NZ792168A - Mitoketoscins: mitochondrial-based therapeutics targeting ketone metabolism in cancer cells - Google Patents

Mitoketoscins: mitochondrial-based therapeutics targeting ketone metabolism in cancer cells

Info

Publication number
NZ792168A
NZ792168A NZ792168A NZ79216818A NZ792168A NZ 792168 A NZ792168 A NZ 792168A NZ 792168 A NZ792168 A NZ 792168A NZ 79216818 A NZ79216818 A NZ 79216818A NZ 792168 A NZ792168 A NZ 792168A
Authority
NZ
New Zealand
Prior art keywords
mitoketoscin
alkenes
cyclic
alkanes
compounds
Prior art date
Application number
NZ792168A
Inventor
Michael P Lisanti
Federica Sotgia
Original Assignee
Lunella Biotech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Lunella Biotech Inc filed Critical Lunella Biotech Inc
Publication of NZ792168A publication Critical patent/NZ792168A/en

Links

Abstract

The present disclosure relates to compounds that bind to at least one of ACAT1/2 and OXCT1/2 and inhibit mitochondrial ATP production, referred to herein as mitoketoscins. Methods of screening compounds for mitochondrial inhibition and anti-cancer properties are disclosed. Also described are methods of using mitoketoscins to prevent or treat cancer, bacterial infections, and pathogenic yeast, as well as methods of using mitoketoscins to provide anti-aging benefits. Specific mitoketoscin compounds are also disclosed. of using mitoketoscins to prevent or treat cancer, bacterial infections, and pathogenic yeast, as well as methods of using mitoketoscins to provide anti-aging benefits. Specific mitoketoscin compounds are also disclosed.

Description

TOSCINS: MITOCHONDRIAL-BASED THERAPEUTICS TARGETING KETONE METABOLISM IN CANCER CELLS FIELD The present disclosure relates to mitoketoscins – non-carcinogenic compounds that bind to at least one of ACAT1/2 and OXCT1/2 and inhibit ondrial ATP production, as well as methods for identifying mitoketoscins, methods of using the inhibitors to target cancer stem cells, to target ia and pathogenic yeast, and to provide ging ts, and pharmaceutical compositions for treating cancer, bacterial infections, yeast infections, and aging, containing one or more mitoketoscins as the active ingredient.
BACKGROUND Researchers have struggled to develop new anti-cancer treatments. Conventional cancer therapies (e.g. irradiation, ting agents such as cyclophosphamide, and antimetabolites such as rouracil) have attempted to selectively detect and eradicate fast-growing cancer cells by interfering with cellular mechanisms involved in cell growth and DNA replication.
Other cancer therapies have used immunotherapies that selectively bind mutant tumor antigens on fast-growing cancer cells (e.g., monoclonal antibodies). Unfortunately, tumors often recur following these therapies at the same or ent site(s), indicating that not all cancer cells have been eradicated. Relapse may be due to insufficient chemotherapeutic dosage and/or emergence of cancer clones resistant to therapy. Hence, novel cancer treatment strategies are needed.
Ketones (3-hydroxybutyrate, acetoacetate and acetone) are high-energy mitochondrial fuels; they are naturally generated by hepatocytes during periods of caloric restriction, fasting, and/or starvation. During nutrient ation, ketone bodies secreted into the blood are then directed towards the brain, where neurons convert them back into -CoA so they may be effectively lized as an energy source. The two most critical neuronal enzymes for this ketone re-utilization process are OXCT1/2 and ACAT1/2, as they are directly involved in the conversion of ketone bodies into Acetyl-CoA. Martinez-Outschoorn et al., Nat Rev Clin Oncol 2017; 14(1):11-31.
The inventors showed that a r “ketone-shuttle” also exists in human tumors, y nic cancer-associated fibroblasts (CAFs) locally e ketone bodies for reutilization by mitochondria in adjacent human breast cancer cells. Martinez-Outschoorn, et al., Cell Cycle 2012; 11(21):3956-63. In further support of this “metabolic-coupling” hypothesis, the inventors found that recombinant over-expression of ACAT1/2 or OXCT1/2 in MDA-MB-231 breast cancer cells was indeed sufficient to promote tumor growth and lung metastasis. These data provide genetic ce that ketone body re-utilization may help drive tumor progression and metastasis.
SUMMARY In view of the foregoing background, it appears that the enzymes ACAT1/2 and OXCT1/2 may be bona-fide metabolic oncogenes. It is therefore an object of this disclosure to demonstrate that ketone re-utilization plays a critical role in the ation and maintenance of many cancers. It is also an object of this disclosure to present methods for identifying mitoketoscins, non-carcinogenic compounds that bind to at least one of ACAT1/2 and OXCT1/2 and inhibit mitochondrial ATP production. It is also an object of this disclosure to identify mitoketoscins having anti-cancer and antibiotic properties. It is also an object of this disclosure to identify mitoketoscins having anti-aging properties. It is also an object of this disclosure to mitoketoscins that function as radiosensitizers and photosensitizers. The term "mitoketoscin" broadly refers to non-carcinogenic compounds that bind to at least one of ACATI/2 and OXCTI/2 and inhibit mitochondrial ATP production. These compounds therefore are ed to target the ondrial enzymes sible for ketone re-utilization and that have anti-cancer and antibiotic properties. These compounds bind to either or both active tic sites of OXCTI/2 and ACATI/2 to inhibit mitochondrial function. The t disclosure further relates to methods of identifying mitoketoscins, methods of making such mitoketoscins, and methods of using mitoketoscins for therapeutic purposes.
Given their mitochondrial inhibition properties, mitoketoscins may similarly be used to target bacteria and enic yeast, provide anti-aging benefits, function as ensitizers and/or photo-sensitizers, sensitize bulk cancer cells and cancer stem cells to chemotherapeutic agents, pharmaceuticals, and/or other natural substances.
Mitoketoscins may be identified through a convergent approach of virtual highthroughput in silico screening followed by in vitro validation for mitochondrial inhibition.
Mitoketoscins may be rapidly developed by combining in silico drug design with phenotypic drug screening.
In a first aspect of the invention, there is provided a method of identifying a mitoketoscin using virtual high-throughput screening and phenotypic drug screening, the method comprising; identifying a compound that targets a mitochondrial ketone re-utilization enzyme using virtual hroughput screening; and testing the compound for mitochondrial inhibition activity.
In a second aspect of the invention, there is provided a mitoketoscin sing the general formula: , wherein R may be selected from the group consisting of chlorine, bromine, , carboxyl, alkanes, cyclic alkanes, alkenes, cyclic alkenes, alkynes, ketones, aldehydes, carboxylic acids, ethers, esters, amines, amides, clic or clic arene, arenes, phenols and benzoic acid.
In a third aspect of the invention, there is provided the use of a mitoketoscin in the manufacture of a medicament for treating cancer, wherein the mitoketoscin comprises the general formula: , wherein R may be selected from the group consisting of hydrogen, fluorine, chlorine, bromine, iodine, carboxyl, alkanes, cyclic alkanes, alkenes, cyclic alkenes, alkynes, ketones, aldehydes, carboxylic acids, ethers, esters, amines, , monocyclic or polycyclic arene, heteroarenes, phenols and benzoic acid.
In a fourth aspect of the invention, there is provided the use of a mitoketoscin in the manufacture of a ment for treating a microbial infection, wherein the toscin comprises the general formula: , wherein R may be selected from the group consisting of hydrogen, fluorine, chlorine, bromine, iodine, carboxyl, alkanes, cyclic alkanes, alkenes, cyclic alkenes, alkynes, ketones, aldehydes, carboxylic acids, ethers, esters, amines, amides, monocyclic or polycyclic arene, heteroarenes, phenols and benzoic acid.
In a fifth aspect of the invention, there is provided the use of a mitoketoscin in the manufacture of a medicament for treating an age-related ion, wherein the mitoketoscin comprises the general formula: , wherein R may be selected from the group consisting of hydrogen, ne, ne, e, iodine, carboxyl, alkanes, cyclic s, s, cyclic alkenes, alkynes, ketones, aldehydes, carboxylic acids, ethers, esters, amines, amides, monocyclic or polycyclic arene, heteroarenes, phenols and benzoic acid.
In a sixth aspect of the invention, there is provided a ceutical composition comprising, as an active ingredient, a mitoketoscin comprising the general formula: , wherein R may be selected from the group consisting of hydrogen, fluorine, chlorine, bromine, iodine, carboxyl, alkanes, cyclic alkanes, alkenes, cyclic alkenes, alkynes, ketones, aldehydes, carboxylic acids, ethers, esters, amines, , monocyclic or polycyclic arene, heteroarenes, phenols, benzoic acid; and a pharmaceutically acceptable carrier.
In a seventh aspect of the ion, there is provided a mitoketoscin comprising the general formula: , wherein R may be selected from the group consisting of hydrogen, , chlorine, bromine, iodine, carboxyl, alkanes, cyclic alkanes, alkenes, cyclic alkenes, alkynes, ketones, aldehydes, carboxylic acids, ethers, esters, , amides, monocyclic or polycyclic arene, heteroarenes, phenols and c acid.
In an eighth aspect of the ion, there is provided the use of a mitoketoscin in the manufacture of a medicament for treating cancer, wherein the mitoketoscin comprises the general formula: , n R may be selected from the group consisting of hydrogen, fluorine, chlorine, bromine, iodine, carboxyl, alkanes, cyclic alkanes, s, cyclic alkenes, alkynes, ketones, aldehydes, carboxylic acids, ethers, esters, , amides, clic or polycyclic arene, heteroarenes, s and benzoic acid.
In a ninth aspect of the invention, there is provided a pharmaceutical composition comprising, as an active ingredient, a mitoketoscin comprising the general formula: , wherein R may be selected from the group consisting of hydrogen, fluorine, chlorine, bromine, iodine, carboxyl, alkanes, cyclic alkanes, alkenes, cyclic alkenes, alkynes, ketones, aldehydes, carboxylic acids, ethers, esters, amines, amides, monocyclic or polycyclic arene, heteroarenes, phenols, benzoic acid; and a pharmaceutically acceptable carrier.
In one embodiment of the present invention, the mitoketoscin comprises the l formula: or a ceutically acceptable salt thereof.
In another embodiment, wherein the mitoketoscin comprises the general formula: or a pharmaceutically acceptable salt thereof, wherein Z is defined as ethylpiperidine or ethylpyrrolidine In a n embodiment, the mitoketoscin comprises the general formula: or a ceutically acceptable salt thereof, wherein Z is d as ethylpiperidine or ethylpyrrolidine.
In another embodiment, the mitoketoscin comprises the general formula: or a pharmaceutically acceptable salt thereof.
In one embodiment, the toscin comprises the general formula: or a pharmaceutically acceptable salt thereof.
In an embodiment, the mitoketoscin comprises the general pharmacophore: or a pharmaceutically acceptable salt thereof.
In one embodiment, the mitoketoscin ses the general pharmacophore: or a pharmaceutically acceptable salt thereof.
In another embodiment, the mitoketoscin comprises the general formula: or a pharmaceutically acceptable salt thereof.
In yet another embodiment, the mitoketoscin comprises the l formula: or a ceutically acceptable salt thereof.
In a n embodiment, the mitoketoscin ses the general formula: or a pharmaceutically able salt thereof.
In one embodiment, the mitoketoscin comprises the general formula: or a pharmaceutically acceptable salt thereof.
In another embodiment, the mitoketoscin comprises the general formula: or a pharmaceutically acceptable salt thereof.
In one embodiment, the mitoketoscin comprises the general formula: or a pharmaceutically acceptable salt f.
In another ment, the mitoketoscin comprises the general formula: or a pharmaceutically acceptable salt thereof.
In yet another embodiment, the mitoketoscin comprises the general formula: or a pharmaceutically acceptable salt thereof.
In one embodiment, the mitoketoscin possesses anti-aging activity.
In r embodiment, the mitoketoscin possesses ensitizing activity.
In a certain embodiment, the toscin possesses photosensitizing activity.
In one embodiment, the mitoketoscin sensitizes cancer cells to chemotherapeutic agents.
In an embodiment, the mitoketoscin sensitizes cancer cells to natural nces.
In another embodiment, the mitoketoscin sensitizes cancer cells to caloric restriction.
In yet another embodiment, the mitoketoscin binds to OXCT1.
In one ment, the mitoketoscin binds to ACAT1.
In an embodiment, the mitoketoscin binds to at least one of OXCT1/2 and ACAT1/2.
In another certain embodiment of the present invention, there is provided a method of ng cancer comprising administering to a patient in need thereof of a pharmaceutically effective amount of the mitoketoscin disclosed herein and a pharmaceutically acceptable carrier.
In one embodiment, the method of treating a microbial infection comprising administering to a patient in need thereof of a pharmaceutically effective amount of the mitoketoscin disclosed herein and a pharmaceutically acceptable carrier.
In another embodiment, the method of treating an lated condition sing administering to a patient in need thereof of a pharmaceutically effective amount of the mitoketoscin disclosed herein and a pharmaceutically able carrier.
In yet another embodiment, a pharmaceutical composition for treating cancer is ed, containing, as the active ingredient, at least one toscin.
In one embodiment, the pharmaceutical composition for treating a bacterial infection containing, as the active ient, at least one mitoketoscin.
In another embodiment, the pharmaceutical composition for treating a pathogenic yeast infection containing, as the active ingredient, at least one mitoketoscin.
In an embodiment, the pharmaceutical composition for treating aging, as the active ingredient, at least one mitoketoscin.
In one embodiment, the present invention provides a method of identifying a mitoketoscin using virtual high-throughput screening and phenotypic drug ing, the method sing; identifying a mitoketoscin that targets a mitochondrial ketone re-utilization enzyme using virtual high-throughput screening; synthesizing the toscin; and testing the mitoketoscin for mitochondrial inhibition activity.
One embodiment r comprising stering a mitochondrial support substrate.
In yet another embodiment, the mitochondrial support substrate comprises at least one of glucose, pyruvate, lactate, fatty acids and acetyl-carnitine.
BRIEF DESCRIPTION OF THE DRAWINGS FIG. I shows a schematic diagram outlining a drug discovery strategy according to embodiments of the present approach. illustrates the chemical structures of eight candidate mitoketoscin compounds 1-8 fied ing phenotypic drug screening.
FIGs. 3A-F shows the s of six candidate mitoketoscin compounds on mammosphere formation in MCF7 cells.
FIGs. 4A-D shows the effects of four candidate mitoketoscin compounds on ATP- ion in MCF7 cells.
FIGs. 5A-B and 6A-B each show the s of two candidate mitoketoscin compounds on basal respiration, proton leak, ATP-linked respiration, maximal respiration, and spare respiratory capacity in MCF7 cells. shows the s of five candidate mitoketoscin compounds on aerobic glycolysis in MCF7 cells. shows the effects of five candidate mitoketoscin compounds on extracellular acidification rate (ECAR) over time in MCF7 cells.
FIGs. 8A-D illustrate docking images of four candidate mitoketoscin nds. shows a schematic diagram outlining how OXCT1 and ACAT1 function to drive ATP production.
A shows four candidate mitoketoscin compounds and their respective IC- 50s for ting CSC propagation. B shows the chemical structure of arecoline, a naturally occurring ACAT1 inhibitor.
A compares the structures of two candidate mitoketoscin compounds. B shows the pharmacophores for two candidate mitoketoscin compounds.
A shows the structure of Coenzyme A (CoA). B compares the structure of CoA with two candidate mitoketoscin compounds. shows a schematic diagram outlining a follow-up treatment strategy with mitochondrial substrates to ameliorate potential side effects of mitoketoscins, according to embodiments of the t approach.
DESCRIPTION The ing description illustrates embodiments of the present approach in sufficient detail to enable practice of the present approach. gh the t approach is described with reference to these specific embodiments, it should be iated that the present approach may be embodied in different forms, and this description should not be construed as limiting any ed claims to the specific embodiments set forth herein. Rather, these embodiments are provided so that this sure will be thorough and complete, and will fully convey the scope of the t approach to those skilled in the art.
Mitochondrial metabolism is an untapped gateway for treating a number of afflictions, ranging from cancer to bacterial and fungal infections to aging. Functional mitochondria are required for the propagation of cancer stem cells. Inhibiting mitochondrial metabolism in cancer cells impedes the propagation of those cells. Mitochondrial inhibitors targeting the re-utilization of ketone bodies as mitochondrial fuels therefore represent a new class of anti-cancer therapeutics. These compounds may also inhibit mitochondrial protein translation, and ore may function as spectrum antibiotics that target both ia and pathogenic yeast. Research has also shown that mitochondrial inhibitors have anti-aging properties; hence mitoketoscins may also impart anti-aging benefits.
Novel inhibitors of mitochondrial ATP production that bind to at least one of OXCT1/2 and ACAT1/2 – mitoketoscins – may be identified through a convergent approach of virtual high-throughput screening followed by in vivo validation for mitochondrial inhibition. is an overview of methods for identifying mitoketoscins by using in silico drug screening and ypic drug screening disclosed herein. All or a portion of the three-dimensional structure of the porcine OXCT1 and human ACAT1 proteins may be used in step S101 to fy novel compounds that bind to these ns through virtual high-throughput screening (vHTS) (i.e., in silico drug screening). The screening may be performed across a library of molecules. For instance, during initial igations the inventors screened a collection of 30,000 small molecule compounds for compounds expected to bind anywhere to the yl-CoA: 3-ketoacid CoA transferase from pig heart covalently bound to CoA (PDB code 3OXO) or to the CoA binding site of human mitochondrial cetyl-CoA thiolase (PDB code 2F2S). Initial vHTS may use various screening programs, such as the eHiTS screening program, to identify a subset of compounds having a strong binding affinity to either protein. For example, the ors used eHiTS to identify the top 1,000 ranked compounds from an initial library, based on predicted binding affinity. eHiTS is a screening method that systematically covers the part of the conformational and positional search space that avoids severe steric clashes, producing highly accurate docking poses at a speed that is well-suited for virtual high-throughput screening.
It should be appreciated that those skilled in the art may select or develop methods for identifying a subset of compounds having a desired binding affinity. To efficiently m the docking, a series of clip files may be prepared corresponding to the entire protein structure and each compound docked sequentially at each of the clip files. Consensus scoring of the top nds may be carried out using AutoDock 4.2, based on the same l binding site for each compound ted from the eHiTS screen. Further analysis of predicted binding affinity and visual inspection may be carried out using a number of methods, including for example a de novo design program such as SPROUT. See Law et al., J Mol Struct. 666: 651-657 (2003), which is incorporated by reference in its entirety, for information about SPROUT. Depending on the l y size and results, a number of compounds may be selected for phenotypic drug screening. For example, the inventors selected 227 compounds that performed well in these analysis steps for ypic drug screening at step S103. 84 compounds were ed for the OXCT1-based phenotypic screen and 143 compounds were selected for the ACAT1-based phenotypic screen.
Phenotypic drug screening S103 may be accomplished by testing the mitochondrial inhibition of selected compounds in a selected cell line. For example, ATP depletion assays may be used. The inventors tested the selected 227 compounds on their ability to functionally induce ATP-depletion in MCF7 human breast cancer cells. Approximately 85% of cellular ATP is normally generated by OXPHOS in mitochondria, so ATP-depletion is a surrogate marker for mitochondrial inhibition. It should be appreciated that those skilled in the art may employ other surrogates for mitochondrial inhibition. However, for the ATP-depletion assay inventors employed, MCF7 cells (6,000 cells/well) were plated into black clear-bottom 96-well plates and incubated overnight before treatment. The 227 compounds identified by vHTS were applied to the plated MCF7 cells at a concentration of 20 µM and were screened for ATP ion. Compounds showing ATP-depletion effects were subsequently re-screened at a lower concentration (10 µM) to identify the top eight compounds that most potently induce ATP-depletion. Compounds were tested after 72 hours of incubation and experiments were performed in duplicate. After treatment, media was ted from the wells and plates were washed with warm phosphate-buffered saline (PBS) mented with Ca2+ and Mg2+. Then, cells were incubated with a t 33342 ) staining solution (10 µg/ml) for 30 min and washed with PBS to te cell viability.
Fluorescence was read with a plate reader using tion/emission wavelengths at 355/460-nm.
Then, a CellTiter-Glo luminescent assay (Promega) was performed to measure metabolic activity (ATP content) in the very same wells that were treated with a given compound. Assays were performed according to the manufacturer’s protocol. Fluorescence intensity (Hoechst staining) and luminescence intensity (ATP content) were normalized to vehicle-alone d controls and were displayed as percent control for ison. All eight test compounds significantly depleted ATP levels in viable cells. It should be appreciated that those of skill in the art may choose to employ the same or similar ATP-depletion assays, modify such assays, or may replace the ATP-depletion assay with another methodology for screening ed compounds for mitochondrial inhibition (e.g., oxygen ption assays).
The present approach includes methods of confirming cell viability. Persons of skill in the art may select one or more methods for ming cell viability suitable for the particular embodiment. The inventors initially used the Sulphorhodamine (SRB) assay, which is based on the measurement of cellular protein content. After treatment for 72 hours in 96-well plates, cells were fixed with 10% trichloroacetic acid (TCA) for 1 hour in the cold room, and were dried overnight at room temperature. Then, cells were incubated with SRB for 15 min, washed twice with 1% acetic acid, and air dried for at least 1 hour. y, the protein-bound dye was dissolved in a 10 mM Tris, pH 8.8 solution and read using the plate reader at 540-nm. Using the SRB assay, the inventors ed only the compounds depleting ATP levels without prominent cytotoxicity for further analysis. Prominent cytotoxicity was defined as fewer than 30% of cells still on the plate. Of , embodiments employing other cell viability confirmation methodology may select nds for further analysis based on other considerations as may be known in the art.
The present approach further es methods of functional validation at step S105, during which a compound’s function as a ondrial inhibitor may be confirmed. A number of methods may be used for functional validation, including, for example, lic flux analysis, mammosphere assays, ity assays, and antibiotic (anti-bacterial and/or anti-fungal) activity. For example, the inventors determined extracellular acidification rates (ECAR) and realtime oxygen ption rates (OCR) for MCF7 cells using the Seahorse Extracellular Flux (XF96) analyzer (Seahorse Bioscience, MA, USA). MCF7 cells were maintained in DMEM supplemented with 10% FBS (fetal bovine , 2 mM GlutaMAX, and 1% Pen- Strep. 5,000 cells per well were seeded into XF96-well cell culture plates, and incubated overnight at 37°C in a 5% CO2 humidified atmosphere. After 24 hours, cells were treated with selected compounds showing ATP-depletion without prominent cytotoxicity at various concentrations (or vehicle alone). After 72 hours of ent, cells were washed in pre-warmed XF assay media (for OCR measurement, XF assay media was supplemented with 10mM glucose, 1mM Pyruvate, 2mM L- glutamine and adjusted at pH 7.4). Cells were maintained in 175 μL/well of XF assay media at 37°C in a non-CO2 incubator for 1 hour. During incubation, 25 μL of 80mM e, 9μM oligomycin, 1M 2-deoxyglucose (for ECAR measurement) and 25 μL of 10μM oligomycin, 9μM FCCP, 10μM rotenone, 10μM antimycin A (for OCR measurement) in XF assay media was loaded into the ion ports of the XFe-96 sensor cartridge. During the experiment, the instrument ed these tors into the wells at a given time point, while ECAR/OCR was measured continuously. ECAR and OCR measurements were normalized by protein content (using the Sulphorhodamine B assay). Data sets were analyzed by XFe-96 software, using one-way ANOVA and Student’s t-test calculations. All experiments were performed in triplicate, and results validated the mitochondrial inhibition effects of the mitoketoscin compounds described herein. It should be appreciated that numerous methods are known for functional validation, and that s of skill in the art may select one or more depending on the tion needs (e.g., other assays that measure or approximate mitochondrial function).
In y, the present approach may include methods of identifying potential mitoketoscins using in silico drug screening and phenotypic drug screening. Novel compounds identified using this ology may be tested for anti-cancer activity (e.g., the ability to inhibit mammosphere ion and cell migration) and may be further tested on distinct bacterial and/or yeast strains to investigate anti-microbial activity. summarizes the general methods for drug screening and validation according to an embodiment of the t approach, but it should be appreciated that those of skill in the art may deviate from the specific examples disclosed herein without departing from the present approach.
The present approach has led to the identification of mitoketoscins that have anticancer properties, and embodiments of the present approach may take the form of one or more of these compounds, as well as pharmaceutical compositions including effective amounts of one or more of these nds, and various methods of treatment using one or more of these compounds. Based on the inventors’ initial screening and validation, the nds identified in have anti-cancer properties and are therefore mitoketoscins. In view of the inventors’ research, these mitoketoscins are ore candidates for clinical trial. It should be appreciated that the mitoketoscins identified in are not exhaustive, but are merely those compounds that have been identified thus far using the novel methodology set forth herein. It should be appreciated by those skilled in the art that the therapeutically-effective amount of each compound, for a particular y can be determined h the application of straightforward procedures as are known in the art.
Some embodiments may take the form of one or more mitoketoscins. The embodiment may be included in a pharmaceutical composition for treating cancer, bacterial infection, and/or pathogenic yeast infection. For e, a mitoketoscin may be a general pharmacophore having the ing structure (or a salt thereof): , , wherein Z is defined as ethylpiperidine or ethylpyrrolidine, , or .
As another example, a mitoketoscin may be a l pharmacophore having the ing structure (or a salt thereof): wherein each R may be the same or different and is selected from the group consisting of hydrogen, carbon, nitrogen, sulfur, oxygen, flourine, chlorine, bromine, iodine, carboxyl, alkanes, cyclic alkanes, alkane-based derivatives, alkenes, cyclic alkenes, -based derivatives, alkynes, alkyne-based derivative, ketones, ketone-based derivatives, aldehydes, aldehyde-based tives, carboxylic acids, carboxylic acid-based derivatives, ethers, ether- based derivatives, esters and ester-based derivatives, amines, amino-based derivatives, amides, amide-based derivatives, monocyclic or polycyclic arene, heteroarenes, arene-based derivatives, heteroarene-based derivatives, phenols, phenol-based derivatives, benzoic acid, and c sed derivatives.
As another example, a mitoketoscin may be a general pharmacophore having the following structure (or a salt thereof): As a further example, a toscin may be a general pharmacophore having the following structure (or a salt thereof): n each R may be the same or different and is selected from the group consisting of hydrogen, carbon, nitrogen, sulfur, oxygen, flourine, chlorine, bromine, iodine, yl, alkanes, cyclic s, alkane-based derivatives, alkenes, cyclic alkenes, alkene-based derivatives, alkynes, alkyne-based derivative, ketones, ketone-based derivatives, aldehydes, aldehyde-based derivatives, carboxylic acids, carboxylic acid-based derivatives, ethers, etherbased derivatives, esters and ester-based derivatives, amines, amino-based tives, amides, based derivatives, monocyclic or polycyclic arene, heteroarenes, arene-based derivatives, heteroarene-based derivatives, phenols, phenol-based derivatives, c acid, and benzoic acidbased derivatives.
As another example, a mitoketoscin may be a general pharmacophore having the following structure (or a salt f): wherein each R may be the same or different and is selected from the group consisting of hydrogen, carbon, en, sulfur, oxygen, flourine, chlorine, bromine, iodine, carboxyl, alkanes, cyclic alkanes, alkane-based derivatives, alkenes, cyclic alkenes, alkene-based derivatives, alkynes, alkyne-based tive, s, ketone-based tives, aldehydes, aldehyde-based derivatives, carboxylic acids, carboxylic acid-based derivatives, , etherbased derivatives, esters and ester-based derivatives, amines, amino-based derivatives, amides, based derivatives, monocyclic or polycyclic arene, heteroarenes, arene-based derivatives, heteroarene-based derivatives, phenols, phenol-based derivatives, benzoic acid, and benzoic acidbased derivatives.
Another example of a mitoketoscin is a general pharmacophore having the following structure (or a salt thereof): A further example of a mitoketoscin is a general pharmacophore having the following structure (or a salt thereof): An additional example of a mitoketoscin is a general pharmacophore having the following structure (or a salt thereof): wherein each R may be the same or different and is selected from the group consisting of hydrogen, carbon, nitrogen, sulfur, oxygen, flourine, chlorine, e, iodine, carboxyl, alkanes, cyclic alkanes, alkane-based tives, alkenes, cyclic alkenes, alkene-based derivatives, alkynes, alkyne-based derivative, ketones, ketone-based derivatives, aldehydes, aldehyde-based derivatives, carboxylic acids, carboxylic acid-based derivatives, ethers, ether-based derivatives, esters and ester-based tives, amines, aminobased derivatives, amides, based derivatives, monocyclic or polycyclic arene, heteroarenes, arene-based derivatives, heteroarene-based derivatives, phenols, phenol-based tives, benzoic acid, and benzoic acid-based tives.
Another example of a mitoketoscin is a general pharmacophore having the following structure (or a salt thereof): wherein each R may be the same or different and is ed from the group consisting of hydrogen, carbon, nitrogen, sulfur, oxygen, flourine, chlorine, bromine, iodine, carboxyl, alkanes, cyclic s, -based derivatives, alkenes, cyclic alkenes, alkene-based derivatives, alkynes, alkyne-based derivative, ketones, ketone-based derivatives, aldehydes, aldehyde-based derivatives, carboxylic acids, carboxylic ased derivatives, ethers, etherbased derivatives, esters and ester-based derivatives, amines, amino-based derivatives, amides, amide-based tives, monocyclic or polycyclic arene, arenes, arene-based derivatives, heteroarene-based derivatives, phenols, phenol-based tives, benzoic acid, and benzoic acidbased derivatives.
A further example of a mitoketoscin is a general pharmacophore having the following structure (or a salt f): Another example of a mitoketoscin is a general pharmacophore having the following structure (or a salt thereof): wherein each R may be the same or different and is selected from the group consisting of hydrogen, carbon, nitrogen, sulfur, oxygen, flourine, chlorine, bromine, iodine, carboxyl, alkanes, cyclic alkanes, alkane-based derivatives, alkenes, cyclic alkenes, alkene-based derivatives, alkynes, alkyne-based derivative, ketones, ketone-based tives, aldehydes, aldehyde-based derivatives, carboxylic acids, carboxylic acid-based derivatives, , etherbased tives, esters and ester-based derivatives, amines, amino-based derivatives, amides, amide-based derivatives, clic or polycyclic arene, heteroarenes, arene-based derivatives, heteroarene-based derivatives, phenols, phenol-based derivatives, benzoic acid, and c acidbased derivatives.It should be appreciated that the mitoketoscins may be selected for therapeutic use individually, or in combination with more than one specific toscin, and/or with other substances to enhance the efficacy of other therapeutics. The therapeutics may be used in the form of usual pharmaceutical compositions which may be prepared using one or more known methods.
For example, a pharmaceutical composition may be prepared by using diluents or excipients such as, for example, one or more fillers, g agents, binders, wetting agents, disintegrating agents, surface active agents, lubricants, and the like as are known in the art. Various types of administration unit forms may be selected depending on the therapeutic e(s). Examples of forms for pharmaceutical compositions include, but are not limited to, tablets, pills, powders, liquids, suspensions, emulsions, granules, capsules, suppositories, injection preparations (solutions and suspensions), topical creams, and other forms as may be known in the art. For the purpose of shaping a pharmaceutical composition in the form of tablets, any excipients which are known may be used, for example carriers such as lactose, white sugar, sodium de, glucose, urea, starch, calcium ate, kaolin, cyclodextrins, crystalline cellulose, silicic acid and the like; binders such as water, ethanol, propanol, simple syrup, glucose solutions, starch solutions, gelatin solutions, carboxymethyl cellulose, shelac, methyl cellulose, potassium phosphate, nylpyrrolidone, etc. Additionally, egrating agents such as dried starch, sodium alginate, agar powder, laminalia powder, sodium en carbonate, calcium ate, fatty acid esters of polyoxyethylene an, sodium laurylsulfate, yceride of stearic acid, starch, lactose, etc., may be used. Disintegration inhibitors such as white sugar, stearin, coconut butter, hydrogenated oils; absorption accelerators such as quaternary ammonium base, sodium laurylsulfate, etc., may be used. Wetting agents such as glycerin, starch, and others known in the art may be used. Adsorbing agents such as, for example, starch, lactose, kaolin, bentonite, colloidal silicic acid, etc., may be used. Lubricants such as purified talc, stearates, boric acid powder, polyethylene glycol, etc., may be used. If tablets are d, they may be further coated with the usual coating materials to make the tablets as sugar coated s, gelatin film coated tablets, s coated with enteric coatings, tablets coated with films, double layered tablets and multi- layered tablets. ceutical compositions adapted for l administration may be formulated as ointments, , suspensions, lotions, powders, solutions, pastes, gels, foams, sprays, ls, or oils. Such pharmaceutical compositions may include conventional additives which include, but are not limited to, preservatives, solvents to assist drug penetration, co-solvents, emollients, propellants, viscosity modifying agents (gelling ), surfactants and carriers.
The present approach may, in some embodiments, involve methods of testing compounds, and in particular mitoketoscins, for anti-cancer properties. As discussed above, vHTS and computational chemistry may be used to identify candidate mitochondrial inhibitors. Those candidates may be tested for specific anti-cancer properties. For example, the inventors compared seven candidate compounds in parallel for their ability to inhibit mammosphere ion in MCF7 cells. demonstrates that six compounds tested inhibited mammosphere formation.
Compound 2 and 8 (FIGs. 3A and 3B, tively) were the two most potent candidates in decreasing the number of mammospheres, a measure of cancer stem cell activity, at a concentration of 25 µM. Compound 6 and 3 were also ive (FIGs. 3C and 3D, respectively), while compound 5 and 1 (FIGs. 3E and 3F, respectively) were less potent inhibitors of mammosphere Compound ID IC-50 (µM) OXCT1 Hits 1 ALB-H01004577 160.4 2 ALB-H09465625 11.3 3 ALB-H15358970 46.7 4 ALB-H15354504 166.8 ACAT1 Hits ALB-H04367562 66.7 6 LEG19576081 22.9 8 ALB-H01005022 10.1 Table 1. Mitoketoscin inhibition of mammosphere formation in MCF7 cells Table 1 summarizes the mammosphere formation inhibition results for seven candidate compounds. Table 1 shows that seven compounds inhibited mammosphere formation with half-maximal inhibitory concentrations (IC-50s) between 10 and 170 µM. nds 1 to 4 were identified from the OXCT1 screen, and Compounds 5, 6, and 8 were identified from the ACAT1 screen.
The present ch may, in some embodiments, involve methods of on validation of toscin compounds. For example, the inventors assessed functional validation of four candidates using the Seahorse Analyzer, which quantitatively measures oxygen consumption rate (OCR) and extracellular acidification rate (ECAR). OCR is a surrogate marker for OXPHOS and ECAR is a surrogate marker for glycolysis and L-lactate production.
The inventors’ results demonstrated that Compounds 2, 3, 6, and 8 all dosedependently inhibited mitochondrial oxygen-consumption in MCF7 cells. FIGs. 4A and 4B show that Compounds 2 and 8 reduced mitochondrial respiration icantly, even at doses as low as µM. Compounds 6 and 3 were also potent tors (FIGs. 4C and 4D). As shown in FIGs. 5 and 6, the compounds dosed dependently reduced basal respiration, proton leak, ATP-linked respiration, and maximal respiration. shows how four compounds significantly inhibited glycolysis compared to the control.
Some embodiments of the present approach may include testing compounds for anti-cancer properties by ering compound effects on mammosphere formation. It should be appreciated that those skilled in the art may use other methods known in the art for ing a candidate mitochondrial tor’s effects on a particular cell line without departing from the present approach. It should also be appreciated that those skilled in the art may assess a candidate mitochondrial inhibitor’s effects on other cancer types, as the inhibitors target cancer stem cells (CSCs). CSCs show conserved or similar features across most cancer types.
FIGs. 8A-8D illustrate the molecular ng of Compounds 2, 8, 3, and 6. The goal of molecular modeling is to predict the predominant binding mode of a compound to a known three dimensional structure. and 8C show the molecular docking of Compounds 2 and 3, respectively, at the yl-CoA g site within the 3D crystal structure of OXCT1. A comparison of and 8C shows that at least one amino acid, ASN-373, is predicted to directly bind to both Compounds 2 and 3. FIGs. 8B and 8D shows the molecular docking of Compounds 8 and 6, respectively, at the CoA binding site within the 3D crystal structure of ACAT1. A comparison of and 8D shows that at least two amino acids, LEU-184 and HIS- 192, are predicted to directly bind to both Compounds 8 and 6. These predicted dominant binding modes may be invaluable to r lead optimization.
Ketone bodies functionally behave as mitochondrial fuels, which may actively drive tumor growth and metastasis. In this context, OXCT1 and ACAT1 are two mitochondrial proteins that participate in ketone re-utilization, as is summarized in The inventors molecularly targeted OXCT1 and ACAT1 to prevent cancer cells from recycling ketone bodies into Acetyl-CoA, which normally enters the TCA cycle, driving mitochondrial ATP production.
A top hit for the OXCT1 screen und 2) and a hit for the ACAT1 screen (Compound 8) have similar al structures, with the exception of minor functional side groups, as shown in A. The underlying “chemical scaffold” or pharmacophore is the same for both small les, as shown in FIG 11B. The structures of Compounds 2 and 8 were also compared to the molecular structure of Coenzyme A in to trate structural similarities.
Two compounds from the OXCT1 screen (Compounds 3 and 4) are structurally similar to each other, as is shown in A. However, based on their observed IC-50 values, Compound 3 is nearly 4 times more potent than Compound 4 in its ability to target CSC propagation. The unique chemical groups that distinguish these two les structurally (highlighted by arrows in A), may be responsible for the observed ences in their IC- 50s observed for their inhibition of CSC propagation.
Recent studies provide additional evidence of a role for ACAT1 as an oncogene, as these studies identified arecoline as a potential ACAT1 inhibitor. Garcia-Bermudez et al, Mol Cell 2016, 64(5):856-857. Arecoline is a nicotinic ased alkaloid found within the areca nut, which is the fruit of the areca palm tree (Areca catechu). Arecoline has shown anti-tumor activity, r validating that drugs targeting ACAT1 might be valuable as anti-cancer agents. However, the inventors did not assess its capacity to target CSCs. As arecoline is very small molecule (shown in B for ison to Compounds 6 and 3), it may need to be modified significantly by medicinal chemistry to increase its potency. ine is not a mitoketoscin, because the compound is known to be carcinogenic.
While normal ketone metabolism occurs under conditions of organismal starvation and/or severe nutrient deprivation, this regulation is lost in human tumors, and ketone metabolism appears to occur constitutively in cancer cells. Targeting ketone lism in human tumors, under normal dietary conditions, would be predicted to have minimal lic side effects.
Nevertheless, the ial side-effects of ketone inhibitors could be significantly ameliorated or “controlled” by including a “rescue” step, consisting of a follow-up treatment with other mitochondrial support ates, such as glucose, pyruvate, lactate, fatty acids and/or acetylcarnitine , as is shown in FIG 13. Sterile ose and L-lactate intravenous solutions (D5W, D5NS, Lactated Ringer’s) are already used routinely in hospitals for other clinical and eutic indications; hence, a follow-up treatment is clinically feasible.
The inventors have shown that compounds inducing acute ATP depletion in cancer cells may sensitize those cells to radiation, ultraviolet light, chemotherapeutic agents, natural substances, and/or caloric restriction. Mitoketoscins, as discussed herein, have demonstrated ATP- ion effects. Based on these preliminary s, mitoketoscins may also be used as radiosensitizers and/or photo-sensitizers. Use as radiosensitizers and/or photo-sensitizers may be in combination with other treatment vectors, including but not limited to other cancer treatment s as may be known in the art, and cancer treatment through inhibiting mitochondrial biogenesis as disclosed herein. Similarly, mitoketoscins may be used to functionally sensitize bulk cancer cells and cancer stem cells to chemotherapeutic agents, pharmaceuticals, and/or other natural substances, such as dietary ments and caloric restriction.
In addition to anti-cancer and anti-biotic behavior, the mitochondrial inhibitors that may be identified by the present ch have the potential to slow the mammalian aging process. c inhibition of ondrial protein translation has been shown to have beneficial sideeffects , and in particular the side effect of slowing the aging process and increasing lifespan in model organisms. Lower steady-state levels of Mrps5 (a mitoribosomal n) are strongly functionally correlated with longer murine lifespan, resulting in a significant lifespan increase of ~250 days. In addition, selective knock-down of Mrps5 in C. elegans dramatically increases lifespan. Mrps5 knock-down worms show significant decreases in ondrial respiration and ATP production. Similarly, down of the worm homologs of mitochondrial complex I, III, IV and V, as well as several TCA cycle enzymes, all robustly extended lifespan, further ating d OXPHOS activity and lower ATP levels as the mechanism. Finally, pharmacological inhibition of mitochondrial biogenesis (using the off-target effects of doxycycline) also significantly increases an in C. elegans. Thus, mitoketoscins may be used to therapeutically target the aging process and to extend an.
Mitoketoscins may also be used to ze and/or reverse drug resistance in cancer cells. Drug resistance is thought to be based, at least in part, on sed mitochondrial function in cancer cells. In ular, cancer cells demonstrating resistance to endocrine therapies, such as tamoxifen, are expected to have increased mitochondrial function. Mitoketoscins inhibit mitochondrial function, and therefore may be useful in reducing and, in some cases reversing, drug resistance in cancer cells.
The terminology used in the description of the invention herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the ion. As used in the description of the invention and the appended claims, the singular forms “a,” “an” and “the” are intended to include the plural forms as well, unless the t clearly indicates otherwise. The invention includes numerous alternatives, modifications, and equivalents as will become apparent from consideration of the following ed description.
It will be understood that gh the terms “first,” d,” “third,” “a),” “b),” and “c),” etc. may be used herein to describe s elements of the invention should not be limited by these terms. These terms are only used to distinguish one element of the invention from another. Thus, a first element discussed below could be termed a element aspect, and similarly, a third without departing from the teachings of the present invention. Thus, the terms “first,” “second,” “third,” “a),” “b),” and “c),” etc. are not intended to necessarily convey a sequence or other hierarchy to the associated elements but are used for identification purposes only. The sequence of operations (or steps) is not limited to the order presented in the claims.
Unless otherwise d, all terms (including technical and scientific terms) used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. It will be r understood that terms, such as those defined in commonly used dictionaries, should be interpreted as having a meaning that is consistent with their meaning in the t of the present application and relevant art and should not be reted in an idealized or overly formal sense unless expressly so defined . The terminology used in the ption of the invention herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. All publications, patent applications, patents and other references mentioned herein are incorporated by reference in their entirety. In case of a conflict in terminology, the present specification is controlling.
] Also as used herein, “and/or” refers to and encompasses any and all le combinations of one or more of the associated listed items, as well as the lack of combinations when interpreted in the alternative (“or”).
Unless the context indicates otherwise, it is specifically intended that the various features of the invention bed herein can be used in any combination. Moreover, the present invention also contemplates that in some embodiments of the invention, any feature or combination of features set forth herein can be excluded or omitted. To illustrate, if the specification states that a complex comprises components A, B and C, it is specifically intended that any of A, B or C, or a ation thereof, can be omitted and disclaimed.
As used herein, the transitional phrase “consisting essentially of” (and grammatical variants) is to be interpreted as encompassing the recited als or steps “and those that do not materially affect the basic and novel characteristic(s)” of the claimed invention. Thus, the term “consisting essentially of” as used herein should not be interpreted as equivalent to “comprising.” The term “about,” as used herein when referring to a measurable value, such as, for example, an amount or concentration and the like, is meant to ass variations of ± 20%, ± 10%, ± 5%, ± 1%, ± 0.5%, or even ± 0.1% of the specified amount. A range provided herein for a measureable value may include any other range and/or individual value therein.
Having thus described certain embodiments of the present ion, it is to be understood that the invention defined by the appended claims is not to be limited by particular details set forth in the above description as many apparent ions thereof are possible without departing from the spirit or scope thereof as hereinafter claimed.

Claims (40)

1. A method of identifying a mitoketoscin using virtual high-throughput screening and phenotypic drug screening, the method comprising; identifying a compound that targets a mitochondrial ketone re-utilization enzyme using virtual high-throughput ing; and testing the compound for mitochondrial inhibition activity.
2. The method of claim 1, n the mitoketoscin binds to OXCT1.
3. The method of claim 1, wherein the mitoketoscin binds ACAT1.
4. The method of any one of claims 1 to 3, wherein the phenotypic drug screening is in vitro drug screening.
5. The method of any one of claims 1 to 3, wherein the ypic drug ing comprises an ATP-depletion assay.
6. The method of any one of claims 1 to 3, wherein the phenotypic drug screening comprises an extracellular acidification rate assay.
7. The method of any one of claims 1 to 3, n the phenotypic drug screening comprises an oxygen consumption rate assay.
8. The method of any one of claims 1 to 7, further comprising testing the mitoketoscin for anti-cancer activity.
9. The method of claim 8, wherein the anti-cancer activity tested is mammosphere
10. The method of claim 8, wherein the anti-cancer activity tested is cell migration.
11. The method of any one of claims 1 to 10 further comprising g the mitoketoscin for anti-microbial activity.
12. A mitoketoscin comprising the general formula: , wherein R may be selected from the group consisting of chlorine, bromine, iodine, yl, alkanes, cyclic alkanes, alkenes, cyclic alkenes, s, ketones, aldehydes, ylic acids, , esters, amines, amides, monocyclic or clic arene, heteroarenes, phenols and benzoic acid.
13. The mitoketoscin of claim 12, wherein the mitoketoscin possesses anti-aging activity.
14. The mitoketoscin of claim 12, wherein the mitoketoscin possesses at least one of radiosensitizing activity and photosensitizing activity.
15. The mitoketoscin of claim 12, wherein the mitoketoscin possesses anti-microbial activity.
16. The mitoketoscin of claim 12, wherein the mitoketoscin sensitizes cancer stem cells to chemotherapeutic agents.
17. The mitoketoscin of claim 12, wherein the toscin sensitizes cancer stem cells to natural substances.
18. The mitoketoscin of claim 12, wherein the mitoketoscin sensitizes cancer stem cells to caloric restriction.
19. The mitoketoscin of any one of claims 12 to 18, wherein the mitoketoscin binds to at least one of OXCT1, OXCT2, ACAT1, and ACAT2.
20. Use of a mitoketoscin in the manufacture of a medicament for treating cancer, wherein the mitoketoscin comprises the general formula: , wherein R may be ed from the group consisting of hydrogen, fluorine, chlorine, bromine, iodine, carboxyl, alkanes, cyclic alkanes, alkenes, cyclic s, alkynes, ketones, aldehydes, carboxylic acids, ethers, esters, amines, amides, monocyclic or polycyclic arene, heteroarenes, phenols and benzoic acid.
21. The use of claim 20, wherein R is fluorine.
22. Use of a mitoketoscin in the manufacture of a medicament for treating a microbial infection, wherein the mitoketoscin comprises the general a: , wherein R may be selected from the group ting of hydrogen, fluorine, chlorine, e, iodine, carboxyl, alkanes, cyclic alkanes, alkenes, cyclic s, alkynes, ketones, aldehydes, carboxylic acids, ethers, esters, amines, amides, clic or polycyclic arene, heteroarenes, phenols and benzoic acid.
23. The use of claim 22, wherein R is ne.
24. Use of a mitoketoscin in the manufacture of a ment for treating an age-related condition, wherein the mitoketoscin comprises the general formula: , wherein R may be selected from the group consisting of hydrogen, fluorine, chlorine, bromine, iodine, carboxyl, alkanes, cyclic alkanes, alkenes, cyclic alkenes, alkynes, ketones, aldehydes, carboxylic acids, ethers, , amines, amides, monocyclic or polycyclic arene, heteroarenes, s and benzoic acid.
25. The use of claim 24, wherein R is fluorine.
26. A pharmaceutical composition comprising, as an active ingredient, a toscin comprising the general formula: , wherein R may be selected from the group consisting of hydrogen, fluorine, chlorine, bromine, iodine, yl, alkanes, cyclic alkanes, alkenes, cyclic alkenes, alkynes, ketones, aldehydes, carboxylic acids, ethers, esters, amines, amides, monocyclic or polycyclic arene, heteroarenes, s, c acid; and a pharmaceutically able carrier.
27. The pharmaceutical composition of claim 26, wherein R is fluorine.
28. A mitoketoscin comprising the general formula: , wherein R may be selected from the group consisting of hydrogen, , chlorine, bromine, iodine, carboxyl, alkanes, cyclic alkanes, alkenes, cyclic alkenes, alkynes, ketones, aldehydes, carboxylic acids, ethers, esters, amines, amides, monocyclic or polycyclic arene, heteroarenes, phenols and benzoic acid.
29. The mitoketoscin of claim 28, wherein the mitoketoscin possesses anti-aging activity.
30. The mitoketoscin of claim 28, wherein the mitoketoscin possesses radiosensitizing activity.
31. The mitoketoscin of claim 28, wherein the mitoketoscin possesses photosensitizing activity.
32. The mitoketoscin of claim 28, wherein the mitoketoscin sensitizes cancer stem cells to chemotherapeutic agents.
33. The mitoketoscin of claim 28, wherein the mitoketoscin sensitizes cancer stem cells to natural substances.
34. The mitoketoscin of claim 28, wherein the toscin sensitizes cancer stem cells to c restriction.
35. The mitoketoscin of any one of claims 28 to 34, wherein the mitoketoscin binds to at least one of OXCT1, OXCT2, ACAT1, and ACAT2.
36. Use of a mitoketoscin in the manufacture of a ment for treating cancer, wherein the mitoketoscin comprises the general formula: , n R may be selected from the group consisting of hydrogen, fluorine, chlorine, bromine, iodine, carboxyl, alkanes, cyclic alkanes, alkenes, cyclic alkenes, s, ketones, aldehydes, ylic acids, ethers, esters, amines, , monocyclic or polycyclic arene, heteroarenes, phenols and benzoic acid.
37. The use of claim 36, wherein R is fluorine.
38. The use of claim 36 or claim 37, n the medicament further comprises a mitochondrial support substrate comprising at least one of glucose, pyruvate, lactate, fatty acids and acetyl-carnitine.
39. A pharmaceutical composition comprising, as an active ingredient, a mitoketoscin comprising the general formula: , n R may be selected from the group consisting of hydrogen, fluorine, chlorine, bromine, iodine, carboxyl, s, cyclic s, alkenes, cyclic alkenes, alkynes, ketones, des, carboxylic acids, ethers, esters, amines, amides, monocyclic or polycyclic arene, heteroarenes, phenols, benzoic acid; and a pharmaceutically acceptable carrier.
40. The pharmaceutical composition of claim 39, wherein R is fluorine. Drug Screening & Validation Virtual high-throughput screening (vHTS) Step 1. Selection of two molecular libraries of 1000 compounds each (both for OXCT1 and ACAT1) from a small molecular screening collection of = : 30,000 compounds Step 2. Further analysis of predicted binding affinity and visual inspection Compounds performing well in all is steps were selected for assay Phenotypic drug screening pletion assays on human breast cancer cells (MCF7) 84 Compounds (OXCTl); 143 nds (ACATl) Observed 8 hits at 20 uM Functional validation Mammosphere assays Metabolic Flux is OCR/ECAR Mitoketoscins Mitoketoscins Compound 1 Compound 5 F? CI H N <0) 2 N ZLOVQ/FHo N Compound 2 Compound 6 ng 9 H \v" \ N N F30 H OE Compound 3 Compound 7 Q N / o N 0 m \ (1flag NJLS / N N\__jN N 3D-Spheroid CSC Assay spheres) ctrl) versus counts 80 Mammosphere 60 Control 10 [1M 25 [1M 50 [1M ctrl) versus 00O counts 0'1O Mammosphere J:o Control 10 [1M 25 [1M 50 [1M WO 05698 control 100 counts 80 Mammosphere ** Control 25 [1M 50 [1M 100 uM ctrl) versus 80 counts 60 Mammosphere 40 Control 25 [1M 50 [1M 100 uM control 100 counts 00O Mammosphere 0'1O Control 25 [1M 50 [1M 100 uM ctrl) versus 100 counts 00O Mammosphere 0'1O Control 25 [1M 50 [1M 100 uM WO 05698 WControl azflsésee Compound 2 20 40 60 8O Time (min) -o— Control AEQEEBEE «\w 2.5 pM ““5 HM Com D.ou nd 8 \\§. 20 40 60 80 Time (min) *Control ~\\\\\\\\\\‘25 ulVl xxxwm uM n: ““5 uM -— 2O o 20 40 60 80 Time (min) 30 +Contro| \\10 [1M \\\\\\\\\\'5 “M m \\\\\\\\\“2.5 “M -— 20 O 20 4O 6O 80 Time (min) 30 Cm0.wm 2 Im.mm 25 N 1pM axfigéaee Nmu.M 50 1.0 MN\\\\ m 5 ______________________________________________________ mmum m N\\\\\\\\\mm m §§m.w kmmk.m VWN\\\\\\\\\\\\\\\\\\\\\\\\\\\\\Nmmmam mm capacity 30 Compound 8 I Control Ammm\:_E\_oE& 2 5 2.5 pM 20 NSpM Basal Proton Leak ATP-link Maximal Spare respiration ation respiration respiratory capacfly FIG. SB C0m D.0U nd 5 I Control 25 ®5pM axfigésee 10 pM 25 pM Basal Proton Leak ATP-link Maximal Spare respiration respiration respiration respiratory capacity 30 Compound 3 I COntrd 25 § 2.5 pM Ammm\:_E\_oE& 20 * §@ 51 u.0Mu.M 0 \\\\\\\\\m \m Spare respiration respiration atory capacfly FIG. GB
NZ792168A 2017-06-26 2018-06-25 Mitoketoscins: mitochondrial-based therapeutics targeting ketone metabolism in cancer cells NZ792168A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US62/524,829 2017-06-26

Publications (1)

Publication Number Publication Date
NZ792168A true NZ792168A (en) 2022-09-30

Family

ID=

Similar Documents

Publication Publication Date Title
AU2020257159B2 (en) Mitoketoscins: mitochondrial-based therapeutics targeting ketone metabolism in cancer cells
AU2022205218B2 (en) Mitoriboscins: mitochondrial-based therapeutics targeting cancer cells, bacteria, and pathogenic yeast
BRPI0512891A (en) compound and a pharmaceutically acceptable salt or prodrug or solvate thereof, composition, methods of inhibiting checkpoint kinase 1 in a cell, of sensitizing cells in an individual undergoing chemotherapeutic or radiotherapeutic treatment for a medical condition and of inhibiting aberrant cell proliferation, use of a compound, and article of manufacture for pharmaceutical use in human
MX2013005661A (en) Pharmaceutical combination of paclitaxel and a cdk inhibitor.
Fan et al. Novel 4-aminoquinazoline derivatives induce growth inhibition, cell cycle arrest and apoptosis via PI3Kα inhibition
US11497749B2 (en) Mitoflavoscins: targeting flavin-containing enzymes eliminates cancer stem cells (CSCS) by inhibiting mitochondrial respiration
Patel et al. Catalyst-free synthesis of imidazo [5, 1-b] quinazolines and their antimicrobial activity
Guo et al. Design, synthesis and molecular docking of salicylic acid derivatives containing metronidazole as a new class of antimicrobial agents
Guo et al. Structure-guided optimization and mechanistic study of a class of quinazolinone-threonine hybrids as antibacterial ThrRS inhibitors
NZ792168A (en) Mitoketoscins: mitochondrial-based therapeutics targeting ketone metabolism in cancer cells
US20220339125A1 (en) Mitoriboscins: mitochondrial-based therapeutics targeting cancer cells, bacteria, and pathogenic yeast
Zhang et al. Regulatory roles of mitochondrial ribosome in lung diseases and single cell biology
WO2024118830A1 (en) Atropisomers of mat2a inhibitors
CN117085019A (en) PARP1 inhibitor and application thereof in preparation of anticancer drugs