Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ANT2 INHIBITOR COMPOUNDS AND METHODS OF USE THEREOF
Document Type and Number:
WIPO Patent Application WO/2009/126335
Kind Code:
A3
Abstract:
The invention relates to methods of treating cell proliferation related disorders or diseases, such as cancer. The invention further relates to pharmaceutical compositions for treating cell proliferation related disorders or diseases.

Inventors:
TERADA NAOHIRO (US)
OSTROV DAVID A (US)
Application Number:
PCT/US2009/002295
Publication Date:
January 14, 2010
Filing Date:
April 08, 2009
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV FLORIDA (US)
TERADA NAOHIRO (US)
OSTROV DAVID A (US)
International Classes:
A61K31/122; A61K31/662; A61P35/00
Domestic Patent References:
WO2002088078A22002-11-07
Other References:
CHEVROLLIER ARNAUD ET AL.: "ANT2 Isoform Required for Cancer Cell Glycolysis", JOURNAL OF BIOENERGETICS AND BIOMEMBRANES, vol. 37, no. 5, 2005, pages 307 - 316
BRAS MORGANE LE ET AL.: "Chemosensitization by Knockdown of Adenine Nucleotide Translocase-2", CANCER RES, vol. 66, no. 18, 2006, pages 9143 - 9152
WELCH KAREN T. ET AL.: "Discovery of non-carbohydrate inhibitors of aminoglycoside modifying enzymes", BIOORGANIC AND MEDICINAL CHEMISTRY, vol. 13, 2005, pages 6252 - 6263
Attorney, Agent or Firm:
HSI, Jeffrey, D. et al. (P.O. Box 55874Boston, MA, US)
Download PDF:
Claims:

What is claimed is:

1. A method of treating a subject suffering from or susceptible to a cell proliferation related disorder or disease, the method comprising administering to a subject in need thereof a therapeutically effective amount of an adenine nucleotide translocase 2 (ANT2) inhibitor, to thereby treat the subject suffering from or susceptible to a cell proliferation related disorder or disease.

2. The method of claim 1, wherein the ANT2 inhibitor is a selective ANT2 inhibitor.

3. The method of claim 1 or 2, wherein the ANT2 inhibitor is selected from a compound of Table 1,

or a pharmaceutically acceptable salt or prodrug thereof.

4. A method for identifying a compound that inhibits ANT2 activity, the method comprising: a) obtaining a crystal structure of ANT2 or obtaining information relating to the crystal structure of ANT2, and b) modeling a test compound into or on the crystal structure coordinates to determine whether the compound binds to ANT2.

5. The method of claim 4, wherein the step of modeling comprises modeling or determining the ability of the compound to bind to or associate with a binding pocket defined by structure coordinates of one or more ANT2 amino acid residues selected from LyslO5 and ArglOό

6. A method for identifying a compound that modulates the activity of ANT2, the method comprising using the atomic coordinates of one or more of ANT2 amino acid residues LyslO5 and ArglOό to generate a three-dimensional structure of a molecule comprising an ANT2 binding pocket, and employing the three-dimensional structure to identify a compound that modulates the activity of ANT2.

7. A kit for treating a cell proliferation related disorder or disease in a subject, comprising an ANT2 activator compound, or a pharmaceutically acceptable ester, salt, and prodrug thereof, and instructions for use.

8. A machine readable storage medium which comprises the structural coordinates of a binding pocket defined (at least in part) by structure coordinates of one or more of ANT2 amino acid residues LyslO5 and ArglOό, or a homologous binding pocket.

9. A computer for producing a three-dimensional representation of a molecule or molecular complex, wherein said molecule or molecular complex comprises a binding pocket defined by structural coordinates of a binding pocket defined at least in part by structure coordinates of one or more of ANT2 amino acid residues LyslO5 and ArglOό, or a homologous binding pocket; or b) a three-dimensional representation of a homologue of said molecule or molecular complex, wherein said homologue comprises a binding pocket that has a root mean square deviation from the backbone atoms of said amino acids of not more than about 2.0 angstroms; the computer comprising (i) a machine-readable data storage medium comprising a data storage material encoded with machine-readable data, wherein said data comprises the structural coordinates of a binding pocket defined at least in part by structure coordinates of one or more of ANT2 amino acid residues LyslO5 and ArglOό, or a homologous binding pocket; (ii) a working memory for storing instructions for processing said machine- readable data;

(iii) a central-processing unit coupled to said working memory and to said machine-readable data storage medium for processing said machine readable data into said three-dimensional representation; and

(iv) a display coupled to said central-processing unit for displaying said three- dimensional representation.

10. A computer for producing a three-dimensional representation of a molecule or molecular complex, wherein said molecule or molecular complex comprises a binding pocket defined by structural coordinates of a binding pocket defined at least in part by structure coordinates of one or more of ANT2 amino acid residues LyslO5 and

ArglOό, or a homologous binding pocket; or b) a three-dimensional representation of a homologue of said molecule or molecular complex, wherein said homologue comprises a binding pocket that has a root mean square deviation from the backbone atoms of said amino acids of not more than about 2.0 angstroms; the computer comprising:

(i) a machine-readable data storage medium comprising a data storage material encoded with machine-readable data, wherein said data comprises the structural coordinates of a binding pocket defined (at least in part) by structure coordinates of one or more of ANT2 amino acid residues LyslO5 and ArglOό, or a homologous binding pocket;

(ii) a working memory for storing instructions for processing said machine- readable data;

(iii) a central-processing unit coupled to said working memory and to said machine-readable data storage medium for processing said machine readable data into said three-dimensional representation; and

(iv) a display coupled to said central-processing unit for displaying said three- dimensional representation.

11. A pharmaceutical composition comprising a compound of Table 1 , or a pharmaceutically acceptable salt or prodrug thereof, together with a pharmaceutically acceptable carrier.

12. A structure of ANT2 having the structural coordinates of a sequence identified in FIG. 4.

13. A method for identifying a chemotherapeutic agent for the treatment of cancer, the method comprising: a) obtaining a structural representation of ANT2 or obtaining information relating to the structure of ANT2, and b) modeling a test compound into or on the structure coordinates of the structural representation of ANT2 to determine whether the compound inhibits ANT2; thereby identifying a chemotherapeutic agent for the treatment of cancer.

14. A method of treating a subject suffering from or susceptible to a cell proliferation related disorder or disease, the method comprising inhibiting the activity of adenine nucleotide translocase 2 (ANT2) in the subject such that the subject is treated.

15. The method of claim 14, wherein the cell proliferation related disorder or disease is cancer.

16. A method of inhibiting the growth of a cell, the method comprising inhibiting the activity of adenine nucleotide translocase 2 (ANT2) in the cell such that growth of the cell is inhibited.

17. The method of claim 16, wherein the cell is a cancer cell.

18. A method of killing a cell, the method comprising inhibiting the activity of adenine nucleotide translocase 2 (ANT2) in the cell such that the cell is killed.

19. The method of claim 18, wherein the cell is a cancer cell.

20. The method of claim 16 or claim 18, wherein the activity of adenine nucleotide translocase 2 (ANT2) in the cell is inhibited by contacting the cell with a selective inhibitor of ANT2.

21. A method of identifying a compound useful for inhibiting ANT2 activity comprising the steps of:

1. Obtaining the crystal structure of human ANT2, whereby a three- dimensional structure of ANT2 will be generated;

2. Determination of ANT2 binding regions or pockets by mapping the atomic coordinates of selected amino acid residues, Lys 105 and Arg 106, to provide an ANT2 modeling system, then providing a machine readable storage medium which comprises the three-dimensional ANT2 structure and binding pocket(s) defined (at least in part) by structure coordinates of one or more of ANT2 amino acid residues LyslO5 and ArglO6;

3. Employing the ANT2 modeling system to identify test compounds that bind within the selected ANT2 binding pockets;

4. Determining the selectivity of compound inhibition, i.e. inhibiting ANT2 over ANTl, by measuring the ADP/ ATP exchange for each ANT, following treatment with compound;

5. Identifying compounds that inhibit ANT2 to the extent of cell growth inhibition or cell death.

22. The method of claim 17, wherein the cancer is breast or ovarian.

23. The method of claim 4, comprising using the atomic coordinates of an amino acid sequence comprising ANT2 amino acid residues Lys 105, Arg 106, and Thr 107.

24. The method of claim 2, wherein the ANT2 inhibitor compound is demonstrated to bind to ANT2 at an amino acid sequence comprising ANT2 amino acid residues LyslO5, ArglOό, and Thr 107.

25. The method of claim 1, wherein the subject is identified as in need of treatment by administration of an ANT2 inhibitor compound.

26. The method of claim 1, wherein the subject is identified as in need of treatment by administration of a ANT2 inhibitor compound that is demonstrated to bind to ANT2 at an amino acid sequence comprising ANT2 amino acid residues LyslO5, ArglOό, and Thr 107.

Description:

ANT2 INHIBITOR COMPOUNDS AND METHODS OF USE THEREOF

CROSS REFERENCE TO RELATED APPLICATIONS

This application claims priority benefit of U.S. Provisional Patent Application No. 61/123,524, filed April 8, 2008, the contents of which are hereby incorporated by reference in their entirety.

BACKGROUND OF THE INVENTION

Adenine nucleotide translocase (ANT) mediates the exchange of ADP and ATP on the inner mitochondrial membrane, thus playing an essential role in energy metabolism of eukaryotic cells. Under aerobic conditions, ATP produced within mitochondria through oxidative phosphorylation is exported to the cytosol through ANT to support cellular activities. In exchange, ADP is imported to provide a substrate for the conversion of ADP to ATP by ATP synthase. In contrast, under anaerobic conditions, glycolytic ATP is imported into mitochondria through ANT. This retro-translocated ATP is necessary for energy-dependent processes within the mitochondria and is critical for maintaining the membrane potential of mitochondria and preventing apoptosis mediated by mitochondrial membrane permeabilization.

All of the known eukaryotic species, including yeasts, have multiple ANTs. For example, Saccharomyces cerevisiae has three ANTs (AACl, 2, and 3), among which AACl and AAC3 are expressed almost exclusively in aerobic and anaerobic conditions, respectively. Thus, the different ANTs are likely utilized for ATP export and import in order to efficiently cope with varying external nutrient and oxygen conditions. Humans possess four distinct ANT isoforms, encoded by four genes, the transcription of which depends on tissue type, developmental stage, cell proliferation, and hormone status. Of these, ANTl (SLC25A4) is expressed primarily in the heart and skeletal muscle, ANT2 (SLC25A5) is expressed in rapidly growing cells and is inducible, and ANT3 (SLC25A6) appears to be constitutively expressed in all tissues (Stepien 1992, Lunardi 1992). We have recently determined that ANT4 is expressed

exclusively in male germ cells, with this expression being particularly high during meiosis (Rodic 2005 and Brower 2007).

Among the four ANTs in humans and other mammals, ANT2 is proposed to play the primary role in importing glycolytic ATP into mitochondria (Chevrollier 2005). Of particular interest, many cancer cells express very high levels of ANT2. This appears to be a consequence of adaptive regulation to the enhanced glycolytic metabolism in cancerous tissue. It is known, for example, that glucose uptake and glycolysis are constitutively upregulated in most cancer cells, and the import of glycolytic ATP into mitochondria is essential for maintenance of mitochondrial membrane potential and intramitochondrial enzymatic activities in hypoxic conditions.

Inhibitors of ADP/ ATP exchange by ANTs have previously been identified, including bongkrekic acid and carboxyatractyloside, but these drugs inhibit all ANTs in a non-specific manner. A particular concern for the use of these prototype drugs in clinics is therefore the inhibition of ANTl, which is known to cause mitochondrial myopathy and hypertrophic cardiomyopathy.

BRIEF SUMMARY OF THE INVENTION

The invention provides compounds and compositions thereof useful in methods of treating a subject suffering from or susceptible to a cell proliferation related disorder or disease, the method comprising administering to a subject in need thereof a therapeutically effective amount of a compound delineated herein (e.g., an adenine nucleotide translocase 2 (ANT2) inhibitor), to thereby treat the subject suffering from or susceptible to a cell proliferation related disorder or disease. The invention also relates to methods for identifying such compounds and compositions. The invention also provides compounds and compositions thereof useful in methods of modulating cells and cell proliferation.

Overexpression of ANT2 , particularly in human breast cancer cell lines (e.g., MCF7, MDA-MB-231, SK-BR-3) and ovarian cancer cell lines (e.g., SK-OV-3 and SNU8) suggests that ANT2 plays a unique role in cancer development, particularly in those forms. Jang, et al., Breast Cancer Res. (2008) 10:Rl 1. Additionally, while ANT2 is highly expressed in MCF7 and MDA-MB-231, it appears that ANTl and ANT3 expressions are not detectable. Furthermore, non-neoplastic MCFlOA cell line is reported to barely express ANT2 as well as ANT 1, but highly transcribes ANT3.

These transcription patterns of ANT iso forms in breast cancer are also identical to that of breast cancer. As such, it is proposed that expression levels of ANT2 correlate with cancer progression, and ANT2 in particular plays a greater role in cancer or tumor cells than in normal cells; and therefore the ANT2 inhibitor compounds herein are more selective for cancer cells than normal cells, thereby providing better targeted therapeutic agents for ANT2 mediated diseases/disorders, which also then provide an improved side effect profile (e.g, lesser adverse side effects) than other existing chemotherapeutic agents.

In yet another aspect, the invention provides the use of a compound of any of the formulae herein alone or together with one or more of the above-described second therapeutic agents in the manufacture of a medicament, either as a single composition or as separate dosage forms, for treatment or prevention in a subject of a disease, disorder or symptom set forth above. Another aspect of the invention is a compound of the formulae herein for use in the treatment or prevention in a subject of a disease, disorder or symptom thereof delineated herein.

BRIEF DESCRIPTION OF THE DRAWINGS

The present invention is further described below with reference to the following non-limiting examples and with reference to the following figures, in which:

FIG 1 is a chart showing the inhibition of HeLa cell growth by certain compounds. HeLa cells were plated in 24 well dishes and treated 24 hrs later, with no drug, DMSO vehicle control, 50μM bongkrekic acid (BKA), or 50μM of one of the twenty four candidate ANT2 small compound inhibitors. After 5 days of culture, plates were washed gently with room temperature PBS, incubated with 0.2% crystal violet solution (Fisher Scientific) for 10 min at room temperature. Cells were then washed twice with water. 1% SDS solution was added to each well and gently agitated on orbital shaker until color was uniform, with no areas of dense coloration in bottom of wells. Absorbance was measured at 570nm. Compounds #1, 5, and 10 all showed marked decrease in cell survival with 79, 81, and 84 inhibition in cell growth respectively.

FIG 2 is a chart showing the inhibition of HeLa cell growth at varying concentration of selected compounds. The compounds that showed the greatest cell growth inhibition were selected for dose response determination. HeLa cells were plated on a 24 well plate and treated 24 hrs later with no, 6.25, 12.5, 25, or 50μM of drug #1, 5, 10 and 12. Crystal violet assay was performed as described above. IC 50 was between 12.5 and 25μM for compound #1, less than 6.25μM for compound #5, and 25~50 μM for compound #10. Compound #12 actually showed very little inhibition at up to 50μM in this series.

FIG 3 shows the predicted tertiary structure of ANT2 with an inhibitor bound to a predicted binding site. There are currently no solved structures of ANT2 in the Protein Data Bank. However, we have experience generating homology models and utilizing them as the basis for molecular docking. We previously identified a novel inhibitor of murine JAK2 in this manner (Sandberg 2005). We utilized SWISS- MODEL, an Automated Comparative Protein Modeling Server, to generate an atomic model of human ANT2 based on the most similar structure in the protein data bank, bovine Antl/Adtl (lokc), which has 89% amino acid identity to human ANT2. The high degree of sequence similarity between them renders the atomic model more accurate, than if it were based on structures with a lower degree of similarity. The resultant computer predicted human ANT2 structure and the molecular docking site, which we selected, that is unique to ANT2 are shown. The site was selected basically by unique amino acid sequence only existing in ANT2 but not in other ANTs (Fig. 4), and characteristics favorable for small molecule binding. Importantly, the selected docking site is located at the ADP/ ATP entry site. Thus, we predict that the small compound interaction at the site would interfere with ADP/ ATP exchange. See, Sandberg EM, Ma X, He K, Frank SJ, Ostrov DA, Sayeski PP (2005) Identification o 1,2,3,4,5,6-hexabromocyclohexane as a small molecule inhibitor of jak2 tyrosine kinase autophosphorylation. J Med Chem 48, 2526-33

FIG 4 shows the sequences of ANTl, ANT2, ANT3 and ANT4. The small molecule docking site we chose include the amino acid sequences unique to ANT2 (Lysine 1 5 , Arginine 1 6 ) as indicated by a red box. Black underlines indicate transmembrane domains that are highly conserved among all ANTs.

FIG 5 illustrates that cancer cells are more susceptible to compounds than non- transformed fibroblasts. The compounds that showed the greatest cell growth

inhibition were selected for dose response determination. NIH 3T3 or HeLa cells were cultured on 24 well plate and treated 24 hrs later with 0, 6.25, 12.5, 25, or 50μM of drug #1, 5, 10 and DMSO vehicle control . Crystal violet staining was performed by incubating cells in 0.2% crystal violet solution (Fisher Scientific) for 10 min at room temperature. Cells were then washed twice with water.

FIG 6 illustrates that cancer cells are more susceptible to compounds than non- transformed fibroblasts. Crystal violet stain from Figure 2 (see above) was solubilized by addition of 1% SDS solution to each well followed by gentle agitation on an orbital shaker until color was uniform, with no areas of dense coloration in bottom of wells. Absorbance was measured at 570 run. Percent survival of NIH 3T3 or HeLa cells was calculated relative to untreated cells. Compound #1 showed a drastic decrease in cell growth at 50 μM with 5% HeLa cell survival compared to 40% cell survival in 3T3 cells. Treatment with compound #5 resulted in 40% and 18% cell survival in HeLa cells compared to 80% and 50% 3T3 cell survival at 12.5 and 25 μM respectively.

DETAILED DESCRIPTION OF THE INVENTION 1. DEFINITIONS

Before further description of the present invention, and in order that the invention may be more readily understood, certain terms are first defined and collected here for convenience.

The term "administration" or "administering" includes routes of introducing the compound of the invention(s) to a subject to perform their intended function. Examples of routes of administration that may be used include injection (subcutaneous, intravenous, parenterally, intraperitoneally, intrathecal), oral, inhalation, rectal and transdermal. The pharmaceutical preparations may be given by forms suitable for each administration route. For example, these preparations are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, etc. administration by injection, infusion or inhalation; topical by lotion or ointment; and rectal by suppositories. Oral administration is preferred. The injection can be bolus or can be continuous infusion. Depending on the route of administration, the compound of the invention can be coated with or disposed in a selected material to protect it from natural conditions which may detrimentally affect

its ability to perform its intended function. The compound of the invention can be administered alone, or in conjunction with either another agent as described above or with a pharmaceutically-acceptable carrier, or both. The compound of the invention can be administered prior to the administration of the other agent, simultaneously with the agent, or after the administration of the agent. Furthermore, the compound of the invention can also be administered in a proform which is converted into its active metabolite, or more active metabolite in vivo.

The term "alkyl" refers to the radical of saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups. The term alkyl further includes alkyl groups, which can further include oxygen, nitrogen, sulfur or phosphorous atoms replacing one or more carbons of the hydrocarbon backbone, e.g., oxygen, nitrogen, sulfur or phosphorous atoms, hi preferred embodiments, a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone (e.g., Ci-C 30 for straight chain, C 3 -C 30 for branched chain), preferably 26 or fewer, and more preferably 20 or fewer, and still more preferably 4 or fewer. Likewise, preferred cycloalkyls have from 3-10 carbon atoms in their ring structure, and more preferably have 3, 4, 5, 6 or 7 carbons in the ring structure. Moreover, the term alkyl as used throughout the specification and sentences is intended to include both "unsubstituted alkyls" and "substituted alkyls," the latter of which refers to alkyl moieties having substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone. Such substituents can include, for example, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety. It will be understood by those skilled in the art that the moieties substituted on the hydrocarbon chain can themselves be substituted, if appropriate. Cycloalkyls can be further substituted, e.g., with the

substituents described above. An "alkylaryl" moiety is an alkyl substituted with an aryl (e.g., phenylmethyl (benzyl)). The term "alkyl" also includes unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively. Unless the number of carbons is otherwise specified, "lower alkyl" as used herein means an alkyl group, as defined above, but having from one to ten carbons, more preferably from one to six, and still more preferably from one to four carbon atoms in its backbone structure, which may be straight or branched-chain. Examples of lower alkyl groups include methyl, ethyl, n-propyl, i-propyl, tert-butyl, hexyl, heptyl, octyl and so forth. In preferred embodiment, the term "lower alkyl" includes a straight chain alkyl having 4 or fewer carbon atoms in its backbone, e.g., Ci-C 4 alkyl.

The terms "alkoxyalkyl," "polyaminoalkyl" and "thioalkoxyalkyl" refer to alkyl groups, as described above, which further include oxygen, nitrogen or sulfur atoms replacing one or more carbons of the hydrocarbon backbone, e.g., oxygen, nitrogen or sulfur atoms.

The terms "alkenyl" and "alkynyl" refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond, respectively. For example, the invention contemplates cyano and propargyl groups. The term "aryl" as used herein, refers to the radical of aryl groups, including 5- and 6-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, benzene, pyrrole, furan, thiophene, imidazole, benzoxazole, benzothiazole, triazole, tetrazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like. Aryl groups also include polycyclic fused aromatic groups such as naphthyl, quinolyl, indolyl, and the like. Those aryl groups having heteroatoms in the ring structure may also be referred to as "aryl heterocycles," "heteroaryls" or "heteroaromatics." The aromatic ring can be substituted at one or more ring positions with such substituents as described above, as for example, halogen, hydroxyl, alkoxy, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarbonyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino

(including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, sulfonate, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety. Aryl groups can also be fused or bridged with alicyclic or heterocyclic rings which are not aromatic so as to form a polycycle (e.g., tetralin).

The language "biological activities" of a compound of the invention includes all activities elicited by compound of the inventions in a responsive cell or subject. It includes genomic and non-genomic activities elicited by these compounds. "Biological composition" or "biological sample" refers to a composition containing or derived from cells or biopolymers. Cell-containing compositions include, for example, mammalian blood, red cell concentrates, platelet concentrates, leukocyte concentrates, blood cell proteins, blood plasma, platelet-rich plasma, a plasma concentrate, a precipitate from any fractionation of the plasma, a supernatant from any fractionation of the plasma, blood plasma protein fractions, purified or partially purified blood proteins or other components, serum, semen, mammalian colostrum, milk, saliva, placental extracts, a cryoprecipitate, a cryosupematant, a cell lysate, mammalian cell culture or culture medium, products of fermentation, ascites fluid, proteins induced in blood cells, and products produced in cell culture by normal or transformed cells (e.g., via recombinant DNA or monoclonal antibody technology). Biological compositions can be cell-free. In a preferred embodiment, a suitable biological composition or biological sample is a red blood cell suspension. In some embodiments, the blood cell suspension includes mammalian blood cells. Preferably, the blood cells are obtained from a human, a non-human primate, a dog, a cat, a horse, a cow, a goat, a sheep or a pig. In preferred embodiments, the blood cell suspension includes red blood cells and/or platelets and/or leukocytes and/or bone marrow cells. In some embodiments, the sample includes a marker (e.g., genetic material, mutant form, chemical or biological tag or identifier) suitable for following disorder or disease progression. The term "chiral" refers to molecules which have the property of non- superimposability of the mirror image partner, while the term "achiral" refers to molecules which are superimposable on their mirror image partner.

The term "diastereomers" refers to stereoisomers with two or more centers of dissymmetry and whose molecules are not mirror images of one another.

The term "effective amount" includes an amount effective, at dosages and for periods of time necessary, to achieve the desired result, e.g., sufficient to treat a cell proliferation related disorder or disease or an associated condition. An effective amount of compound of the invention may vary according to factors such as the disease state, age, and weight of the subject, and the ability of the compound of the invention to elicit a desired response in the subject. Dosage regimens may be adjusted to provide the optimum therapeutic response. An effective amount is also one in which any toxic or detrimental effects (e.g., side effects) of the compound of the invention are outweighed by the therapeutically beneficial effects.

A therapeutically effective amount of compound of the invention (i.e., an effective dosage) may range from about 0.001 to 30 mg/kg body weight, or from about 0.01 to 10 mg/kg body weight, or from about 0.05 to 5 mg/kg body weight, or from about 0.1 to 1 mg/kg, 0.2 to 0.9 mg/kg, 0.3 to 0.8 mg/kg, 0.4 to 0.7 mg/kg, or 0.5 to 0.6 mg/kg body weight. The skilled artisan will appreciate that certain factors may influence the dosage required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present. Moreover, treatment of a subject with a therapeutically effective amount of a compound of the invention can include a single treatment or, preferably, can include a series of treatments, hi one example, a subject is treated with a compound of the invention in the range of between about 0.1 to 20 mg/kg body weight, one time per week for between about 1 to 10 weeks, preferably between 2 to 8 weeks, more preferably between about 3 to 7 weeks, and even more preferably for about 4, 5, or 6 weeks. It will also be appreciated that the effective dosage of a compound of the invention used for treatment may increase or decrease over the course of a particular treatment.

The term "enantiomers" refers to two stereoisomers of a compound which are non-superimposable mirror images of one another. An equimolar mixture of two enantiomers is called a "racemic mixture" or a "racemate."

The term "haloalkyl" is intended to include alkyl groups as defined above that are mono-, di- or polysubstituted by halogen, e.g., fluoromethyl and trifluoromethyl.

The term "halogen" designates -F, -Cl, -Br or -I. The term "hydroxyl" means -OH.

The term "heteroatom" as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, sulfur and phosphorus.

The term "homeostasis" is art-recognized to mean maintenance of static, or constant, conditions in an internal environment.

The language "improved biological properties" refers to any activity inherent in a compound of the invention that enhances its effectiveness in vivo. In a preferred embodiment, this term refers to any qualitative or quantitative improved therapeutic property of a compound of the invention, such as reduced toxicity.

The term "optionally substituted" is intended to encompass groups that are unsubstituted or are substituted by other than hydrogen at one or more available positions, typically 1, 2, 3, 4 or 5 positions, by one or more suitable groups (which may be the same or different). Such optional substituents include, for example, hydroxy, halogen, cyano, nitro, Ci-Csalkyl, C 3 -C 8 cycloalkyl, C 2 -C 8 alkenyl, C 2 - Cgalkynyl, Ci-C 8 alkoxy, C 2 -C 8 alkyl ether, C 3 -C 8 alkanone, Ci-C 8 alkylthio, amino, mono- or di-( Ci-C 8 alkyl)amino, haloCj-Qalkyl, Ci-Cgalkoxy, Ci-C 8 alkanoyl, C 2 - Qalkanoyloxy, Ci-Cgalkoxycarbonyl, -COOH, -CONH2, mono- or di-( Ci- C 8 alkyl)aminocarbonyl, -SO 2 NH 2 , and/or mono or di(C]-C 8 alkyl)sulfonamido, as well as carbocyclic and heterocyclic groups. Optional substitution is also indicated by the phrase "substituted with from O to X substituents," where X is the maximum number of possible substituents. Certain optionally substituted groups are substituted with from O to 2, 3 or 4 independently selected substituents (i.e., are unsubstituted or substituted with up to the recited maximum number of substitutents).

The term "isomers" or "stereoisomers" refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.

The term "obtaining" as in "obtaining the ACE activator" is intended to include purchasing, synthesizing or otherwise acquiring the ACE activator.

The phrases "parenteral administration" and "administered parenterally" as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticulare, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.

The terms "polycyclyl" or "polycyclic radical" refer to the radical of two or more cyclic rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls) in which two or more carbons are common to two adjoining rings, e.g., the rings are "fused rings". Rings that are joined through non-adjacent atoms are termed "bridged" rings. Each of the rings of the polycycle can be substituted with such substituents as described above, as for example, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkyl, alkylaryl, or an aromatic or heteroaromatic moiety.

The term "prodrug" includes compounds with moieties that can be metabolized in vivo. Generally, the prodrugs are metabolized in vivo by esterases or by other mechanisms to active drugs. Examples of prodrugs and their uses are well known in the art (See, e.g., Berge et al. (1977) "Pharmaceutical Salts", J. Pharm. Sci. 66: 1-19). The prodrugs can be prepared in situ during the final isolation and purification of the compounds, or by separately reacting the purified compound in its free acid form or hydroxyl with a suitable esterifying agent. Hydroxyl groups can be converted into esters via treatment with a carboxylic acid. Examples of prodrug moieties include substituted and unsubstituted, branch or unbranched lower alkyl ester moieties, {e.g., propionoic acid esters), lower alkenyl esters, di-lower alkyl-amino lower-alkyl esters (e.g., dimethylaminoethyl ester), acylamino lower alkyl esters (e.g., acetyloxymethyl ester), acyloxy lower alkyl esters (e.g., pivaloyloxymethyl ester), aryl esters (phenyl ester), aryl-lower alkyl esters (e.g., benzyl ester), substituted (e.g., with

methyl, halo, or methoxy substituents) aryl and aryl-lower alkyl esters, amides, lower- alkyl amides, di-lower alkyl amides, and hydroxy amides. Preferred prodrug moieties are propionoic acid esters and acyl esters. Prodrugs which are converted to active forms through other mechanisms in vivo are also included. The language "cell proliferation related disorder or disease" refers to diseases or conditions related to uncontrolled or disregulated cell growth or proliferation. Exemplary cell proliferation related disorders or diseases include, but are not limited to cancer, atherosclerosis, amyloidosis, polycythemia vera, and restenosis. Cancers include cancer of the colon, breast, bone, brain and others (e.g., osteosarcoma, neuroblastoma, colon adenocarcinoma), cardiac cancer (e.g., sarcoma, myxoma, rhabdomyoma, fibroma, lipoma and teratoma); lung cancer (e.g., bronchogenic carcinoma, alveolar carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma); various gastrointestinal cancer (e.g., cancers of esophagus, stomach, pancreas, small bowel, and large bowel); genitourinary tract cancer (e.g., kidney, bladder and urethra, prostate, testis; liver cancer (e.g., hepatoma, cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma); bone cancer (e.g., osteogenic sarcoma, fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma, multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma, benign chondroma, chondroblastoma, chondromyxofϊbroma, osteoid osteoma and giant cell tumors); cancers of the nervous system (e.g., of the skull, meninges, brain, and spinal cord); gynecological cancers (e.g., uterus, cervix, ovaries, vulva, vagina); hematologic cancer (e.g., cancers relating to blood, Hodgkin's disease, non-Hodgkin's lymphoma); skin cancer (e.g., malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis); and cancers of the adrenal glands (e.g., neuroblastoma). In one aspect, the cancer is breast or ovarian.

As used herein, "obtaining a biological sample from a subject," includes obtaining a sample for use in the methods described herein. A biological sample is described above.

The language "a prophylactically effective amount" of a compound refers to an amount of a compound of the invention of the formula (I) or otherwise described

herein which is effective, upon single or multiple dose administration to the patient, in preventing or treating a cell proliferation related disorder or disease.

The language "reduced toxicity" is intended to include a reduction in any undesired side effect elicited by a compound of the invention when administered in vivo.

The language "selective ANT2 inhibitor" refers to a compounds capable of inhibiting the activity of ANT2 while not substantially inhibiting the activity of at least one of ANTl, ANT3, and ANT4 at the same compound concentration. For example, a selective ANT2 inhibitor may have ANT2 inhibitory activity 2, 5, 10, 20, 50, 100, 500, or 1000-fold more potent than its ANTl, ANT3, or ANT4 inhibitory activity. In certain embodiments, the ANT2 inhibitory activity of a selective ANT2 inhibitor is at least 5-fold, 10-fold, or 20-fold more potent than the ANTl inhibitory activity of the compound.

The term "sulfhydryl" or "thiol" means -SH. The term "subject" includes organisms which are capable of suffering from a cell proliferation related disorder or disease or who could otherwise benefit from the administration of a compound of the invention of the invention, such as human and non-human animals. Preferred human animals include human patients suffering from or prone to suffering from a cell proliferative disease or disorder, as described herein. The term "non-human animals" of the invention includes all vertebrates, e.g., mammals, e.g., rodents, e.g., mice, and non-mammals, such as non-human primates, e.g., sheep, dog, cow, chickens, amphibians, reptiles, etc. "Susceptible to a cell proliferative disease or disorder" is meant to include subjects at risk of developing a cell proliferative disease or disorder, including cancer. The phrases "systemic administration," "administered systemically",

"peripheral administration" and "administered peripherally" as used herein mean the administration of a compound of the invention(s), drug or other material, such that it enters the patient's system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration. The language "therapeutically effective amount" of a compound of the invention of the invention refers to an amount of an agent which is effective, upon single or multiple dose administration to the patient, in treating or preventing a cell

proliferative disease or disorder or an associated condition or symptom, or in prolonging the survivability of the patient with such condition beyond that expected in the absence of such treatment.

With respect to the nomenclature of a chiral center, terms "d" and "1" configuration are as defined by the IUPAC Recommendations. As to the use of the terms, diastereomer, racemate, epimer and enantiomer will be used in their normal context to describe the stereochemistry of preparations.

2. COMPOUNDS OF THE INVENTION In one aspect, the invention provides a compound capable of inhibiting ANT2 activity. In certain embodiments, the compound is capable of inhibiting ANT2 activity selectively, e.g., without concomitant inhibition of other ANTs, including ANTl, ANT3, or ANT4.

In certain embodiments, the ANT2 inhibitor compound can be represented by compounds of Table 1 , or a pharmaceutically acceptable salt or prodrug thereof.

In general, a compound of the invention will be selected such that the compound is capable of binding to a binding site or pocket of ANT2 that is defined (at least in part) by structure coordinates of one or more of ANT2 amino acid residues LyslO5 and ArglOό. Moreover, in certain embodiments, a compound has one or more of the following properties: (1) not more than 5 hydrogen bond donors; (2) not more than 10 hydrogen bond acceptors; (3) a molecular weight of 1000 or less, 800 or less, 600 or less, 500 or less; and (4) a partition coefficient log P of less than 5.

Compounds according to the invention can generally be made according to techniques known in the field (see, e.g., Comprehensive Organic Synthesis, Trost, B. M. and Fleming, I. eds., Pergamon Press, Oxford; and references cited therein).

Furthermore, compounds of the invention can be purified, separated, or isolated, e.g., by crystallization, chromatographic separation (e.g., by liquid chromatography), or by other methods known in the art.

Naturally occurring or synthetic isomers can be separated in several ways known in the art. Methods for separating a racemic mixture of two enantiomers include chromatography using a chiral stationary phase (see, e.g., "Chiral Liquid Chromatography," WJ. Lough, Ed. Chapman and Hall, New York (1989)).

Enantiomers can also be separated by classical resolution techniques. For example, formation of diastereomeric salts and fractional crystallization can be used to separate enantiomers. For the separation of enantiomers of carboxylic acids, the diastereomeric salts can be formed by addition of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, and the like. Alternatively, diastereomeric esters can be formed with enantiomerically pure chiral alcohols such as menthol, followed by separation of the diastereomeric esters and hydrolysis to yield the free, enantiomerically enriched carboxylic acid. For separation of the optical isomers of amino compounds, addition of chiral carboxylic or sulfonic acids, such as camphorsulfonic acid, tartaric acid, mandelic acid, or lactic acid can result in formation of the diastereomeric salts.

3. USES OF THE COMPOUNDS OF THE INVENTION

As described herein below, it has now been found that the compounds of the invention and analogs can treat and prevent cell proliferative diseases and disorders, including cancer.

Thus, in one aspect, the invention provides a method of treating a subject suffering from or susceptible to a cell proliferation related disorder or disease, the method comprising administering to a subject in need thereof a therapeutically effective amount of a compound delineated herein (e.g., an adenine nucleotide translocase 2 (ANT2) inhibitor), to thereby treat the subject suffering from or susceptible to a cell proliferation related disorder or disease. The ANT2 inhibitor acts to impair growth and/or induce death of abnormal cells. In one aspect, the compound is a selective ANT2 inhibitor compound (e.g., as demonstrated by protocols know in the art, including those delineated hereon). In one embodiment, the compound is capable of binding to or interacting with a binding site or pocket defined (at least in part) by structure coordinates of one or more ANT2 amino acid residues selected from LyslO5 and ArglOβ. In certain embodiments, the compound is a compound disclosed herein, e.g., a compound of Table 1. In certain embodiments, the subject is a mammal, e.g., a primate, e.g., a human.

In another aspect, the invention provides a method of treating a subject suffering from or susceptible to a cell proliferation related disorder or disease, the

method comprising inhibiting the activity of adenine nucleotide translocase 2 (ANT2) in the subject such that the subject is treated.

In one aspect, the invention provides a method of inhibiting the growth of a cell, the method comprising inhibiting the activity of adenine nucleotide translocase 2 (ANT2) in the cell such that growth of the cell is inhibited. In one embodiment, the compound is capable of binding to or interacting with a binding site or pocket defined (at least in part) by structure coordinates of one or more ANT2 amino acid residues LyslO5 and ArglOό. In another embodiment, the ANT2 inhibitor compound is demonstrated to bind to ANT2 at an amino acid sequence comprising ANT2 amino acid residues LyslO5, ArglOό, and Thr 107. In certain embodiments, the compound is a compound disclosed herein, e.g., a compound of Table 1.

In one aspect, the invention provides a method of killing a cell, the method comprising inhibiting the activity of adenine nucleotide translocase 2 (ANT2) in the cell such that the cell is killed. In one aspect the cell is a cancer cell. In another aspect, the invention provides a method for inhibiting activity or expression of ANT2 (e.g., in vitro, in vivo) in a cell or a subject, the method comprising contacting the cell or subject with an effective amount of a compound capable of inhibiting activity of ANT2, such that activity of ANT2 is inhibited. In another aspect, the method is any of those herein wherein the the activity of adenine nucleotide translocase 2 (ANT2) in the cell is inhibited by contacting the cell with a selective inhibitor of ANT2.

The present methods can be performed on cells in culture, e.g. in vitro or ex vivo, or on cells present in an animal subject, e.g., in vivo. Compounds of the inventions can be initially tested in vitro using primary cultures of cells. The present methods can be performed on cells in culture, e.g. in vitro or ex vivo, or on cells present in an animal subject, e.g., in vivo. Compounds of the invention can be initially tested in vitro using cells from the respiratory tract from embryonic rodent pups (See e.g. U.S. Patent No. 5,179,109-fetal rat tissue culture), or other mammalian (See e.g. U.S. Patent No. 5,089,517-fetal mouse tissue culture) or non-mammalian animal models.

Alternatively, the effects of a compound of the invention can be characterized in vivo using animals models.

In one aspect, the methods herein modulate (e.g, inhibit, kill, induce apopotosis, reduce proliferation) cells that are cancer cells.

In certain embodiments, the methods of the invention include administering to a subject a therapeutically effective amount of a compound of the invention in combination with another pharmaceutically active compound. In a further embodiment, the additional therapeutic agent is an anti-cancer agent, chemotherapeutic agent, an anti-angiogenesis agent, cytotoxic agent, or an anti- proliferation agent. Examples of such chemotherapeutic agents include but are not limited to daunorubicin, daunomycin, dactinomycin, doxorubicin, epirubicin, idarubicin, esorubicin, bleomycin, mafosfamide, ifosfamide, cytosine arabinoside, bis- chloroethylnitrosurea, busulfan, mitomycin C, actinomycin D, mithramycin, prednisone, hydroxyprogesterone, testosterone, tamoxifen, dacarbazine, procarbazine, hexamethylmelamine, pentamethylmelamine, mitoxantrone, amsacrine, chlorambucil, methylcyclohexylnitrosurea, nitrogen mustards, melphalan, cyclophosphamide, 6- mercaptopurine, 6-thioguanine, cytarabine (CA), 5-azacytidine, hydroxyurea, deoxycoformycin, 4-hydroxyperoxycyclophosphoramide, 5-fluorouracil (5-FU), 5- fluorodeoxyuridine (5-FUdR), methotrexate (MTX), colchicine, vincristine, vinblastine, etoposide, trimetrexate, teniposide, cisplatin and diethylstilbestrol (DES). See, generally, The Merck Manual of Diagnosis and Therapy, 15th Ed., pp. 1206- 1228, Berkow et al., eds., Rahway, N. J., 1987). The compound of the invention and the pharmaceutically active compound may be administered to the subject in the same pharmaceutical composition or in different pharmaceutical compositions (at the same time or at different times).

Determination of a therapeutically effective amount or a prophylactically effective amount of the compound of the invention, can be readily made by the physician or veterinarian (the "attending clinician"), as one skilled in the art, by the use of known techniques and by observing results obtained under analogous circumstances. The dosages may be varied depending upon the requirements of the patient in the judgment of the attending clinician; the severity of the condition being treated and the particular compound being employed. In determining the therapeutically effective amount or dose, and the prophylactically effective amount or dose, a number of factors are considered by the attending clinician, including, but not limited to: the specific cell proliferative disease or disorder involved;

pharmacodynamic characteristics of the particular agent and its mode and route of administration; the desired time course of treatment; the species of mammal; its size, age, and general health; the degree of or involvement or the severity of the disease; the response of the individual patient; the particular compound administered; the mode of administration; the bioavailability characteristics of the preparation administered; the dose regimen selected; the kind of concurrent treatment (i.e., the interaction of the compound of the invention with other co-administered therapeutics); and other relevant circumstances.

Treatment can be initiated with smaller dosages, which are less than the optimum dose of the compound. Thereafter, the dosage may be increased by small increments until the optimum effect under the circumstances is reached. For convenience, the total daily dosage may be divided and administered in portions during the day if desired. A therapeutically effective amount and a prophylactically effective amount of a compound of the invention of the invention is expected to vary from about 0.1 milligram per kilogram of body weight per day (mg/kg/day) to about lOO mg/kg/day.

Compounds determined to be effective for the prevention or treatment of cell proliferative diseases or disorders in animals, e.g., dogs, chickens, and rodents, may also be useful in treatment of similar conditions in humans. Those skilled in the art of treatment in humans will know, based upon the data obtained in animal studies, the dosage and route of administration of the compound to humans. In general, the dosage and route of administration in humans is expected to be similar to that in animals.

In one aspect, the methods herein are those wherein the subject is identified as in need of treatment by administration of a compound herein (e.g., an ANT2 inhibitor compound). In one aspect, the methods herein are those wherein the subject is identified as in need of treatment by admiistration of a compound herein (e.g., an ANT2 inhibitor compound) that is demonstrated to bind to ANT2 at an amino acid sequence comprising ANT2 amino acid residues LyslO5, ArglOό, and Thr 107. The identification of those patients who are in need of prophylactic treatment for cell proliferative diseases or disorders is well within the ability and knowledge of one skilled in the art. Certain of the methods for identification of patients which are at risk of developing cell proliferative diseases or disorders which can be treated by the

subject methods are appreciated in the medical arts, such as family history, the presence of other risk factors associated with the development of that disease state in the subject patient, and the like. A clinician skilled in the art can readily identify such candidate patients, by the use of, for example, clinical tests, physical examination and medical/family/travel history.

A method of assessing the efficacy of a disease treatment in a subject includes determining the physical condition of the subject (e.g., blood pressure, degree or extent of disorder progression, obtaining a biological sample, and the like) and then administering a therapeutically effective amount of an ANT2 inhibitor compound of the invention to the subject. After a appropriate period of time after the administration of the compound, e.g., 2 hours, 4 hours, 8 hours, 12 hours, or 72 hours, or one week, the physical condition of the subject is determined again. The modulation of the cell proliferative disease or disorder indicates efficacy of an treatment. The physical condition of the subject may be determined periodically throughout treatment. For example, the physical condition of the subject may be checked every few hours, days or weeks to assess the further efficacy of the treatment. The method described may be used to screen or select patients that may benefit from treatment with an ANT2 inhibitor.

In another aspect, the invention provides a method for identifying a compound that modulates (e.g., inhibits) the activity of ANT2, the method comprising using the atomic coordinates of one or more of ANT2 amino acid residues LyslO5 and ArglOό to generate a three-dimensional structure of a molecule comprising an ANT2 binding pocket, and employing the three-dimensional structure to identify a compound that modulates the activity of ANT2. In another aspect, a compound of the invention is packaged in a therapeutically effective amount with a pharmaceutically acceptable carrier or diluent. The composition may be formulated for treating a subject suffering from or susceptible to a cell proliferation related disorder or disease, and packaged with instructions to treat a subject suffering from or susceptible to such a disease or condition. In aspects, the method of packaging comprises combining the compound or compositions delineated herein in a pharmaceutically acceptable packaging material, In other aspects the method of packaging comprises inserting, printing or attaching instructions (e.g., insert, printing on package, label) in or on the packaging.

In another aspect, the invention provides a packaged composition including a therapeutically effective amount of an ANT2 inhibitor compound and a pharmaceutically acceptable carrier or diluent. The composition may be formulated for treating a subject suffering from or susceptible to a cell proliferation related disorder or disease, and packaged with instructions to treat a subject suffering from or susceptible to a cell proliferation related disorder or disease.

In one aspect, the invention provides a kit for treating a cell proliferation related disorder or disease in a subject is provided and includes a compound disclosed herein, e.g., a compound of Table 1, or a pharmaceutically acceptable ester, salt, and prodrug thereof, and instructions for use. In further aspects, the invention provides kits for treating a cell proliferation related disorder or disease, assessing the efficacy of an anti-cell-proliferative treatment in a subject using an ANT2 inhibitor, monitoring the progress of a subject being treated with an ANT2 inhibitor, selecting a subject with or susceptible to a cell proliferation related disorder or disease, and/or treating a subject suffering from or susceptible to a cell proliferation related disorder or disease. In certain embodiments, the invention provides: a kit for treating a cell proliferation related disorder or disease in a subject, the kit comprising a compound capable of inhibiting activity (or expression) of ANT2, or pharmaceutically acceptable esters, salts, and prodrugs thereof, and instructions for use; in certain embodiments, the compound is represented any of the structures of Table 1, or a pharmaceutically acceptable salt thereof.

4. PHARMACEUTICAL COMPOSITIONS

The invention also provides a pharmaceutical composition, comprising an effective amount of a compound of the invention, e.g., a compound of Table 1, or otherwise described herein and a pharmaceutically acceptable carrier. In a further embodiment, the effective amount is effective to treat a cell proliferation related disorder or disease, as described previously.

In an embodiment, the compound of the invention is administered to the subject using a pharmaceutically-acceptable formulation, e.g., a pharmaceutically- acceptable formulation that provides sustained delivery of the compound of the invention to a subject for at least 12 hours, 24 hours, 36 hours, 48 hours, one week,

two weeks, three weeks, or four weeks after the pharmaceutically-acceptable formulation is administered to the subject.

In certain embodiments, these pharmaceutical compositions are suitable for topical or oral administration to a subject. In other embodiments, as described in detail below, the pharmaceutical compositions of the present invention may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or nonaqueous solutions or suspensions), tablets, boluses, powders, granules, pastes; (2) parenteral administration, for example, by subcutaneous, intramuscular or intravenous injection as, for example, a sterile solution or suspension; (3) topical application, for example, as a cream, ointment or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam; or (5) aerosol, for example, as an aqueous aerosol, liposomal preparation or solid particles containing the compound.

The phrase "pharmaceutically acceptable" refers to those compounds of the present invention, compositions containing such compounds, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. The phrase "pharmaceutically-acceptable carrier" includes pharmaceutically- acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject chemical from one organ, or portion of the body, to another organ, or portion of the body. Each carrier is "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate;

(I3) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations. Wetting agents, emulsifϊers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.

Examples of pharmaceutically-acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfϊte, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.

Compositions containing a compound of the invention(s) include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal, aerosol and/or parenteral administration. The compositions may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred per cent, this amount will range from about 1 per cent to about ninety-nine percent of active ingredient, preferably from about 5 per cent to about 70 per cent, more preferably from about 10 per cent to about 30 per cent.

Methods of preparing these compositions include the step of bringing into association a compound of the invention(s) with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a compound of the invention with liquid

carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.

Compositions of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the invention(s) as an active ingredient. A compound may also be administered as a bolus, electuary or paste.

In solid dosage forms of the invention for oral administration (capsules, tablets, pills, dragees, powders, granules and the like), the active ingredient is mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, acetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.

A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be

made by molding in a suitable machine a mixture of the powdered active ingredient moistened with an inert liquid diluent.

The tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.

Liquid dosage forms for oral administration of the compound of the invention(s) include pharmaceutically-acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifϊers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.

In addition to inert diluents, the oral compositions can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.

Suspensions, in addition to the active compound of the invention(s) may contain suspending agents as, for example, ethoxylated isostearyl alcohols,

polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.

Pharmaceutical compositions of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compound of the invention(s) with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent. Compositions of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.

Dosage forms for the topical or transdermal administration of a compound of the invention(s) include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound of the invention(s) may be mixed under sterile conditions with a pharmaceutically-acceptable carrier, and with any preservatives, buffers, or propellants which may be required.

The ointments, pastes, creams and gels may contain, in addition to compound of the invention(s) of the present invention, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.

Powders and sprays can contain, in addition to a compound of the invention(s), excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.

The compound of the invention(s) can be alternatively administered by aerosol. This is accomplished by preparing an aqueous aerosol, liposomal preparation or solid particles containing the compound. A nonaqueous (e.g., fluorocarbon propellant) suspension could be used. Sonic nebulizers are preferred because they minimize exposing the agent to shear, which can result in degradation of the compound.

Ordinarily, an aqueous aerosol is made by formulating an aqueous solution or suspension of the agent together with conventional pharmaceutically-acceptable carriers and stabilizers. The carriers and stabilizers vary with the requirements of the particular compound, but typically include nonionic surfactants (T weens, Pluronics, or polyethylene glycol), innocuous proteins like serum albumin, sorbitan esters, oleic acid, lecithin, amino acids such as glycine, buffers, salts, sugars or sugar alcohols. Aerosols generally are prepared from isotonic solutions.

Transdermal patches have the added advantage of providing controlled delivery of a compound of the invention(s) to the body. Such dosage forms can be made by dissolving or dispersing the agent in the proper medium. Absorption enhancers can also be used to increase the flux of the active ingredient across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the active ingredient in a polymer matrix or gel.

Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of the invention.

Pharmaceutical compositions of the invention suitable for parenteral administration comprise one or more compound of the invention(s) in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.

Examples of suitable aqueous and nonaqueous carriers, which may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and

antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.

In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally-administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.

Injectable depot forms are made by forming microencapsule matrices of compound of the invention(s) in biodegradable polymers such as polylactide- polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly( anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue. When the compound of the invention(s) are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically-acceptable carrier.

Regardless of the route of administration selected, the compound of the invention(s), which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art.

Actual dosage levels and time course of administration of the active ingredients in the pharmaceutical compositions of the invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration,

without being toxic to the patient. An exemplary dose range is from 0.01 to 10 mg per day.

A preferred dose of the compound of the invention for the present invention is the maximum that a patient can tolerate and not develop serious or unacceptable side effects. In certain embodiments, the compound of the present invention is administered at a concentration of about 10 micrograms to about 100 mg per kilogram of body weight per day, about 0.1 - about 10 mg/kg or about 1.0 mg - about 10 mg/kg of body weight per day. Ranges intermediate to the above-recited values are also intended to be part of the invention.

5. SCREENING METHODS AND SYSTEMS

In another aspect, the invention provides a method for identifying a compound that inhibits ANT2 activity (e.g., a chemotherapeutic agent), the method comprising obtaining a structure of ANT2 or obtaining information relating to the structure of ANT2, and modeling a test compound into or on the crystal structure coordinates to determine whether the compound inhibits ANT2. In certain embodiments, the step of modeling comprises modeling or determining the ability of the compound to bind to or associate with a binding pocket defined by structure coordinates of one or more ANT2 amino acid residues Lysl05 and ArglOβ. Yet another aspect of the invention is a method for identifying a compound that modulates the activity of ANT2, the method comprising using the atomic coordinates of one or more ANT2 amino acid residues LyslO5 and ArglOό to generate a three-dimensional structure of a molecule comprising an ANT2 binding site or binding pocket, and employing the three-dimensional structure to identify a compound that modulates (e.g., inhibits) the activity of ANT2.

In another aspect, the invention relates to a three-dimensional structure of ANT2. The invention provides key structural features of ANT2.

In another aspect, the invention provides a machine readable storage medium which comprises the structural coordinates of either one or both of the binding pockets identified herein, or similarly shaped, homologous binding pockets. Such storage media encoded with these data are capable of displaying a three-dimensional graphical representation of a molecule or molecular complex which comprises such binding

pockets on a computer screen or similar viewing device. Thus, in one embodiment, the invention provides a machine readable storage medium which comprises the structural coordinates of a binding pocket defined (at least in part) by structure coordinates of one or more of ANT2 amino acid residues LyslO5 and ArglOό, or a homologous binding site or binding pocket.

In another aspect, the invention provides a computer for producing a three- dimensional representation of a molecule or molecular complex, wherein said molecule or molecular complex comprises a binding pocket defined by structural coordinates of a binding pocket defined (at least in part) by structure coordinates of one or more of ANT2 amino acid residues LyslO5 and ArglOό, or a homologous binding site or pocket; or b) a three-dimensional representation of a homologue of said molecule or molecular complex, wherein said homologue comprises a binding pocket that has a root mean square deviation from the backbone atoms of said amino acids of not more than about 2.0 angstroms. The computer includes (i) a machine-readable data storage medium comprising a data storage material encoded with machine- readable data, wherein said data comprises the structural coordinates of a binding pocket defined (at least in part) by structure coordinates of one or more of ANT2 amino acid residues LyslO5 and ArglOό, or a homologous binding site pocket; (ii) a working memory for storing instructions for processing said machine-readable data; (iii) a central-processing unit coupled to said working memory and to said machine- readable data storage medium for processing said machine readable data into said three-dimensional representation; and (iv) a display coupled to said central-processing unit for displaying said three-dimensional representation.

Thus, the computer produces a three-dimensional graphical structure of a molecule or a molecular complex which comprises a binding site or pocket.

In another embodiment, the invention provides a computer for producing a three-dimensional representation of a molecule or molecular complex defined by structure coordinates of all or some of the ANT2 amino acids, or a three-dimensional representation of a homologue of said molecule or molecular complex, wherein said homologue comprises a binding pocket that has a root mean square deviation from the backbone atoms of said amino acids of not more than 2.0 (more preferably not more than 1.5) angstroms.

In exemplary embodiments, the computer or computer system can include components which are conventional in the art, e.g., as disclosed in U.S. Patent No.

5,978,740 and/or 6,183,121 (incorporated herein by reference). For example, a computer system can includes a computer comprising a central processing unit ("CPU"), a working memory (which may be, e.g., RAM (random-access memory) or "core" memory), a mass storage memory (such as one or more disk drives or CD- ROM drives), one or more cathode-ray tube (CRT) or liquid crystal display (LCD) display terminals, one or more keyboards, one or more input lines, and one or more output lines, all of which are interconnected by a conventional system bus.

Machine-readable data of this invention may be inputted to the computer via the use of a modem or modems connected by a data line. Alternatively or additionally, the input hardware may include CD-ROM drives, disk drives or flash memory. In conjunction with a display terminal, a keyboard may also be used as an input device.

Output hardware coupled to the computer by output lines may similarly be implemented by conventional devices. By way of example, output hardware may include a CRT or LCD display terminal for displaying a graphical representation of a binding pocket of this invention using a program such as QUANTA or PYMOL.

Output hardware might also include a printer, or a disk drive to store system output for later use.

In operation, the CPU coordinates the use of the various input and output devices, coordinates data accesses from the mass storage and accesses to and from working memory, and determines the sequence of data processing steps. A number of programs may be used to process the machine-readable data of this invention, including commercially-available software.

A magnetic storage medium for storing machine-readable data according to the invention can be conventional. A magnetic data storage medium can be encoded with a machine-readable data that can be carried out by a system such as the computer system described above. The medium can be a conventional floppy diskette or hard disk, having a suitable substrate which may be conventional, and a suitable coating , which may also be conventional, on one or both sides, containing magnetic domains whose polarity or orientation can be altered magnetically. The medium may also have an opening for receiving the spindle of a disk drive or other data storage device.

The magnetic domains of the medium are polarized or oriented so as to encode in manner which may be conventional, machine readable data such as that described herein, for execution by a system such as the computer system described herein.

An optically-readable data storage medium also can be encoded with machine- readable data, or a set of instructions, which can be carried out by a computer system. The medium can be a conventional compact disk read only memory (CD-ROM) or a rewritable medium such as a magneto-optical disk which is optically readable and magneto-optically writable.

In the case of CD-ROM, as is well known, a disk coating is reflective and is impressed with a plurality of pits to encode the machine-readable data. The arrangement of pits is read by reflecting laser light off the surface of the coating. A protective coating, which preferably is substantially transparent, is provided on top of the reflective coating.

In the case of a magneto-optical disk, as is well known, a data-recording coating has no pits, but has a plurality of magnetic domains whose polarity or orientation can be changed magnetically when heated above a certain temperature, as by a laser. The orientation of the domains can be read by measuring the polarization of laser light reflected from the coating. The arrangement of the domains encodes the data as described above.

Structure data, when used in conjunction with a computer programmed with software to translate those coordinates into the 3-dimensional structure of a molecule or molecular complex comprising a binding pocket, may be used for a variety of purposes, such as drug discovery. The structure can be an ANT structure having the amino acid sequence of any delineated in FIG 4 herein, or fragment thereof.

For example, the structure encoded by the data may be computationally evaluated for its ability to associate with chemical entities. Chemical entities that associate with a binding site or pocket of ANT2 as disclosed herein may inhibit ANT2 activity, and are potential drug candidates. Alternatively, the structure encoded by the data may be displayed in a graphical three-dimensional representation on a computer screen. This allows visual inspection of the structure, as well as visual inspection of the structure's association with chemical entities.

Thus, according to another embodiment, the invention relates to a method for evaluating the potential of a chemical entity to associate with a) a molecule or molecular complex comprising a binding pocket defined, at least in part, by structure coordinates of one or more ANT2 amino acid residues LyslO5 and ArglOβ, as described herein, or b) a homologue of said molecule or molecular complex, wherein said homologue comprises a binding pocket that has a root mean square deviation

from the backbone atoms of said amino acids of not more than 2.0 (more preferably 1.5) angstroms.

This method comprises the steps of: i) employing computational means to perform a fitting operation between the chemical entity and a binding pocket of the molecule or molecular complex; and ii) analyzing the results of the fitting operation to quantify the association between the chemical entity and the binding pocket.

The term "chemical entity", as used herein, refers to chemical compounds, complexes of at least two chemical compounds, and fragments of such compounds or complexes.

In certain embodiments, the method evaluates the potential of a chemical entity to associate with a molecule or molecular complex defined by structure coordinates of all or some of the amino acids of ANT2, as described herein, or a homologue of said molecule or molecular complex having a root mean square deviation from the backbone atoms of said amino acids of not more than 2.0 (more preferably not more than 1.5) angstroms.

In a further embodiment, the structural coordinates one of the binding pockets described herein can be utilized in a method for identifying a potential agonist or antagonist of a molecule comprising an ANT2 binding site or pocket. This method comprises the steps of: a) using the atomic coordinates of ANT2 amino acid residues LyslO5 and ArglOό, as described herein, with a root mean square deviation from the backbone atoms of said amino acids of not more than about 2.0 (more preferably not more than 1.5) angstroms, to generate a three-dimensional structure of molecule comprising an ANT2 binding site or pocket; b) employing the three-dimensional structure to design or select the potential agonist or antagonist. The method further includes the optional steps of c) synthesizing the agonist or antagonist; and d) contacting the agonist or antagonist with the molecule to determine the ability of the potential agonist or antagonist to interact with the molecule.

In another aspect, the invention is a method of identifying a compound useful for inhibiting ANT2 activity comprising the steps of:

1. Obtaining the crystal structure of human ANT2, whereby a three- dimensional structure of ANT2 will be generated;

2. Determination of ANT2 binding regions or pockets by mapping the atomic coordinates of selected amino acid residues, Lys 105 and Arg 106, to provide an ANT2 modeling system, then providing a machine readable storage medium which comprises the three-dimensional ANT2 structure and binding pocket(s) defined (at least in part) by structure coordinates of one or more of ANT2 amino acid residues LyslO5 and ArglOό;

3. Employing the ANT2 modeling system to identify test compounds that bind within the selected ANT2 binding pockets;

4. Determining the selectivity of compound inhibition, i.e. inhibiting ANT2 over ANTl , by measuring the ADP/ ATP exchange for each ANT, following treatment with compound;

5. Identifying compounds that inhibit ANT2 to the extent of cell growth inhibition or cell death.

The present inventors' elucidation of heretofore unknown binding sites or pockets in the structure of ANT2 provides the necessary information for designing new chemical entities and compounds that may interact with ANT2, in whole or in part, and may therefore modulate (e.g., inhibit or decrease) the activity of ANT2, preferably with selectivity relative to other ANTs.

The design of compounds that bind to ANT2 binding sites or pockets according to this invention generally involves consideration of several factors. First, the entity must be capable of physically and structurally associating with parts or all of an ANT2 binding site or pocket. Non-covalent molecular interactions important in this association include hydrogen bonding, van der Waals interactions, hydrophobic interactions and electrostatic interactions. Second, the entity must be able to assume a conformation that allows it to associate with an ANT2 binding site or pocket directly.

Although certain portions of the entity will not directly participate in these associations, those portions of the entity may still influence the overall conformation of the molecule. This, in turn, may have a significant impact on potency. Such conformational requirements include the overall three-dimensional structure and orientation of the chemical entity in relation to all or a portion of the binding pocket, or the spacing between functional groups of an entity comprising several chemical entities that directly interact with the binding pocket or homologues thereof.

The potential inhibitory or binding effect of a chemical entity on an ANT2 binding site or pocket may be analyzed prior to its actual synthesis and testing by the use of computer modeling techniques. If the theoretical structure of the given entity suggests insufficient interaction and association between it and the target binding pocket, testing of the entity is obviated. However, if computer modeling indicates a strong interaction, the molecule may then be synthesized and tested for its ability to bind to a binding pocket. This may be achieved, e.g., by testing the ability of the molecule to inhibit ANT2 activity, e.g., using assays described herein or known in the art. In this manner, synthesis of inoperative compounds may be avoided.

A potential inhibitor of an ANT2 -related binding pocket may be computationally evaluated by means of a series of steps in which chemical entities or fragments are screened and selected for their ability to associate with the ANT2 - related binding pockets.

One skilled in the art may use one of several methods to screen chemical entities or fragments for their ability to associate with an ANT2 binding site or pocket. This process may begin by visual inspection of, for example, an ANT2 binding site or pocket on the computer screen based on the structure coordinates described herein, or other coordinates which define a similar shape generated from the machine-readable storage medium. Selected fragments or chemical entities may then be positioned in a variety of orientations, or docked, within that binding pocket as defined supra. Docking may be accomplished using software such as Quanta and DOCK, followed by energy minimization and molecular dynamics with standard molecular mechanics force fields, such as CHARMM and AMBER.

Specialized computer programs (e.g., as known in the art and/or commercially available and/or as described herein) may also assist in the process of selecting fragments or chemical entities.

Once suitable chemical entities or fragments have been selected, they can be assembled into a single compound or complex. Assembly may be preceded by visual inspection of the relationship of the fragments to each other on the three-dimensional image displayed on a computer screen in relation to the structure coordinates of the target binding pocket.

Instead of proceeding to build a compound capable of binding to a binding pocket in a step-wise fashion one fragment or chemical entity at a time as described above, inhibitory or other binding compounds may be designed as a whole or "de novo" using either an empty binding site or optionally including some portion(s) of a known inhibitor(s). There are many de novo ligand design methods known in the art, some of which are commercially available (e.g., LeapFrog, available from Tripos Associates, St. Louis, Mo.).

Other molecular modeling techniques may also be employed in accordance with this invention (see, e.g., N. C. Cohen et al., "Molecular Modeling Software and Methods for Medicinal Chemistry, J. Med. Chem., 33, pp. 883-894 (1990); see also, M. A. Navia and M. A. Murcko, "The Use of Structural Information in Drug Design", Current Opinions in Structural Biology, 2, pp. 202-210 (1992); L. M. Balbes et al., "A Perspective of Modern Methods in Computer- Aided Drug Design", in Reviews in Computational Chemistry, Vol. 5, K. B. Lipkowitz and D. B. Boyd, Eds., VCH, New York, pp. 337-380 (1994); see also, W. C. Guida, "Software For Structure-Based Drug Design", Curr. Opin. Struct. Biology,, 4, pp. 777-781 (1994)).

Once a compound has been designed or selected, the efficiency with which that entity may bind to a binding pocket may be tested and optimized by computational evaluation. Specific computer software is available in the art to evaluate compound deformation energy and electrostatic interactions. Examples of programs designed for such uses include: AMBER; QUANTA/CHARMM (Accelrys, Inc., Madison, WI) and the like. These programs may be implemented, for instance, using a commercially- available graphics workstation. Other hardware systems and software packages will be known to those skilled in the art.

Another technique involves the in silico screening of virtual libraries of compounds, e.g., as described herein (see, e.g., the Examples herein below). Many thousands of compounds can be rapidly screened and the best virtual compounds can be selected for further screening (e.g., by synthesis and in vitro testing). Small

molecule databases can be screened for chemical entities or compounds that can bind, in whole or in part, to an ANT2 binding site or pocket. In this screening, the quality of fit of such entities to the binding site may be judged either by shape complementarity or by estimated interaction energy. Finally, additional computational techniques can be used for automated structure-based optimization with software packages such as RACHEL (Tripos, Inc.). RACHEL allows a database of fragments to be screened and evaluated (i.e., scored) as each fragment is considered as an extension of the lead compound. The lead compound can then be grown in silico at user defined sites and ranked again. This approach can provide a "filtered" library of derivatives likely to have an increased affinity for the target.

In the methods ddescribed herein, the additional steps of procuring (e.g., purchasing or obtaining from a commercial source or compound library , synthesizing, etc.) the compuond and/or testing the compound in a laboratory protocol (e.g., assay, experiment, etc.) may also be included.

The invention also provides methods for designing, evaluating and identifying compounds which bind to the aforementioned binding site/pocket. Such compounds are potential inhibitors or modulators of ANT2 activity. Other embodiments of the invention are disclosed herein.

The invention is further illustrated by the following examples which should in no way be construed as being further limiting.

EXAMPLES

Materials and Methods

Virtual Screening

The software package of DOCKv5.2 (Ewing et al. 2001) was used for in silico screening of -240,000 compounds available from the National Cancer Institute, Developmental Therapeutics Program. The structure coordinates and chemical

information for each compound were processed either with accessory software from DOCK or with the ZINC server (Irwin and Shoichet 2005).

The grid-based scoring system was used for scoring with the non-bonded force field energy function implemented in DOCK. A standard 6-12 Lennard- Jones potential was used to evaluate van der Waals contacts. Spheres were generated by SPHGEN (Kuntz et al. 1982) and clusters were edited by hand to target specific sites on the molecular surface of ANT2.

EXAMPLE 1 Approximately 240,000 compounds were virtually screened with DOCKv5.2

(E wing et al. 2001) and ranked by energy score. This computer database was prepared with DOCK accessory software (SF2MOL2, UCSF) and Sybyl (Tripos, Inc.).

We first predicted the three-dimensional structure of ANT2 (SWISS-MODEL) based on the solved crystal structure of bovine Antl . The ANT2 structure and the molecular docking site are shown in Figs. 3 & 4 . The docking site was selected based on its unique specificity to ANT2 and not the other ANTs, and on characteristics favorable for small molecule binding. Importantly, the selected docking site is located at the ADP/ ATP entry site. Thus, we predict that the small compound interaction at the site would interfere with ADP/ ATP exchange. Moreover, the site selected for molecular docking overlaps with the site at which HIV-1/Vpr is known to bind . HIV- 1/Vpr is known to induce mtPTP -mediated cellular apoptosis.

EXAMPLE 2

The 24 compounds that had the highest scores for potential ANT2 binding were selected and screened for their potential activities for inhibition of cancer cell growth. The compounds were obtained from NCI Developmental Therapeutics Program.

A human cervical cancer cell line, HeLa, was treated with either vehicle alone (DMSO), each chemical compound (50 μM), or a positive control, bongkrekic acid (BKA, general ANT inhibitor) for 5 days. The cell growth was evaluated and quantitated using a crystal violet staining that measures total DNA amounts. For the

compounds that exhibit an inhibitory effect on cancer cell growth, their dose responses were determined.

Results: Four compounds (#1, 5, 10 and 12) out of 24 inhibited the growth of HeLa cells over 75% at the concentration of 50 μM (Fig. 1). IC 5O of each compound was; 12.5-25 μM, <6.25 μM and 25-50 μM in #1, #5 and #10 compound, respectively (Fig. 2).

The structures of these compounds are shown below:

Also from, TABLE 1

Compound #1:

NSC: 5754 CAS: 83-28-3 Molecular Formula: C 14H 14O3

Molecular Weight: 230.0 g/mol

Compound #5: NSC: 18477

CAS: 5462-46-4

Molecular Formula: C6H20N2O12P4.4Pb

Molecular Weight: 1265.0 g/mol

Compound #10:

NSC: 91852 CAS: 10437-95-3 Molecular Formula: C18H12O4 Molecular Weight: 292.0 g/mol

Conclusion: Three out of the 24 small compounds selected for ANT2 binding showed the growth inhibitory effect on HeLa cells, with various IC 50 .

EXAMPLE 3 Susceptibility of cancer cells v. non -transformed fibroblasts (see, FIG 5): The compounds that showed the greatest cell growth inhibition were selected for dose response determination. NIH 3T3 or HeLa cells were cultured on 24 well plate and treated 24 hrs later with 0, 6.25, 12.5, 25, or 50μM of compounds #1, 5, 10 and DMSO vehicle control . Crystal violet staining was performed by incubating cells in 0.2% crystal violet solution (Fisher Scientific) for 10 min at room temperature. Cells were then washed twice with water.

Susceptibility of cancer cells v. non -transformed fibroblasts (see, FIG 6): Crystal violet stain from FIG 5 (see above) was solubilized by addition of 1% SDS solution to each well followed by gentle agitation on an orbital shaker until color was uniform, with no areas of dense coloration in bottom of wells. Absorbance was measured at 570 nm. Percent survival of NIH 3T3 or HeLa cells was calculated relative to untreated cells. Compound #1 showed a drastic decrease in cell growth at 50 μM with 5% HeLa cell survival compared to 40% cell survival in 3T3 cells. Treatment with compound #5 resulted in 40% and 18% cell survival in HeLa cells compared to 80% and 50% 3T3 cell survival at 12.5 and 25 μM respectively.

The disclosures of each and every patent, patent application and publication cited herein are hereby incorporated herein by reference in their entirety.

The recitation of a listing of chemical groups in any definition of a variable herein includes definitions of that variable as any single group or combination of listed groups. The recitation of an embodiment for a variable herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof.

Although the invention has been disclosed with reference to specific embodiments, it is apparent that other embodiments and variations of the invention may be devised by others skilled in the art without departing from the true spirit and scope of the invention. The sentences are intended to be construed to include all such embodiments and equivalent variations.