dichloromethane. The reaction stirred at room temperature and was monitored by TLC (95:5 dichloromethane:methanol). When the reactant was consumed, toluene (2 ml_) was added, and the reaction was filtered and washed with methanol and diethyl ether to afford the product.

2-(5-(4-aminophenyl)furan-2-yl)-6-nitroquinoline-4-carbox ylic acid (46).

[00198] 1 H NMR (400 MHz, DMSO) 5 9.64 (d, J = 2.6 Hz, 1H), 8.51 (s, 1 H), 8.50 (dd, J = 9.3, 2.7 Hz 1 H), 8.21 (d, J = 9.2 Hz, 1 H), 7.70 (d, J = 3.7 Hz, 1 H), 7.63 (d, J = 8.5 Hz, 2H), 6.96 (d, J = 3.7 Hz, 1 H), 6.71 (d, J = 8.5 Hz, 2H). 13 C NMR (126 MHz, DMSO) 5 181.43, 170.79, 167.47, 160.15, 151.45, 144.90, 138.48, 134.48, 130.86, 126.55, 125.65, 122.63, 116.21, 106.77, 99.75, 98.24, 96.36. HRMS (ESI) calcd for C 20 H 14 N 3 O 5 [M+H] + 376.0855; found 376.0927.

2-(5-(3-aminophenyl)furan-2-yl)-6-nitroquinoline-4-carbox ylic acid (47).

[00199] 1 H NMR (500 MHz, CDCI 3 ) 5 9.69 (d, J = 2.6 Hz, 1H), 8.56 (s, 1H), 8.54 (dd, J = 9.3, 2.7 Hz, 1 H), 8.27 (d, J = 9.2 Hz, 1 H), 7.75 (dd, J = 3.7, 0.9 Hz, 1 H), 7.30 - 7.21 (m, 3H), 7.18 (dd, J = 3.6, 1.2 Hz, 1H), 6.76 (d, J = 6.8 Hz, 1H) 3.86 (s, 2H). 13 C NMR (126 MHz, DMSO) 5 185.65, 184.23, 182.42, 166.86, 151.56, 145.80, 139.08, 131.36, 130.30, 124.44, 123.53, 123.22, 122.93, 121.12, 116.51 , 109.79, 105.87, 103.07, 102.77. HRMS (ESI) calcd for C 2 oH 14 N 3 0 5 [M+H] + 376.0855; found 376.0927.

[00200] As used herein, the term “about” refers to plus or minus 10% of the referenced number. Although there has been shown and described the preferred embodiment of the present invention, it will be readily apparent to those skilled in the art that modifications may be made thereto which do not exceed the scope of the appended claims. Therefore, the scope of the invention is only to be limited by the following claims. In some embodiments, the figures presented in this patent application are drawn to scale, including the angles, ratios of dimensions, etc. In some embodiments, the figures are representative only and the claims are not limited by the dimensions of the figures. In some embodiments, descriptions of the inventions described herein using the phrase “comprising” includes embodiments that could be described as “consisting essentially of’ or “consisting of’, and as such the written description requirement for claiming one or more embodiments of the present invention using the phrase “consisting essentially of’ or “consisting of’ is met.