WO2007014247A2 - Novel compounds, pharmaceutical compositions containing same, and methods of use for same - Google Patents

Novel compounds, pharmaceutical compositions containing same, and methods of use for same Download PDF

Info

Publication number
WO2007014247A2
WO2007014247A2 PCT/US2006/028979 US2006028979W WO2007014247A2 WO 2007014247 A2 WO2007014247 A2 WO 2007014247A2 US 2006028979 W US2006028979 W US 2006028979W WO 2007014247 A2 WO2007014247 A2 WO 2007014247A2
Authority
WO
WIPO (PCT)
Prior art keywords
subject
alkyl
aryl
alkenyl
alkylaryl
Prior art date
Application number
PCT/US2006/028979
Other languages
French (fr)
Other versions
WO2007014247A3 (en
Inventor
Francis P. Kuhajda
Craig A. Townsend
Susan M. Medghalchi
Jill M. Mcfadden
Original Assignee
Johns Hopkins University
Fasgen, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Johns Hopkins University, Fasgen, Inc. filed Critical Johns Hopkins University
Priority to US12/309,419 priority Critical patent/US20100029761A1/en
Publication of WO2007014247A2 publication Critical patent/WO2007014247A2/en
Publication of WO2007014247A3 publication Critical patent/WO2007014247A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/56Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D307/68Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents

Definitions

  • Fatty acids have three primary roles in the physiology of cells. First, they are the building bocks of biological membranes. Second, fatty acid derivatives serve as hormones and intracellular messengers. Third, and of particular importance to the present invention, fatty acids are fuel molecules that can be stored in adipose tissue as triacylglycerols, which are also known as neutral fats.
  • FAS fatty acid synthase
  • ACC acetyl CoA carboxylase
  • malic enzyme acetyl CoA carboxylase
  • citric lyase The principal enzyme, FAS, catalyzes the NADPH-dependent condensation of the precursors malonyl-CoA and acetyl-CoA to produce fatty acids.
  • NADPH is a reducing agent that generally serves as the essential electron donor at two points in the reaction cycle of FAS.
  • the other three enzymes i.e., ACC, malic enzyme, and citric lyase
  • Other enzymes for example the enzymes that produce NADPH, are also involved in fatty acid synthesis.
  • FAS has an Enzyme Commission (E.C.) No. 2.3.1.85 and is also known as fatty acid synthetase, fatty acid ligase, as well as its systematic name acyl-CoA:malonyl-CoA C- acyltransferase (decarboxylating, oxoacyl- and enoyl-reducing and thioester-hydrolysing).
  • E.C. Enzyme Commission
  • the FAS catalyzed synthesis of fatty acids is similar in lower organisms, such as, for example, bacteria, and in higher organisms, such as, for example, mycobacteria, yeast and humans, there are some important differences.
  • bacteria the seven enzymatic reactions are carried out by seven separate polypeptides that are non-associated. This is classified as Type II FAS.
  • mycobacteria, yeast and humans are carried out by multifunctional polypeptides.
  • yeast have a complex composed of two separate polypeptides whereas in mycobacterium and humans, all seven reactions are carried out by a single polypeptide. These are classified as Type I FAS.
  • FAS inhibitors can be identified by the ability of a compound to inhibit the enzymatic activity of purified FAS.
  • FAS activity can be assayed by measuring the incorporation of radiolabeled precursor (i.e., acetyl-CoA or malonyl-CoA) into fatty acids or by spectrophotometrically measuring the oxidation of NADPH. (DiIs, et al., Methods Enzymol., 35:74-83).
  • Table 1 set forth below, lists several FAS inhibitors.
  • N-ethylmaleimide oxalyl thiol esters such as S-oxalylglutathione gossypol phenylglyoxal
  • TDG 2-tetradecanylglutarate
  • the condensing enzyme of the FAS complex is well characterized in terms of structure and function.
  • the active site of the condensing enzyme contains a critical cysteine thiol, which is the target of antilipidemic reagents, such as, for example, the inhibitor cerulenin.
  • Preferred inhibitors of the condensing enzyme include a wide range of chemical compounds, including alkylating agents, oxidants, and reagents capable of undergoing disulphide exchange.
  • the binding pocket of the enzyme prefers long chain, E, E, dienes.
  • Cerulenin [ ⁇ IS, 3i?)-2,3-epoxy-4-oxo-7,10 dodecadienoyl amide] is an example:
  • Cerulenin covalently binds to the critical cysteine thiol group in the active site of the condensing enzyme of fatty acid synthase, inactivating this key enzymatic step (Funabashi, et al., J. Biochem., 105:751-755, 1989). While cerulenin has been noted to possess other activities, these either occur in microorganisms which may not be relevant models of human cells (e.g., inhibition of cholesterol synthesis in fungi, Omura (1976), Bacterid. Rev., 40:681-697; or diminished RNA synthesis in viruses, Perez, et al.
  • F ⁇ BS, 280: 129-133 occur at a substantially higher drug concentrations (inhibition of viral HIV protease at 5 mg/ml, Moelling, et al. (1990), FEBS, 261 :373-377) or may be the direct result of the inhibition of endogenous fatty acid synthesis (inhibition of antigen processing in B lymphocytes and macrophages, FaIo, et al. (1987), J. Immunol., 139:3918-3923).
  • cerulenin does not specifically inhibit myristoylation of proteins (Simon, et al., J. Biol. Chem., 267:3922-3931, 1992).
  • FAS inhibitors are disclosed in U.S. Patent Application No. 08/096,908 and its CIP filed Jan. 24, 1994, the disclosures of which are hereby incorporated by reference. Included are inhibitors of fatty acid synthase, citrate lyase, acetyl CoA carboxylase, and malic enzyme.
  • Triacsin C (sometimes termed WS- 1228A), a naturally occurring acyl-CoA synthetase inhibitor, which is a product of Streptomyces sp. SK-1894.
  • the chemical structure of Triacsin C is l-hydroxy-3-(E, E, E-2 ⁇ 4',T- undecatrienylidine) triazene.
  • Triacsin C causes 50% inhibition of rat liver acyl-CoA synthetase at 8.7 ⁇ M; a related compound, Triacsin A, inhibits acyl CoA-synthetase by a mechanism which is competitive with long-chain fatty acids. Inhibition of acyl-CoA synthetase is toxic to animal cells.
  • Tomoda et al. (Tomoda el. al., J. Biol. Chem. 266:4214-4219, 1991) teaches that Triacsin C causes growth inhibition in Raji cells at 1.0 ⁇ M, and have also been shown to inhibit growth of Vero and HeIa cells. Tomoda el. al. further teaches that acyl-CoA synthetase is essential in animal cells and that inhibition of the enzyme has lethal effects.
  • the compounds disclosed in the '575 Patent have several advantages over the natural product cerulenin for therapeutic applications: [1] they do not contain the highly reactive epoxide group of cerulenin, [2] they are stable and soluble in aqueous solution, [3] they can be produced by a two-step synthetic reaction and thus easily produced in large quantities, and [4] they are easily tritiated to high specific activity for biochemical and pharmacological analyses.
  • the synthesis of this family of compounds, which are fatty acid synthase inhibitors is described in the '575 Patent, as is their use as a means to treat tumor cells expressing FAS, and their use as a means to reduce body weight.
  • the '575 Patent also discloses the use of any fatty acid synthase inhibitors to systematically reduce adipocyte mass (adipocyte cell number or size) as a means to reduce body weight.
  • mice and humans The primary sites for fatty acid synthesis in mice and humans are the liver (see Roncari, Can. J. Biochem., 52:221-230, 1974; Triscari et al., 1985, Metabolism, 34:580-7; Barakat et al., 1991, Metabolism, 40:280-5), lactating mammary glands (see Thompson, et al., Pediatr. Res., 19:139-143, 1985) and adipose tissue (Goldrick et al., 1974, Clin. Sci. MoI. Med., 46:469-79).
  • Cerulenin was originally isolated as a potential antifungal antibiotic from the culture broth of Cephalosporium caerulens. Structurally cerulenin has been characterized as (2i?,3 ⁇ S)-epoxy-4-oxo-7,10-trans,trans-dodecanoic acid amide. Its mechanism of action has been shown to be inhibition, through irreversible binding, of beta-ketoacyl-ACP synthase, the condensing enzyme required for the biosynthesis of fatty acids. Cerulenin has been categorized as an antifungal, primarily against Candida and Saccharomyces sp.
  • Infectious diseases which are particularly susceptible to treatment are diseases which cause lesions in externally accessible surfaces of the infected animal.
  • Externally accessible surfaces include all surfaces that may be reached by non-invasive means (without cutting or puncturing the skin), including the skin surface itself, mucus membranes, such as those covering nasal, oral, gastrointestinal, or urogenital surfaces, and pulmonary surfaces, such as the alveolar sacs.
  • Susceptible diseases include: (1) cutaneous mycoses or tineas, especially if caused by Microsporum, Trichophyton, Epidermophyton, or Mucocutaneous candidiasis; (2) mucotic keratitis, especially if caused by Aspergillus, Fusarium or Candida; (3) amoebic keratitis, especially if caused by Acanthamoeba; (4) gastrointestinal disease, especially if caused by Giardia lamblia, Entamoeba, Cryptosporidium, Microsporidium, or Candida (most commonly in immunocompromised animals); (5) urogenital infection, especially if caused by Candida albicans or Trichomonas vaginalis; and (6) pulmonary disease, especially if caused by Mycobacterium tuberculosis, Aspergillus, or Pneumocystis carinii. Infectious organisms that are susceptible to treatment with fatty acid synthesis inhibitors include Mycobacterium tuberculosis, especially multiply-
  • Any compound that inhibits fatty acid synthesis may be used to inhibit microbial cell growth.
  • compounds administered to a patient must not be equally toxic to both patient and the target microbial cells. Accordingly, it is beneficial to select inhibitors that only, or predominantly, affect target microbial cells.
  • Eukaryotic microbial cells which are dependent on their own endogenously synthesized fatty acid will express Type I FAS. This is shown both by the fact that FAS inhibitors are growth inhibitory and by the fact that exogenously added fatty acids can protect normal patient cells but not these microbial cells from FAS inhibitors. Therefore, agents which prevent synthesis of fatty acids by the cell may be used to treat infections.
  • fatty acids are synthesized by Type I FAS using the substrates acetyl CoA, malonyl CoA and NADPH.
  • other enzymes which can feed substrates into this pathway may also effect the rate of fatty acid synthesis and thus be important in microbes that depend on endogenously synthesized fatty acid.
  • the product of Type I FAS differs in various organisms. For example, in the fungus S. cerevisiae the products are predominately palmitate and sterate sterif ⁇ ed to coenzyme- A. In Mycobacterium smegmatis, the products are saturated fatty acid CoA esters ranging in length from 16 to 24 carbons. These lipids are often further processed to fulfill the cells need for various lipid components.
  • Inhibition of key steps in down-stream processing or utilization of fatty acids may be expected to inhibit cell function, whether the cell depends on endogenous fatty acid or utilizes fatty acid supplied from outside the cell, and so inhibitors of these down-stream steps may not be sufficiently selective for microbial cells that depend on endogenous fatty acid.
  • Type I fatty acid synthesis inhibitor to such microbes makes them more sensitive to inhibition by inhibitors of down-stream fatty acid processing and/or utilization. Because of this synergy, administration of a fatty acid synthesis inhibitor in combination with one or more inhibitors of down-stream steps in lipid biosynthesis and/or utilization will selectively affect microbial cells that depend on endogenously synthesized fatty acid.
  • Preferred combinations include an inhibitor of FAS and acetyl CoA carboxylase, or FAS and an inhibitor of MAS.
  • the mammal or patient may be treated by administering a fatty acid synthesis inhibitor (Pat No.
  • CPT-I carnitine palmitoyl transferase-1
  • Patent No. 5,759,837 the disclosure of which is hereby incorporated by reference. That application does not describe or disclose any of the compounds disclosed herein.
  • New classes of compounds of formulae I, II, III, and IV have been discovered which have a variety of therapeutically valuable properties, including, FAS -inhibition, NPY- inhibition, CPT-I stimulation, ability to induce weight loss, and anti-cancer and anti-microbial properties.
  • FIG. 1 shows a synthetic scheme to make certain compounds according to the
  • FIG. 2 shows a synthetic scheme to make certain compounds according to the invention.
  • the compounds of the invention can be prepared by conventional means. The synthesis of a number of the compounds is described in the examples. The compounds may be useful for the treatment of obesity, cancer, or microbially-based infections.
  • R 1 H, or C 1 -C 20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms
  • R 2 H, or Ci-C 2O alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms; with the proviso that when R 1 is -(CH 2 ) 5 CH 3 or -(CH 2 ) I0 CH 3 , R 2 is not -CH 2 CH 3 , and the further proviso that when R 1 is -CH 2 CH 3 , R 2 is not -CH 2 CH 3 , and the further proviso that when R 1 is -C 6 H 5 , R 2 is not -CH 2 CH 3 ; with the further proviso that when R 1 is -CH(CH 3 ) 2 , R 2 is not -CH 2 CH 3 ; with the
  • R 1 is -(CH 2 ) 7 CH 3
  • R 2 is -CH 2 CH 3 .
  • R 1 is -(CH 2 ) 3 CH 3
  • R 2 is -CH 2 CH 3 .
  • R 1 and R 2 have the meanings given above, but where when R 2 is -CH 2 CH 3 , R 1 is not Q- C 3 alkyl, optionally substituted with halogen atoms, and when R 2 is -CH 2 CHs, R 1 is not C 2 - alkenyl or C 6 -aryl, optionally substituted with halogen atoms, when R 2 is H, R 1 is not C 1 -C 2 alkyl or C 6 -aryl, optionally substituted with halogen atoms.
  • Another embodiment of the invention is compounds having the following general
  • R 3 H, or C]-C 20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms;
  • R 4 H, or C 1 -C 20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms;
  • R 5 H 5 or Ci-C 20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms, -C(O)R 6 , where R 6 is H, Cj-C 2O alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms, or -OH, -OR 7 or -NR 7 , where R 7 is H, Ci-C 2O alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms; with the proviso that when R 5 is H and R 3 is -CH 2 CH 3 , R 4 is not -CH 2 CH 3 ; and the further proviso that when R 3 is -CH 3 , and R 5 is H, R 4 is not -
  • R 3 is -(CHs) 2 CH 3 , R 4 is not -CH 2 CH 3 ; with the further proviso that when R 5 is H, and R 3 is R 4 is not - CH 3 ; and the further proviso that when R 3 is -CH 3 , and R 5 is H, R 4 is not -CH 3 ; with the further proviso that when R 5 is H and R 3 is -CH 3 , R 4 is not -C(CH 3 ) 3 and the further proviso that when R 5 is -CH 3 and R 3 is -C 6 Hs, R 4 is not -CH 3 ; and the further proviso that when R 5 is -CH 3 and R 3 is -CH 3 , R 4 is not -CH 3 ; and the further proviso that when R 5 is -CH 3 , and R 3 is -CH 2 (CO)OCH 2 CH 3 , R 4 is
  • Another embodiment of this invention is a pharmaceutical composition comprising a pharmaceutical diluent and a compound of formula V:
  • R 8 H, or C J -C 2O alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms;
  • R 9 H, or C 1 -C 20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms.
  • the compounds of formulas I and II above may also be included in a pharmaceutical composition.
  • R l ⁇ H, or Ci-C 2O alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms
  • R u H, or C1-C2 0 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms;
  • R 12 H, or C)-C 2O alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms, -C(O)R 13 , where R 13 is H 5 Ci-C 20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms, or -OH, -OR 14 or -NR 14 , where R 14 is H, CJ-C 20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms.
  • compositions of the present invention can be presented for administration to humans and other animals in unit dosage forms, such as tablets, capsules, pills, powders, granules, sterile parenteral solutions or suspensions, oral solutions or suspensions, oil in water and water in oil emulsions containing suitable quantities of the compound, suppositories and in fluid suspensions or solutions.
  • unit dosage forms such as tablets, capsules, pills, powders, granules, sterile parenteral solutions or suspensions, oral solutions or suspensions, oil in water and water in oil emulsions containing suitable quantities of the compound, suppositories and in fluid suspensions or solutions.
  • pharmaceutical diluent and “pharmaceutical carrier,” have the same meaning.
  • solid or fluid unit dosage forms can be prepared.
  • the compound can be mixed with conventional ingredients such as talc, magnesium stearate, dicalcium phosphate, magnesium aluminum silicate, calcium sulfate, starch, lactose, acacia, methylcellulose and functionally similar materials as pharmaceutical diluents or carriers.
  • Capsules are prepared by mixing the compound with an inert pharmaceutical diluent and filling the mixture into a hard gelatin capsule of appropriate size.
  • Soft gelatin capsules are prepared by machine encapsulation of a slurry of the compound with an acceptable vegetable oil, light liquid petrolatum or other inert oil.
  • Fluid unit dosage forms or oral administration such as syrups, elixirs, and suspensions can be prepared.
  • the forms can be dissolved in an aqueous vehicle together with sugar, aromatic flavoring agents and preservatives to form a syrup.
  • Suspensions can be prepared with an aqueous vehicle with the aid of a suspending agent such as acacia, tragacanth, methylcellulose and the like.
  • parenteral administration fluid unit dosage forms can be prepared utilizing the compound and a sterile vehicle.
  • the compound can be dissolved in water for injection and filter sterilized before filling into a suitable vial or ampoule and sealing.
  • Adjuvants such as a local anesthetic, preservative and buffering agents can be dissolved in the vehicle.
  • the composition can be fro2;en after filling into a vial and the water removed under vacuum. The lyophilized powder can then be scaled in the vial and reconstituted prior to use.
  • the clinical therapeutic indications envisioned for the compounds of the invention include: (1) infections due to invasive micro-organisms such as staphylococci and enterococci; (2) cancers, and (3) obesity due to the ingestion of excess calories. Dose and duration of therapy will depend on a variety of factors, including (1) the patient's age, body weight, and organ function (e ⁇ g., liver and kidney function); (2) the nature and extent of the disease process to be treated, as well as any existing significant co-morbidity and concomitant medications being taken, and (3) drug-related parameters such as the route of administration, the frequency and duration of dosing necessary to effect a cure, and the therapeutic index of the drug. In general, doses will be chosen to achieve serum levels of 1 ng/ml to 100ng/ml with the goal of attaining effective concentrations at the target site of approximately 1 ⁇ g/ml to 10 ⁇ g/ml. EXAMPLES
  • a series of compounds according to the invention were synthesized as described below.
  • Biological activity of certain compounds were profiled as follows: Each compound was tested for at least some of the following properties: [1] inhibition of purified human FAS, [2] inhibition of fatty acid synthesis activity in whole cells and [3] cytotoxicity against cultured MCF-7 human breast cancer cells, known to possess high levels of FAS and fatty acid synthesis activity, using the crystal violet and XTT assays. Select compounds with low levels of cytotoxicity were then tested for weight loss in Balb/C mice. Certain compounds were also tested for activity against gram positive and/or negative bacteria. .
  • Ten Tl 50 flasks of confluent cells are lysed with 1.5 ml lysis buffer (20 mM Tris- HCl, pH 7.5, 1 mM EDTA 5 0.1 mM phenylmethanesulfonyl fluoride (PMSF), 0.1% Igepal CA- 630) and dounce homogenized on ice for 20 strokes.
  • the lysate is centrifuged in JA-20 rotor (Beckman) at 20,000 rpm for 30 minutes at 4 0 C and the supernatant is brought to 42 ml with lysis buffer.
  • a solution of 50% PEG 8000 in lysis buffer is added slowly to the supernatant to a final concentration of 7.5%.
  • the solution is centrifuged in JA-20 rotor (Beckman) at 15,000 rpm for 30 minutes at 4 0 C.
  • Solid PEG 8000 is then added to the supernatant to a final concentration of 15%.
  • the pellet is resuspended overnight at 4 0 C in 10 ml of Buffer A (20 mM K 2 HPO 4 , pH 7.4).
  • Buffer A (20 mM K 2 HPO 4 , pH 7.4
  • the protein solution is applied to a Mono Q 5/5 anion exchange column (Pharmacia).
  • FAS activity is measured by monitoring the malonyl-CoA dependent oxidation of NADPH spectrophotometrically at OD 340 in 96-well plates (DiIs et al and Arslanian et al, 1975). Each well contains 2 ⁇ g purified FAS, 100 mM K 2 HPO 4 , pH 6.5, 1 mM dithiothreitol (Sigma), and 187.5 ⁇ M /3-NADPH (Sigma). Stock solutions of inhibitors are prepared in DMSO at 2, 1, and 0.5 mg/ml resulting in final concentrations of 20, 10, and 5 ⁇ g/ml when 1 ⁇ l of stock is added per well. For each experiment, cerulenin (Sigma) is run as a positive control along with DMSO controls, inhibitors, and blanks (no FAS enzyme) all in duplicate.
  • the assay is performed on a Molecular Devices SpectraMax Plus
  • Spectrophotometer The plate containing FAS, buffers, inhibitors, and controls are placed in the spectrophotometer heated to 37 0 C. Using the kinetic protocol, the wells are blanked on duplicate wells containing 100 ⁇ l of 100 mM K 2 HPO 4 , pH 6.5 and the plate is read at OD 340 at 10 sec intervals for 5 minutes to measure any malonyl-CoA independent oxidation of NADPH. The plate is removed from the spectrophotometer and malonyl-CoA (67.4 ⁇ M, final concentration per well) and alkynyl-CoA (61.8 ⁇ M, final concentration per well) are added to each well except to the blanks.
  • malonyl-CoA 67.4 ⁇ M, final concentration per well
  • alkynyl-CoA (61.8 ⁇ M, final concentration per well
  • the plate is read again as above with the kinetic protocol to measure the malonyl- CoA dependent NADPH oxidation.
  • the difference between the ⁇ OD 340 for the malonyl-CoA dependent and non-malonyl-CoA dependent NADPH oxidation is the specific FAS activity. Because of the purity of the FAS preparation, non-malonyl-CoA dependent NADPH oxidation is negligible.
  • the IC 50 for the compounds against FAS is determined by plotting the ⁇ OD 340 for each inhibitor concentration tested, performing linear regression and computing the best-fit line, r 2 values, and 95% confidence intervals.
  • the concentration of compound yielding 50% inhibition of FAS is the IC 50 .
  • Graphs of ⁇ OD 340 versus time are plotted by the SOFTmax PRO software (Molecular Devices) for each compound concentration. Computation of linear regression, best-fit line, r 2 , and 95% confidence intervals are calculated using Prism Version 3.0 (Graph Pad Software).
  • the crystal violet assay measures cell growth but not cytotoxicity. This assay employs crystal violet staining of fixed cells in 96-well plates with subsequent solubilization and measurement of OD 490 on a spectrophotometer. The OD 490 corresponds to cell growth per unit time measured. Cells are treated with the compounds of interest or vehicle controls and IC 50 for each compound is computed.
  • the XTT assay is a non-radioactive alternative for the [ 51 Cr] release cytotoxicity assay.
  • XTT is a tetrazolium salt that is reduced to a formazan dye only by metabolically active, viable cells. The reduction of XTT is measured spectrophotometrically as OD 490 - OD 650 .
  • 9 x 10 3 MCF-7 human breast cancer cells obtained from the American Type Culture Collection are plated per well in 96 well plates in DMEM medium with 10% fetal bovine serum, insulin, penicillin, and streptomycin. Following overnight culture at 37 0 C and 5% CO 2 , the compounds to be tested, dissolved in DMSO, are added to the wells in 1 ⁇ l volume at the following concentrations: 80, 40, 20, 10, 5, 2.5, 1.25, and 0.625 ⁇ g/ml in triplicate. Additional concentrations are tested if required. 1 ⁇ l of DMSO is added to triplicate wells are the vehicle control. C75 is run at 40, 20, 10, 15, 12.5, 10, and 5 ⁇ g/ml in triplicate as positive controls.
  • XTT Cell Proliferation Kit II
  • XTT Cell Proliferation Kit II
  • Plates are read at OD 490 and OD 65 o on a Molecular Devices SpectraMax Plus Spectrophotometer. Three wells containing the XTT reagent without cells serve as the plate blank. XTT data are reported as OD 490 - OD 65 o. Averages and standard error of the mean are computed using SOFTmax Pro software (Molecular Dynamics).
  • the IC 50 for the compounds is defined as the concentration of drug leading to a 50% reduction in OD49C) - OD 65 O compared to controls.
  • OD 4 9 0 - OD 6S o are computed by the SOFTmax PRO software (Molecular Devices) for each compound concentration.
  • IC 50 is calculated by linear regression, plotting the FAS activity as percent of control versus drug concentrations. Linear regression, best-fit line, r 2 , and 95% confidence intervals are determined using Prism Version 3.0 (Graph Pad Software).
  • This assay measures the incorporation of [ 14 C]acetate into total lipids and is a measure of fatty acid synthesis pathway activity in vitro. It is utilized to measure inhibition of fatty acid synthesis in vitro.
  • MCF-7 human breast cancer cells cultured as above are plated at 5 x 10 4 cells per well in 24-well plates. Following overnight incubation, the compounds to be tested, solubilized in DMSO, are added at 5, 10, and 20 ⁇ g/ml in triplicate, with lower concentrations tested if necessary. DMSO is added to triplicate wells for a vehicle control. C75 is run at 5 and 10 ⁇ g/ml in triplicate as positive controls. After 4 hours of incubation, 0.25 ⁇ Ci of [ 14 C] acetate (10 ⁇ l volume) is added to each well.
  • the IC 50 for the compounds is defined as the concentration of drug leading to a 50% reduction in [ I4 C]acetate incorporation into lipids compared to controls. This is determined by plotting the average cpm for each inhibitor concentration tested, performing linear regression and computing the best-fit line, r 2 values, and 95% confidence intervals. The average cpm values are computed by the Beckman scintillation counter (Model LS6500) for each compound concentration. Computation of linear regression, best-fit line, r 2 , and 95% confidence intervals are calculated using Prism Version 3.0 (Graph Pad Software).
  • mice (Jackson Labs) are utilized for the initial weight loss screening. Animals are housed in temperature and 12 hour day/night cycle rooms and fed mouse chow and water ad lib. Three mice are utilized for each compound tested with vehicle controls in triplicate per experiment. For the experiments, mice are housed separately for each compound tested three mice to a cage. Compounds are diluted in DMSO at 10 mg/ml and mice are injected intraperitoneally with 60 mg/kg in approximately 100 ⁇ l of DMSO or with vehicle alone. Mice are observed and weighed daily; average weights and standard errors are computed with Excel (Microsoft). The experiment continues until treated animals reach their pretreatment weights. Select compounds are tested in animals housed in metabolic cages. Dosing of animals are identical to the screening experiments with three animals to a single metabolic cage. Animal weights, water and food consumption, and urine and feces production are measured
  • a broth microdilution assay is used to assess the antimicrobial activity of the compounds. Compounds are tested at twofold serial dilutions, and the concentration that inhibits visible growth (OD 6O o at 10% of control) is defined as the MIC. Microorganisms tested include Staphylococcus aureus (ATCC # 29213), Enterococcus faecalis (ATCC # 29212), Pseudomonas aeruginosa (ATCC # 27853), and Escherichia coli (ATCC # 25922). The assay is performed in two growth media, Mueller Hinton Broth and Trypticase Soy Broth.
  • a blood (Tsoy/5% sheep blood) agar plate is inoculated from frozen stocks maintained in T soy broth containing 10% glycerol and incubated overnight at 37° C. Colonies are suspended in sterile broth so that the turbidity matches the turbidity of a 0.5 McFarland standard. The inoculum is diluted 1 :10 in sterile broth (Mueller Hinton or Trypticase soy) and 195 ul is dispensed per well of a 96- well plate. The compounds to be tested, dissolved in DMSO, are added to the wells in 5 ul volume at the following concentrations: 25, 12.5, 6.25, 3.125, 1.56 and 0.78 ug/ml in duplicate.
  • OD 600 OD 600 on a Molecular Devices SpectraMax Plus Spectrophotometer. Average OD 60O values are computed using SOFTmax Pro Software (Molecular Devices) and MIC values are determined by linear regression analysis using Prism version 3.02 (Graph Pad Software, San Diego). The MIC is defined as the concentration of compound required to produce an OD 60O reading equivalent to 10% of the vehicle control reading.
  • a 24 well plate with 1 ml per well was prepared with 2.5 x 105 cells /well. The cells were incubated O/N.
  • a solubilized palmitate solution was prepared. 50 ⁇ ls Of(I- 14 C) Palmitic acid was added to a 2 ml centrifuge tube and dried under nitrogen gas. 2 mis of ⁇ -CD ( ⁇ -Cyclodextran)- 10 mg/ml in 10 mM Tris were added. This solution was incubated in a 37 0 C water bath for 30 minutes.
  • a hot mix was prepared by adding 25 ⁇ l of this solution to 2.5 ⁇ ls of 200 ⁇ M Carnitine and 222.5 ⁇ l of serum free medium that is used for cells.
  • the cells were then treated with the test compound in triplicate, and incubated at 37°C for 60 minutes.
  • the medium was removed and 250 ⁇ l of the hot mix were added.
  • the test compound was added again, and further incubated at 37°C for 60 minutes.
  • the reaction was stopped with 50 ⁇ l of 2.6 N HClO 4 .
  • the contents of the plate were transferred to a 1.5 ml centrifuge tube, and 50 ⁇ l of 4N KOH were then added, and the tube incubated in a 6O 0 C water bath for 30 min.
  • Sodium acetate (IM, 75 ⁇ l) and sulfuric acid (3N, 50 ⁇ l) were added to the solution and vortexed.
  • the tube was spun for 7 minutes at 1000 rpm at room temperature.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Child & Adolescent Psychology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

A pharmaceutical diluent and a compound of formula VI: wherein: R10 = H, or C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms; R11 = H3 or C]-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms; R12 = H, or C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms, -C(O)R13, where R13 is H, Ci-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms, or -OH, -OR14 or -NR14, where R14 is H, C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms.

Description

JNOVEl, CUMJfUUNDiS. PHARMACEUTICAL COMPOSITIONS CONTAINING SAME, AND METHODS OF USE FOR SAME
BACKGROUND OF THE INVENTION
Fatty acid synthase
Fatty acids have three primary roles in the physiology of cells. First, they are the building bocks of biological membranes. Second, fatty acid derivatives serve as hormones and intracellular messengers. Third, and of particular importance to the present invention, fatty acids are fuel molecules that can be stored in adipose tissue as triacylglycerols, which are also known as neutral fats.
There are four primary enzymes involved in the fatty acid synthetic pathway, fatty acid synthase (FAS), acetyl CoA carboxylase (ACC), malic enzyme, and citric lyase. The principal enzyme, FAS, catalyzes the NADPH-dependent condensation of the precursors malonyl-CoA and acetyl-CoA to produce fatty acids. NADPH is a reducing agent that generally serves as the essential electron donor at two points in the reaction cycle of FAS. The other three enzymes (i.e., ACC, malic enzyme, and citric lyase) produce the necessary precursors. Other enzymes, for example the enzymes that produce NADPH, are also involved in fatty acid synthesis.
FAS has an Enzyme Commission (E.C.) No. 2.3.1.85 and is also known as fatty acid synthetase, fatty acid ligase, as well as its systematic name acyl-CoA:malonyl-CoA C- acyltransferase (decarboxylating, oxoacyl- and enoyl-reducing and thioester-hydrolysing). There are seven distinct enzymes - or catalytic domains - involved in the FAS catalyzed synthesis of fatty acids: acetyl transacylase, malonyl transacylase, beta-ketoacyl synthetase (condensing enzyme), beta-ketoacyl reductase, beta-hydroxyacyl dehydrase, enoyl reductase, and thioesterase. (Wakil, S. J., Biochemistry, 28: 4523-4530, 1989). All seven of these enzymes together form FAS.
Although the FAS catalyzed synthesis of fatty acids is similar in lower organisms, such as, for example, bacteria, and in higher organisms, such as, for example, mycobacteria, yeast and humans, there are some important differences. In bacteria, the seven enzymatic reactions are carried out by seven separate polypeptides that are non-associated. This is classified as Type II FAS. In contrast, the enzymatic reactions in mycobacteria, yeast and humans are carried out by multifunctional polypeptides. For example, yeast have a complex composed of two separate polypeptides whereas in mycobacterium and humans, all seven reactions are carried out by a single polypeptide. These are classified as Type I FAS.
FAS inhibitors
Various compounds have been shown to inhibit fatty acid synthase (FAS). FAS inhibitors can be identified by the ability of a compound to inhibit the enzymatic activity of purified FAS. FAS activity can be assayed by measuring the incorporation of radiolabeled precursor (i.e., acetyl-CoA or malonyl-CoA) into fatty acids or by spectrophotometrically measuring the oxidation of NADPH. (DiIs, et al., Methods Enzymol., 35:74-83). Table 1, set forth below, lists several FAS inhibitors.
Table 1
Representative Inhibitors Of The Enzvmes Of The Fatty Acid Synthesis Pathway
Inhibitors of Fatty Acid Synthase I
1 ,3-dibromopropanone cerulenin
Ellman's reagent (5,5'-dithiobis(2-nitrobenzoic phenyocerulenin acid), DTNB) melarsoprol
4-(4'-chlorobenzyloxy) benzyl nicotinate (KCD- iodoacetate
232) phenylarsineoxide
4-(4'-chlorobenzyloxy) benzoic acid (Mil) pentostam
2(5(4-chlorophenyl)pentyl)oxirane-2- melittin carboxylate (POCA) and its CoA derivative thiolactomycin ethoxyformic anhydride
Inhibitors for citrate lyase Inhibitors for malic enzyme
(-) hydroxycitrate periodate-oxidized 3-aminopyridine adenine
(R,S)-S-(3,4-dicarboxy-3-hydroxy-3-methyl- dinucleotide phosphate butyl)-CoA 5,5'-dithiobis(2-nitrobenzoic acid)
S-carboxymethyl-CoA p-hydroxymercuribenzoate
N-ethylmaleimide oxalyl thiol esters such as S-oxalylglutathione gossypol phenylglyoxal
2,3-butanedione bromopyruvate pregnenolone
Inhibitors for acetyl CoA carboxylase sethoxydim 9-decenyl-l-pentenedioic acid haloxyfop and its CoA ester decanyl-2-pentenedioic acid diclofop and its CoA ester decanyl-1-pentenedioic acid clethodim (S)-ibuprofenyl-CoA alloxydim (R)-ibuprofenyl-CoA trifop fluazifop and its CoA ester clofibric acid clofop
2,4-D mecoprop 5-(tetradecycloxy)-2-furoic acid dalapon beta, beta'-tetramethylhexadecanedioic acid
2-alkyl glutarate tralkoxydim
2-tetradecanylglutarate (TDG) free or monothioester of beta, beta prime-
2-octylglutaric acid methyl-substituted hexadecanedioic acid
N6,02-dibutyryl adenosine cyclic 3 ',5'- (MEDICA 16) monophosphate alpha-cyanco-4-hydroxycinnamate
N2,02-dibutyryl guanosine cyclic 3',5'- S-(4-bromo-2,3-dioxobutyl)-CoA monophosphate p-hydroxymercuribenzoate (PHMB)
CoA derivative of 5-(tetradecyloxy)-2-furoic N6,02-dibutyryl adenosine cyclic 3',5'- acid (TOFA) monophosphate
2,3,7,8-tetrachlorodibenzo-p-dioxin υi tne tour enzymes in the fatty acid synthetic pathway, FAS is the preferred target for inhibition because it acts only within the pathway to fatty acids, while the other three enzymes are implicated in other cellular functions. Therefore, inhibition of one of the other three enzymes is more likely to affect normal cells. Of the seven enzymatic steps carried out by FAS, the step catalyzed by the condensing enzyme {i.e., beta-ketoacyl synthetase) and the enoyl reductase have been the most common candidates for inhibitors that reduce or stop fatty acid synthesis. The condensing enzyme of the FAS complex is well characterized in terms of structure and function. The active site of the condensing enzyme contains a critical cysteine thiol, which is the target of antilipidemic reagents, such as, for example, the inhibitor cerulenin.
Preferred inhibitors of the condensing enzyme include a wide range of chemical compounds, including alkylating agents, oxidants, and reagents capable of undergoing disulphide exchange. The binding pocket of the enzyme prefers long chain, E, E, dienes.
In principal, a reagent containing the sidechain diene and a group which exhibits reactivity with thiolate anions could be a good inhibitor of the condensing enzyme. Cerulenin [{IS, 3i?)-2,3-epoxy-4-oxo-7,10 dodecadienoyl amide] is an example:
Figure imgf000005_0001
Cerulenin covalently binds to the critical cysteine thiol group in the active site of the condensing enzyme of fatty acid synthase, inactivating this key enzymatic step (Funabashi, et al., J. Biochem., 105:751-755, 1989). While cerulenin has been noted to possess other activities, these either occur in microorganisms which may not be relevant models of human cells (e.g., inhibition of cholesterol synthesis in fungi, Omura (1976), Bacterid. Rev., 40:681-697; or diminished RNA synthesis in viruses, Perez, et al. (1991), FΕBS, 280: 129-133), occur at a substantially higher drug concentrations (inhibition of viral HIV protease at 5 mg/ml, Moelling, et al. (1990), FEBS, 261 :373-377) or may be the direct result of the inhibition of endogenous fatty acid synthesis (inhibition of antigen processing in B lymphocytes and macrophages, FaIo, et al. (1987), J. Immunol., 139:3918-3923). Some data suggest that cerulenin does not specifically inhibit myristoylation of proteins (Simon, et al., J. Biol. Chem., 267:3922-3931, 1992).
Several more FAS inhibitors are disclosed in U.S. Patent Application No. 08/096,908 and its CIP filed Jan. 24, 1994, the disclosures of which are hereby incorporated by reference. Included are inhibitors of fatty acid synthase, citrate lyase, acetyl CoA carboxylase, and malic enzyme.
Tomoda and colleagues (Tomoda et..al., Biochim. Biophys. Act 921:595-598 1987; Omura el. al., J. Antibiotics 39:1211-1218 1986) describe Triacsin C (sometimes termed WS- 1228A), a naturally occurring acyl-CoA synthetase inhibitor, which is a product of Streptomyces sp. SK-1894. The chemical structure of Triacsin C is l-hydroxy-3-(E, E, E-2\4',T- undecatrienylidine) triazene. Triacsin C causes 50% inhibition of rat liver acyl-CoA synthetase at 8.7 μM; a related compound, Triacsin A, inhibits acyl CoA-synthetase by a mechanism which is competitive with long-chain fatty acids. Inhibition of acyl-CoA synthetase is toxic to animal cells. Tomoda et al. (Tomoda el. al., J. Biol. Chem. 266:4214-4219, 1991) teaches that Triacsin C causes growth inhibition in Raji cells at 1.0 μM, and have also been shown to inhibit growth of Vero and HeIa cells. Tomoda el. al. further teaches that acyl-CoA synthetase is essential in animal cells and that inhibition of the enzyme has lethal effects.
A family of compounds (gamma-substituted-alpha-rnethylene-beta-carboxy- gamma-butyrolactones) has been shown in U.S. Patent No. 5,981,575 (the disclosure of which is hereby incorporated by reference) to inhibit fatty acid synthesis, inhibit growth of tumor cells, and induce weight loss. The compounds disclosed in the '575 Patent have several advantages over the natural product cerulenin for therapeutic applications: [1] they do not contain the highly reactive epoxide group of cerulenin, [2] they are stable and soluble in aqueous solution, [3] they can be produced by a two-step synthetic reaction and thus easily produced in large quantities, and [4] they are easily tritiated to high specific activity for biochemical and pharmacological analyses. The synthesis of this family of compounds, which are fatty acid synthase inhibitors, is described in the '575 Patent, as is their use as a means to treat tumor cells expressing FAS, and their use as a means to reduce body weight. The '575 Patent also discloses the use of any fatty acid synthase inhibitors to systematically reduce adipocyte mass (adipocyte cell number or size) as a means to reduce body weight.
The primary sites for fatty acid synthesis in mice and humans are the liver (see Roncari, Can. J. Biochem., 52:221-230, 1974; Triscari et al., 1985, Metabolism, 34:580-7; Barakat et al., 1991, Metabolism, 40:280-5), lactating mammary glands (see Thompson, et al., Pediatr. Res., 19:139-143, 1985) and adipose tissue (Goldrick et al., 1974, Clin. Sci. MoI. Med., 46:469-79).
Inhibitors of fatty acid synthesis as antimicrobial agents Cerulenin was originally isolated as a potential antifungal antibiotic from the culture broth of Cephalosporium caerulens. Structurally cerulenin has been characterized as (2i?,3ιS)-epoxy-4-oxo-7,10-trans,trans-dodecanoic acid amide. Its mechanism of action has been shown to be inhibition, through irreversible binding, of beta-ketoacyl-ACP synthase, the condensing enzyme required for the biosynthesis of fatty acids. Cerulenin has been categorized as an antifungal, primarily against Candida and Saccharomyces sp. In addition, some in vitro activity has been shown against some bacteria, actinomycetes, and mycobacteria, although no activity was found against Mycobacterium tuberculosis. The activity of fatty acid synthesis inhibitors and cerulenin in particular has not been evaluated against protozoa such as Toxoplasma gondii or other infectious eucaryotic pathogens such as Pneumocystis carinii, Giardia lamblia, Plasmodium sp., Trichomonas vaginalis, Cryptosporidium, Trypanosoma, Leishmania, and Schistosoma.
Infectious diseases which are particularly susceptible to treatment are diseases which cause lesions in externally accessible surfaces of the infected animal. Externally accessible surfaces include all surfaces that may be reached by non-invasive means (without cutting or puncturing the skin), including the skin surface itself, mucus membranes, such as those covering nasal, oral, gastrointestinal, or urogenital surfaces, and pulmonary surfaces, such as the alveolar sacs. Susceptible diseases include: (1) cutaneous mycoses or tineas, especially if caused by Microsporum, Trichophyton, Epidermophyton, or Mucocutaneous candidiasis; (2) mucotic keratitis, especially if caused by Aspergillus, Fusarium or Candida; (3) amoebic keratitis, especially if caused by Acanthamoeba; (4) gastrointestinal disease, especially if caused by Giardia lamblia, Entamoeba, Cryptosporidium, Microsporidium, or Candida (most commonly in immunocompromised animals); (5) urogenital infection, especially if caused by Candida albicans or Trichomonas vaginalis; and (6) pulmonary disease, especially if caused by Mycobacterium tuberculosis, Aspergillus, or Pneumocystis carinii. Infectious organisms that are susceptible to treatment with fatty acid synthesis inhibitors include Mycobacterium tuberculosis, especially multiply-drug resistant strains, and protozoa such as Toxoplasma.
Any compound that inhibits fatty acid synthesis may be used to inhibit microbial cell growth. However, compounds administered to a patient must not be equally toxic to both patient and the target microbial cells. Accordingly, it is beneficial to select inhibitors that only, or predominantly, affect target microbial cells.
Eukaryotic microbial cells which are dependent on their own endogenously synthesized fatty acid will express Type I FAS. This is shown both by the fact that FAS inhibitors are growth inhibitory and by the fact that exogenously added fatty acids can protect normal patient cells but not these microbial cells from FAS inhibitors. Therefore, agents which prevent synthesis of fatty acids by the cell may be used to treat infections. In eukaryotes, fatty acids are synthesized by Type I FAS using the substrates acetyl CoA, malonyl CoA and NADPH. Thus, other enzymes which can feed substrates into this pathway may also effect the rate of fatty acid synthesis and thus be important in microbes that depend on endogenously synthesized fatty acid. Inhibition of the expression or activity of any of these enzymes will effect growth of the microbial cells that are dependent upon endogenously synthesized fatty acid. The product of Type I FAS differs in various organisms. For example, in the fungus S. cerevisiae the products are predominately palmitate and sterate sterifϊed to coenzyme- A. In Mycobacterium smegmatis, the products are saturated fatty acid CoA esters ranging in length from 16 to 24 carbons. These lipids are often further processed to fulfill the cells need for various lipid components.
Inhibition of key steps in down-stream processing or utilization of fatty acids may be expected to inhibit cell function, whether the cell depends on endogenous fatty acid or utilizes fatty acid supplied from outside the cell, and so inhibitors of these down-stream steps may not be sufficiently selective for microbial cells that depend on endogenous fatty acid. However, it has been discovered that administration of Type I fatty acid synthesis inhibitor to such microbes makes them more sensitive to inhibition by inhibitors of down-stream fatty acid processing and/or utilization. Because of this synergy, administration of a fatty acid synthesis inhibitor in combination with one or more inhibitors of down-stream steps in lipid biosynthesis and/or utilization will selectively affect microbial cells that depend on endogenously synthesized fatty acid. Preferred combinations include an inhibitor of FAS and acetyl CoA carboxylase, or FAS and an inhibitor of MAS.
When it has been determined that a mammal is infected with cells of an organism which expresses Type I FAS, or if FAS has been found in a biological fluid from a patient, the mammal or patient may be treated by administering a fatty acid synthesis inhibitor (Pat No.
5,614,551).
The inhibition of neuropeptide- Y to depress appetite and stimulate weight loss is described in International Patent Application No. PCT/USOl/05316 the disclosure of which is hereby incorporated by reference. That application, however, does not describe or disclose any of the compounds disclosed in the present application
The stimulation of carnitine palmitoyl transferase-1 (CPT-I) to stimulate weight loss is described in U.S. Patent Application Serial No. 60/354,480, the disclosure of which is hereby incorporated by reference. That application does not describe or disclose any of the compounds disclosed herein, either.
The use of FAS inhibitors to inhibit the growth of cancer cells is described in U.S.
Patent No. 5,759,837, the disclosure of which is hereby incorporated by reference. That application does not describe or disclose any of the compounds disclosed herein.
Summary of the Invention
New classes of compounds of formulae I, II, III, and IV have been discovered which have a variety of therapeutically valuable properties, including, FAS -inhibition, NPY- inhibition, CPT-I stimulation, ability to induce weight loss, and anti-cancer and anti-microbial properties.
It is a further object of this invention to provide a method of inducing weight loss in animals and humans by administering a pharmaceutical composition comprising a pharmaceutical diluent and compounds of formula V or formula VI.
It is a further object of the invention to provide a method of inhibiting fatty acid synthase activity in humans or animals by administering a pharmaceutical composition comprising a pharmaceutical diluent and compounds of formula V or formula VI.
It is a further object of this invention to provide a method of treating cancer in animals and humans by administering a pharmaceutical composition comprising a pharmaceutical diluent and a compound of formula V or formula VI.
It is still a further object of this invention to provide a method of preventing the growth of cancer cells in animals and humans by administering a pharmaceutical composition comprising a pharmaceutical diluent and a compound of formula V or formula VI.
It is a further object of this invention to provide a method of inhibiting growth of invasive microbial cells by administering a pharmaceutical composition comprising a pharmaceutical diluent and a compound of formula V or formula VI.
Brief Description of the Drawings
FIG. 1 shows a synthetic scheme to make certain compounds according to the
invention.
FIG. 2 shows a synthetic scheme to make certain compounds according to the invention.
Detailed Description of the Invention
The compounds of the invention can be prepared by conventional means. The synthesis of a number of the compounds is described in the examples. The compounds may be useful for the treatment of obesity, cancer, or microbially-based infections.
One embodiment of the invention is compounds having the following general
formula:
Figure imgf000012_0001
wherein:
R1 = H, or C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms; R2 = H, or Ci-C2O alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms; with the proviso that when R1 is -(CH2)5CH3 or -(CH2)I0CH3, R2 is not -CH2CH3, and the further proviso that when R1 is -CH2CH3, R2 is not -CH2CH3, and the further proviso that when R1 is -C6H5, R2 is not -CH2CH3; with the further proviso that when R1 is -CH(CH3)2, R2 is not -CH2CH3; with the further proviso that when R1 is -CH2CHBr2, R2 is not -CH2CH3; when R1 is - CH=CH2, R2 is not -CH2CH3; with the further proviso that when R1 is -CH3, R2 is not -CH2CH3; and the further proviso that when R1 is -^-C6H4F, R2 is not -CH2CH3; and the further proviso that when R1 is -(CHa)2CH3, R2 is not -CH2CH3; with the further proviso that when R1 is -CH3, R2 is not H; with the further proviso that when R1 is -(CH2)2CH3, R2 is not H; and with the further proviso that when R1 is -C6Hs, R2 is not H. with the further proviso that when R1 is -CH3, R2 is not -(CH2)2CH3.
In a preferred embodiment, R1 is -(CH2)7CH3, and R2 is -CH2CH3.
In another preferred embodiment, R1 is -(CH2)3CH3, and R2 is -CH2CH3.
A further embodiment of the invention is compounds having the following structural formula:
Figure imgf000013_0001
II
where R1 and R2 have the meanings given above, but where when R2 is -CH2CH3, R1 is not Q- C3 alkyl, optionally substituted with halogen atoms, and when R2 is -CH2CHs, R1 is not C2- alkenyl or C6-aryl, optionally substituted with halogen atoms, when R2 is H, R1 is not C1-C2 alkyl or C6-aryl, optionally substituted with halogen atoms. Another embodiment of the invention is compounds having the following general
formula:
Figure imgf000014_0001
πi
wherein:
R3 = H, or C]-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms;
R4 = H, or C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms;
R5 = H5 or Ci-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms, -C(O)R6, where R6 is H, Cj-C2O alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms, or -OH, -OR7 or -NR7, where R7 is H, Ci-C2O alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms; with the proviso that when R5 is H and R3 is -CH2CH3, R4 is not -CH2CH3; and the further proviso that when R3 is -CH3, and R5 is H, R4 is not -CH2CH3; and the further proviso that when R3 is -C6H5, and R5 is H, R4 is not -CH2CH3; with the proviso that when R5 is H and
R3 is -(CHs)2CH3, R4 is not -CH2CH3; with the further proviso that when R5 is H, and R3 is
Figure imgf000015_0001
R4 is not - CH3; and the further proviso that when R3 is -CH3, and R5 is H, R4 is not -CH3; with the further proviso that when R5 is H and R3 is -CH3, R4 is not -C(CH3)3 and the further proviso that when R5 is -CH3 and R3 is -C6Hs, R4 is not -CH3; and the further proviso that when R5 is -CH3 and R3 is -CH3, R4 is not -CH3; and the further proviso that when R5 is -CH3, and R3 is -CH2(CO)OCH2CH3, R4 is
not -CH2CH3.
Another embodiment of the invention is compounds having the following general formula:
Figure imgf000015_0002
where R3, R4, and R5 have the meanings given above, but with the proviso that when R4 is -CH2CH3, and R5 is H, R3 is not C1-C3 alkyl or C6-aryl, and with the further proviso that when R3 is -CH3, R4 is not Cj-C4 alkyl, and the further proviso that when R5 is -CH3, and R3 is -CH2(CO)OCH2CH3, R4 is not -CH2CH3; and the further proviso that when R4 and R5 are -CH3, and R3 is not -C6H5; and the further proviso that when R4 is -CH3, R3 is not =CHCH(CH3)2. Another embodiment of this invention is a pharmaceutical composition comprising a pharmaceutical diluent and a compound of formula V:
Figure imgf000016_0001
wherein:
R8 = H, or CJ-C2O alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms;
R9 = H, or C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms.
Optionally, the compounds of formulas I and II above may also be included in a pharmaceutical composition.
A further embodiment of the invention comprises a pharmaceutical diluent and a compound of formula VI:
Figure imgf000016_0002
Vl
wherein: R = H, or Ci-C2O alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms; Ru = H, or C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms;
R12 = H, or C)-C2O alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms, -C(O)R13, where R13 is H5 Ci-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms, or -OH, -OR14 or -NR14, where R14 is H, CJ-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms.
The compositions of the present invention can be presented for administration to humans and other animals in unit dosage forms, such as tablets, capsules, pills, powders, granules, sterile parenteral solutions or suspensions, oral solutions or suspensions, oil in water and water in oil emulsions containing suitable quantities of the compound, suppositories and in fluid suspensions or solutions. As used in this specification, the terms "pharmaceutical diluent" and "pharmaceutical carrier," have the same meaning. For oral administration, either solid or fluid unit dosage forms can be prepared. For preparing solid compositions such as tablets, the compound can be mixed with conventional ingredients such as talc, magnesium stearate, dicalcium phosphate, magnesium aluminum silicate, calcium sulfate, starch, lactose, acacia, methylcellulose and functionally similar materials as pharmaceutical diluents or carriers. Capsules are prepared by mixing the compound with an inert pharmaceutical diluent and filling the mixture into a hard gelatin capsule of appropriate size. Soft gelatin capsules are prepared by machine encapsulation of a slurry of the compound with an acceptable vegetable oil, light liquid petrolatum or other inert oil. Fluid unit dosage forms or oral administration such as syrups, elixirs, and suspensions can be prepared. The forms can be dissolved in an aqueous vehicle together with sugar, aromatic flavoring agents and preservatives to form a syrup. Suspensions can be prepared with an aqueous vehicle with the aid of a suspending agent such as acacia, tragacanth, methylcellulose and the like.
For parenteral administration fluid unit dosage forms can be prepared utilizing the compound and a sterile vehicle. In preparing solutions the compound can be dissolved in water for injection and filter sterilized before filling into a suitable vial or ampoule and sealing. Adjuvants such as a local anesthetic, preservative and buffering agents can be dissolved in the vehicle. The composition can be fro2;en after filling into a vial and the water removed under vacuum. The lyophilized powder can then be scaled in the vial and reconstituted prior to use.
The clinical therapeutic indications envisioned for the compounds of the invention include: (1) infections due to invasive micro-organisms such as staphylococci and enterococci; (2) cancers, and (3) obesity due to the ingestion of excess calories. Dose and duration of therapy will depend on a variety of factors, including (1) the patient's age, body weight, and organ function (e^g., liver and kidney function); (2) the nature and extent of the disease process to be treated, as well as any existing significant co-morbidity and concomitant medications being taken, and (3) drug-related parameters such as the route of administration, the frequency and duration of dosing necessary to effect a cure, and the therapeutic index of the drug. In general, doses will be chosen to achieve serum levels of 1 ng/ml to 100ng/ml with the goal of attaining effective concentrations at the target site of approximately 1 μg/ml to 10 μg/ml. EXAMPLES
The invention will be illustrated, but not limited, by the following examples: A series of compounds according to the invention were synthesized as described below. Biological activity of certain compounds were profiled as follows: Each compound was tested for at least some of the following properties: [1] inhibition of purified human FAS, [2] inhibition of fatty acid synthesis activity in whole cells and [3] cytotoxicity against cultured MCF-7 human breast cancer cells, known to possess high levels of FAS and fatty acid synthesis activity, using the crystal violet and XTT assays. Select compounds with low levels of cytotoxicity were then tested for weight loss in Balb/C mice. Certain compounds were also tested for activity against gram positive and/or negative bacteria. .
Chemical Synthesis of Compounds
Figure imgf000019_0001
1
To diethyloxaloacetate sodium salt (3.0 g, 14.3 mmol) in EtOH (10.3 mL) was added nonyl aldehyde (3.19 mL, 18.6 mmol) and the solution was heated to 50 0C for 24 h. The mixture was then cooled and poured into HCl (1 N, 20 mL) and extracted with Et2O (3 x 25 mL). The combined organics were dried (MgSO4), filtered and evaporated to give crude 1. Flash chromatography 20% EtOAc/Hex gave pure 1. Recrystallization in hexanes at -78 °C gave 1 as a
fluffy white solid (2.7 g, 73%); mp 38.5-30.5 0C 1H NMR (300 MHz, CDCl3) δ 0.86 (t, J= 7
Hz, 3 H), 1.25 (m, 12 H), 1.35 (t, J= 7 Hz, 3 H), 1.57 (m, 1 H), 2.03 (m, 1 H), 4.34 (m, 2 H), 5.05-5.07 (m, 1 H). 13C NMR (100 MHz, CDCl3) δ 14.0, 14.1, 22.6, 24.3, 29.1, 29.1 , 29.2, 31.8,
33.0, 61.9, 78.3, 118.7, 152.3, 164.6, 165.9. IR (NaCl) 1769, 1689, 1596 cm"1.
Figure imgf000020_0001
To diethyloxaloacete sodium salt (3.0 g, 14.3 mmol) in EtOH (10.3 mL) was added veraldehyde (1.98 mL, 18.6 mmol) and the solution was heated to 5O0C for 24 h. The mixture was then cooled and poured into HCl (1 N, 20 mL) and extracted with Et2O (3 x 25 mL). The combined organics were dried (MgSO4), filtered and evaporated to give crude 2. Flash chromatography 20% EtOAc/Hex gave pure 2. Recrystallization in hexanes at -78 0C gave 2 as a
fluffy white solid (1.9 g, 59%); mp 34- 35 0C. 1H NMR (300 MHz, CDCl3) δ 0.89 (t, J= 7 Hz, 3
H), 1.23- 1.37 (m, 4 H)1 1.35 (t, J= 7 Hz, 3 H), 1.56-1.62 (m, 1 H), 2.03-2.12 (m, 1 H), 4.27-4.44 (m, 2 H), 5.06-5.10 (dd, J= 2.9, 7.9 Hz, 1 H). 13C NMR (100 MHz, CDCl3) δ 13.7, 14.1, 22.2, 26.3, 32.7, 61.9, 78.2, 118.8, 152.1, 164.6, 165.8. IR (NaCl) 1779, 1686, 1655 cm"1.
Figure imgf000020_0002
3
To a precooled solution (-400C) of 1 (78 mg, 0.29 mmol) in DMF (1.2 mL) was added NaH (16.5 mg, 0.4 mmol) and this solution was allowed to warm to 00C and stir for 30 minutes. Then ethyl bromoacetate (66 uL, 0.59 mmol) was added and the mixture was allowed to warm and stir at room temperature (10-12 h). NH4Cl (saturated solution, 20 mL) was added and the mixture was extracted with Et2O (3 x 15 mL). The combined organics were dried (MgSO4), filtered and evaporated to give crude 3. Flash chromatography using 2%AcOH/20%EtOAc/Hex gave pure 3 (65 mg, 63%). 1H NMR (300 MHz, CDCl3) δ 0.84 (t, J
= 7.1 Hz, 3 H), 1.22-1.34 (m, 18 H), 1.56-1.61 (m, 1 H), 2.03-2.06 (m, 1 H), 4.19-4.28 (m, 4 H),
5.08 (dd, J= 2.7, 7.8 Hz, 1 H), 5.12 (d, J= 6.6 Hz, 2 H)1 13C NMR (100 MHz, CDCl3) δ 14.0,
14.1, 22.5, 24.2, 29.1, 29.1, 29.1, 29.2, 31.7, 33.1, 61.2, 61.5, 66.5, 78.8, 124.2, 146.4, 161.1, 166.5, 168.2.
Figure imgf000021_0001
To 1 (100 mg, 0.38 mmol) and dimethyl sulfate (79 uL, 0.84 mmol) following the above procedure gave 4 (32 mg, 30%) after flash chromatography 10% EtOAc/Hex. 1H NMR
(300 MHz, CDCl3) δ 0.85 (t, J= 6.9 Hz, 3 H), 1.24 (m, 12 H), 1.32 (t, J= 7.1 Hz, 3 H), 1.50- 1.62 (m, 1 H), 1.99-2.06 (m, 1 H), 4.21 (s, 3 H), 4.28-4.33 (m, 2 H), 5.05 (dd, J= 2.9, 7.8 Hz, 1
H). 13C NMR (300 MHz, CDCl3) δ 14.0, 14.1, 22.6, 24.2, 29.1, 29.1, 29.2, 31.8, 33.2, 59.8, 61.3, 78.5, 123.4, 148.0, 161.4, 166.7.
Figure imgf000021_0002
5
To 1 (75 mg, 0.28 mmol) and benzyl bromide (68 uL, 0.58 mmol) following the above procedure gave 5 (36 mg, 36%) after flash chromatography 10% EtOAc/Hexanes. 1H
NMR (300 MHz, CDCl3) δ 0.84 (t, J= 6.8 Hz, 3 H), 1.25 (m, 12 H), 1.32 (t, J= 7.1 Hz, 3 H),
1.53-1.70 (m, 1 H), 1.99-2.05 (m, 1 H), 4.22-4.33 (m, 2 H), 5.05 (dd, J= 2.9, 7.8 Hz, 1 H), 5.6
(s, 2 H), 7.28-7.42 (m, 5 H). 13C NMR (100 MHz, CDCl3) δ 14.1, 14.1, 22.6, 23.9, 29.1, 29.1,
29.3, 31.8, 32.9, 61.2, 72.9, 78.6, 126.0, 127.8, 128.4, 128.5, 135.8, 147.4, 161.4, 166.9.
Figure imgf000022_0001
6
To 1 (132 mg, 0.5 mmol) and allyl bromide (82 uL, 0.95 mmol) following the above procedure gave 6 (33 mg, 22%) after flash chromatography 10% EtOAc/Hex. 1H NMR
(300 MHz, CDCl3) δ 0.86 (t, J= 6.9 Hz, 3 H), 1.24 (m, 12 H), 1.32 (t, J= 7.1 Hz, 3 H), 1.51-
1.61 (m, 1 H), 1.99-2.08 (m, 1 H), 4.22-4.34 (m, 2 H), 5.05-5.09 (m, 3 H), 5.27 (dq, J= 9.2, 1.5 Hz, 1 H), 5.43 (dq, J= 17.1, 1.5 Hz, 1 H)5 5.89-6.02 (m, 1 H).
Figure imgf000022_0002
7
To 1 (100 mg, 0.35 mmol) in pyridine (1.4 mL) was added ethyl chloroformate (50 uL, 0.53 mmol) and the solution was allowed to stir at room temperature. The mixture was then poured into CuSO4 (10 mL) and extracted with CH2Cl2 (3 x 10 mL). The combined organics were dried (MgSO4) filtered and evaporated to give crude 7. Column chromatography 20%
EtOAc/Hexanes gave pure 7 (39 mg, 31 %). 1H NMR (400 MHz, CDCl3) δ 0.83 ( t, J= 6.0 Hz, 3 H), 1.98-1.39 (m, 18 H), 1.62-1.71 (m, 1 H), 2.08-2.11 (m, 1 H), 4.23-4.33 (m, 4 H), 5.17-5.19
(m, 1 H); 13C NMR (100 MHz, CDCl3) δ 12.9, 12.9, 13.0, 21.6, 23.1, 28.0, 28.1, 28.2, 30.2, 31.4,
61.1, 65.3, 78.0, 136.3, 140.0, 149.1, 158.7, 164.4.
BIOLOGICAL AND BIOCHEMICAL METHODS
Purification of FAS from ZR-75-1 Human Breast Cancer Cells.
Human FAS was purified from cultured ZR-75-1 human breast cancer cells )btained from the American Type Culture Collection. The procedure, adapted from Linn et al, 1981, and Kuhajda et al, 1994, utilizes hypotonic lysis, successive polyethyleneglycol (PEG) precipitations, and anion exchange chromatography. ZR-75-1 cells are cultured at 37 0C with 5% CO2 in RPMI culture medium with 10% fetal bovine serum, penicillin and streptomycin.
Ten Tl 50 flasks of confluent cells are lysed with 1.5 ml lysis buffer (20 mM Tris- HCl, pH 7.5, 1 mM EDTA5 0.1 mM phenylmethanesulfonyl fluoride (PMSF), 0.1% Igepal CA- 630) and dounce homogenized on ice for 20 strokes. The lysate is centrifuged in JA-20 rotor (Beckman) at 20,000 rpm for 30 minutes at 4 0C and the supernatant is brought to 42 ml with lysis buffer. A solution of 50% PEG 8000 in lysis buffer is added slowly to the supernatant to a final concentration of 7.5%. After rocking for 60 minutes at 4 0C, the solution is centrifuged in JA-20 rotor (Beckman) at 15,000 rpm for 30 minutes at 4 0C. Solid PEG 8000 is then added to the supernatant to a final concentration of 15%. After the rocking and centrifugation is repeated as above, the pellet is resuspended overnight at 4 0C in 10 ml of Buffer A (20 mM K2HPO4, pH 7.4). After 0.45 μM filtration, the protein solution is applied to a Mono Q 5/5 anion exchange column (Pharmacia). The column is washed for 15 minutes with buffer A at 1 ml/minute, and bound material is eluted with a linear 60-ml gradient over 60 minutes to 1 M KCl. FAS (MW~ 270 kD) typically elutes at 0.25 M KCl in three 0.5 ml fractions identified using 4-15% SDS- PAGE with Coomassie G250 stain (Bio-Rad). FAS protein concentration is determined using the Coomassie Plus Protein Assay Reagent (Pierce) according to manufacturer's specifications using BSA as a standard. This procedure results in substantially pure preparations of FAS (>95%) as judged by Coomassie-stained gels.
Measurement of FAS Enzymatic Activity and Determination ofthe JCso of the Compounds
FAS activity is measured by monitoring the malonyl-CoA dependent oxidation of NADPH spectrophotometrically at OD340 in 96-well plates (DiIs et al and Arslanian et al, 1975). Each well contains 2 μg purified FAS, 100 mM K2HPO4, pH 6.5, 1 mM dithiothreitol (Sigma), and 187.5 μM /3-NADPH (Sigma). Stock solutions of inhibitors are prepared in DMSO at 2, 1, and 0.5 mg/ml resulting in final concentrations of 20, 10, and 5 μg/ml when 1 μl of stock is added per well. For each experiment, cerulenin (Sigma) is run as a positive control along with DMSO controls, inhibitors, and blanks (no FAS enzyme) all in duplicate.
The assay is performed on a Molecular Devices SpectraMax Plus
Spectrophotometer. The plate containing FAS, buffers, inhibitors, and controls are placed in the spectrophotometer heated to 370C. Using the kinetic protocol, the wells are blanked on duplicate wells containing 100 μl of 100 mM K2HPO4, pH 6.5 and the plate is read at OD340 at 10 sec intervals for 5 minutes to measure any malonyl-CoA independent oxidation of NADPH. The plate is removed from the spectrophotometer and malonyl-CoA (67.4 μM, final concentration per well) and alkynyl-CoA (61.8 μM, final concentration per well) are added to each well except to the blanks. The plate is read again as above with the kinetic protocol to measure the malonyl- CoA dependent NADPH oxidation. The difference between the Δ OD340 for the malonyl-CoA dependent and non-malonyl-CoA dependent NADPH oxidation is the specific FAS activity. Because of the purity of the FAS preparation, non-malonyl-CoA dependent NADPH oxidation is negligible.
The IC50 for the compounds against FAS is determined by plotting the Δ OD340 for each inhibitor concentration tested, performing linear regression and computing the best-fit line, r2 values, and 95% confidence intervals. The concentration of compound yielding 50% inhibition of FAS is the IC50. Graphs of Δ OD340 versus time are plotted by the SOFTmax PRO software (Molecular Devices) for each compound concentration. Computation of linear regression, best-fit line, r2, and 95% confidence intervals are calculated using Prism Version 3.0 (Graph Pad Software).
Crystal Violet Cell Growth Assay
The crystal violet assay measures cell growth but not cytotoxicity. This assay employs crystal violet staining of fixed cells in 96-well plates with subsequent solubilization and measurement of OD490 on a spectrophotometer. The OD490 corresponds to cell growth per unit time measured. Cells are treated with the compounds of interest or vehicle controls and IC50 for each compound is computed.
To measure the cytotoxicity of specific compounds against cancer cells, 5 x 104 MCF-7 human breast cancer cells, obtained from the American Type Culture Collection are plated per well in 24 well plates in DMEM medium with 10% fetal bovine serum, penicillin, and streptomycin. Following overnight culture at 370C and 5% CO2, the compounds to be tested, dissolved in DMSO, are added to the wells in 1 μl volume at the following concentrations: 50, 40, 30, 20, and 10 μg/ml in triplicate. Additional concentrations are tested if required. 1 μl of DMSO is added to triplicate wells are the vehicle control. C75 is run at 10, and 5 μg/ml in triplicate as positive controls.
After 72 hours of incubation, cells are stained with 0.5 ml of Crystal Violet stain (0.5% in 25% methanol) in each well. After 10 minutes, wells are rinsed, air dried, and then solubilized with 0.5 ml 10% sodium dodecylsulfate with shaking for 2 hours. Following transfer of 100 μl from each well to a 96-well plate, plates are read at OD4P0 on a Molecular Devices SpectraMax Plus Spectrophotometer Average OD490 values are computed using SOFTmax Pro Software (Molecular Devices) and IC50 values are determined by linear regression analysis using Prism version 3.02 (Graph Pad Software, San Diego).
XTT Cytotoxicity Assay
The XTT assay is a non-radioactive alternative for the [51Cr] release cytotoxicity assay. XTT is a tetrazolium salt that is reduced to a formazan dye only by metabolically active, viable cells. The reduction of XTT is measured spectrophotometrically as OD490 - OD650.
To measure the cytotoxicity of specific compounds against cancer cells, 9 x 103 MCF-7 human breast cancer cells, obtained from the American Type Culture Collection are plated per well in 96 well plates in DMEM medium with 10% fetal bovine serum, insulin, penicillin, and streptomycin. Following overnight culture at 370C and 5% CO2, the compounds to be tested, dissolved in DMSO, are added to the wells in 1 μl volume at the following concentrations: 80, 40, 20, 10, 5, 2.5, 1.25, and 0.625 μg/ml in triplicate. Additional concentrations are tested if required. 1 μl of DMSO is added to triplicate wells are the vehicle control. C75 is run at 40, 20, 10, 15, 12.5, 10, and 5 μg/ml in triplicate as positive controls.
After 72 hours of incubation, cells are incubated for 4 hours with the XTT reagent as per manufacturer's instructions (Cell Proliferation Kit II (XTT) Roche). Plates are read at OD490 and OD65o on a Molecular Devices SpectraMax Plus Spectrophotometer. Three wells containing the XTT reagent without cells serve as the plate blank. XTT data are reported as OD490 - OD65o. Averages and standard error of the mean are computed using SOFTmax Pro software (Molecular Dynamics). The IC50 for the compounds is defined as the concentration of drug leading to a 50% reduction in OD49C) - OD65O compared to controls. The OD490 - OD6So are computed by the SOFTmax PRO software (Molecular Devices) for each compound concentration. IC50 is calculated by linear regression, plotting the FAS activity as percent of control versus drug concentrations. Linear regression, best-fit line, r2, and 95% confidence intervals are determined using Prism Version 3.0 (Graph Pad Software).
Measurement of [14C] acetate Incorporation into Total Lipids and Determination of IC so of Compounds
This assay measures the incorporation of [14C]acetate into total lipids and is a measure of fatty acid synthesis pathway activity in vitro. It is utilized to measure inhibition of fatty acid synthesis in vitro.
MCF-7 human breast cancer cells cultured as above, are plated at 5 x 104 cells per well in 24-well plates. Following overnight incubation, the compounds to be tested, solubilized in DMSO, are added at 5, 10, and 20 μg/ml in triplicate, with lower concentrations tested if necessary. DMSO is added to triplicate wells for a vehicle control. C75 is run at 5 and 10 μg/ml in triplicate as positive controls. After 4 hours of incubation, 0.25 μCi of [14C] acetate (10 μl volume) is added to each well.
After 2 hours of additional incubation, medium is aspirated from the wells and 800 μl of chlorofornrmethanol (2:1) and 700 μl of 4 mM MgCl2 is added to each well. Contents of each well are transferred to 1.5 Eppendorf tubes, and spun at full-speed for 2 minutes in a high-speed Eppendorf Microcentrifuge 5415D. After removal of the aqueous (upper) layer, an additional 700 μl of chloroform .-methanol (2:1) and 500 μl of 4 mM MgCl2 are added to each tube and then centrifuged for 1 minutes as above. The aqueous layer is removed with a Pasteur pipette and discarded. An additional 400 μl of chlorofornrmethanol (2:1) and 200 μl of 4 mM MgCl2 are added to each tube, then centrifuged and aqueous layer is discarded. The lower (organic) phase is transferred into a scintillation vial and dried at 40 0C under N2 gas. Once dried, 3 ml of scintillant (APB #NBC5104) is added and vials are counted for 14C. The Beckman Scintillation counter calculates the average cpm values for triplicates.
The IC50 for the compounds is defined as the concentration of drug leading to a 50% reduction in [I4C]acetate incorporation into lipids compared to controls. This is determined by plotting the average cpm for each inhibitor concentration tested, performing linear regression and computing the best-fit line, r2 values, and 95% confidence intervals. The average cpm values are computed by the Beckman scintillation counter (Model LS6500) for each compound concentration. Computation of linear regression, best-fit line, r2, and 95% confidence intervals are calculated using Prism Version 3.0 (Graph Pad Software).
Weight Loss Screen
Balb/C mice (Jackson Labs) are utilized for the initial weight loss screening. Animals are housed in temperature and 12 hour day/night cycle rooms and fed mouse chow and water ad lib. Three mice are utilized for each compound tested with vehicle controls in triplicate per experiment. For the experiments, mice are housed separately for each compound tested three mice to a cage. Compounds are diluted in DMSO at 10 mg/ml and mice are injected intraperitoneally with 60 mg/kg in approximately 100 μl of DMSO or with vehicle alone. Mice are observed and weighed daily; average weights and standard errors are computed with Excel (Microsoft). The experiment continues until treated animals reach their pretreatment weights. Select compounds are tested in animals housed in metabolic cages. Dosing of animals are identical to the screening experiments with three animals to a single metabolic cage. Animal weights, water and food consumption, and urine and feces production are measured
daily.
Antimicrobial Properties
A broth microdilution assay is used to assess the antimicrobial activity of the compounds. Compounds are tested at twofold serial dilutions, and the concentration that inhibits visible growth (OD6Oo at 10% of control) is defined as the MIC. Microorganisms tested include Staphylococcus aureus (ATCC # 29213), Enterococcus faecalis (ATCC # 29212), Pseudomonas aeruginosa (ATCC # 27853), and Escherichia coli (ATCC # 25922). The assay is performed in two growth media, Mueller Hinton Broth and Trypticase Soy Broth.
A blood (Tsoy/5% sheep blood) agar plate is inoculated from frozen stocks maintained in T soy broth containing 10% glycerol and incubated overnight at 37° C. Colonies are suspended in sterile broth so that the turbidity matches the turbidity of a 0.5 McFarland standard. The inoculum is diluted 1 :10 in sterile broth (Mueller Hinton or Trypticase soy) and 195 ul is dispensed per well of a 96- well plate. The compounds to be tested, dissolved in DMSO, are added to the wells in 5 ul volume at the following concentrations: 25, 12.5, 6.25, 3.125, 1.56 and 0.78 ug/ml in duplicate. Additional concentrations are tested if required. 5 ul of DMSO added to duplicate wells are the vehicle control. Serial dilutions of positive control compounds, vancomycin (E. faecalis and S. aureus) and tobramycin (E. coli and P. aeruginosa), are included
in each run. After 24 hours of incubation at 37 0C, plates are read at OD600 on a Molecular Devices SpectraMax Plus Spectrophotometer. Average OD60O values are computed using SOFTmax Pro Software (Molecular Devices) and MIC values are determined by linear regression analysis using Prism version 3.02 (Graph Pad Software, San Diego). The MIC is defined as the concentration of compound required to produce an OD60O reading equivalent to 10% of the vehicle control reading.
β-oxidation Assay- Isolation of Acid Soluble Products
A 24 well plate with 1 ml per well was prepared with 2.5 x 105 cells /well. The cells were incubated O/N.
The next day, a solubilized palmitate solution was prepared. 50μls Of(I-14C) Palmitic acid was added to a 2 ml centrifuge tube and dried under nitrogen gas. 2 mis of α-CD (α-Cyclodextran)- 10 mg/ml in 10 mM Tris were added. This solution was incubated in a 370C water bath for 30 minutes.
A hot mix was prepared by adding 25 μl of this solution to 2.5 μls of 200μM Carnitine and 222.5 μl of serum free medium that is used for cells.
The cells were then treated with the test compound in triplicate, and incubated at 37°C for 60 minutes. The medium was removed and 250 μl of the hot mix were added. The test compound was added again, and further incubated at 37°C for 60 minutes. The reaction was stopped with 50 μl of 2.6 N HClO4. The contents of the plate were transferred to a 1.5 ml centrifuge tube, and 50 μl of 4N KOH were then added, and the tube incubated in a 6O0C water bath for 30 min. Sodium acetate (IM, 75 μl) and sulfuric acid (3N, 50 μl) were added to the solution and vortexed. The tube was spun for 7 minutes at 1000 rpm at room temperature. A portion (225 μl) was removed, and the following were added (vortex after each addition, twice at the end): 938 μl of 1 :1 Chloroform: Methanol; 468 μl of Chloroform; 281 μl distilled Water. The tubes were spun at 1000 rpm for 5 minutes. The upper phase was removed into a large glass scintillation vial and 5 ml of Budget solvent (scintillation liquid) was added. The tubes were well vortexed. Finally, 14C was counted for one minute.
Results of the biological testing
Figure imgf000031_0001
Figure imgf000031_0002
Figure imgf000031_0003
Figure imgf000032_0001
Figure imgf000032_0002
Figure imgf000032_0003
Figure imgf000032_0004
Beta Oxidation Results
0.04 ug/ml 0.19 ug/ml .78 ug/ml 3.13 ug/ml 12.5ug/ml 50 ug/ml
Compound 1 94 108 104 113 114 54
103 104 104 109 107 49
120 113 106 107 100 45
0.19 ug/ml 0.39 ug/ml I .78 ug/ml 1.56 ug/ml 12.5ug/ml 50 ug/ml
Compound 2 102 102 107 112 108 45 I
1.25 ug/ml 2.5 ug/ml 5 ug/ml 10 ug/ml
I Compound 3 I 113 I 131 I 138 I 79
1.25 ug/ml 2.5 ug/ml 5 ug/ml 10 ug/ml |
I Compound 4 ! 112 I 119 I 116 104 I
1.25 ug/ml 2.5 ug/ml 5 ug/ml 10 ug/ml |
I Compound 5 108 101 115 74 I
I 1.25 ug/ml 2.5 ug/ml 5 ug/ml I 10 ug/ml (
Compound 6 118 125 112 101 I
0.09 ug/ml 0.39 ug/ml 1.56 ug/ml | 6.25 ug/ml 25 ug/ml 100 ug/ml
I Compound 7 | ! 99 I 107 I I 113 I 146 I 131 I I 54 I

Claims

We claim:
1. A compound of formula I:
Figure imgf000034_0001
wherein:
R1 = H, or CpC20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms; R2 = H, or Ci-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms; with the proviso that when R1 is -(CH2)5CH3 or -(CH2)ioCH3, R2 is not -CH2CH3, and the further proviso that when R1 is -CH2CH3, R2 is not -CH2CH3, and the further proviso that when R1 is -C6H5, R2 is not -CH2CH3; with the further proviso that when R1 is -CH(CH3)2, R2 is not -CH2CH3; with the further proviso that when R1 is -CH2CHBr2, R2 is not -CH2CH3; when R1 is - CH=CH2, R2 is not -CH2CH3; with the further proviso that when R1 is -CH3, R2 is not -CH2CH3; and the further proviso that when R1 is -JD-C6H4F, R2 is not -CH2CH3; and the further proviso that when R1 is -(CH2)2CH3, R2 is not -CH2CH3; with the further proviso that when R1 is -CH3, R2 is not H; with the further proviso that when R1 is -(CH2)2CH3, R2 is not H; and with the further proviso that when R1 is -C6H5, R2 is
not H. with the further proviso that when R1 is -CH3, R2 is not -(CH2)2CH3.
2. A compound according to claim 1, wherein R1 is -(CH2)7CH3, and R2 is -CH2CH3.
3. A compound according to claim 1, wherein R1 is -(CH2)SCH3, and R2 is -CH2CH3.
4. A compound of formula III:
Figure imgf000035_0001
wherein:
R3 = H, or C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms;
R4 = H, or C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms;
R5 = H, or C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms, -C(O)R6, where R6 is H, C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms, or -OH, -OR7 or -NR7, where R7 is H, C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms; with the proviso that when R5 is H and R3 is -CH2CH3, R4 is not -CH2CH3; and the further proviso that when R3 is -CH3, and R5 is H, R4 is not -CH2CH3; and the further proviso that when R3 is -C6H5, and R5 is H, R4 is not -CH2CH3; with the proviso that when R5 is H and
R3 is -(CH2)2CH3, R4 is not -CH2CH3; with the further proviso that when R5 is H, and R3 is =CHCH(CH3)2, R4 is not - CH3; and the further proviso that when R3 is -CH3, and R5 is H, R4 is not -CH3; with the further proviso that when R5 is H and R3 is -CH3, R4 is not -C(CH3)3 and the further proviso that when R5 is -CH3 and R3 is -C6Hs, R4 is not -CH3; and the further proviso that when R5 is -CH3 and R3 is -CH3, R4 is not -CH3; and the further proviso that when R5 is -CH3, and R3 is -CH2(CO)OCH2CH3, R4 is not -CH2CH3.
5. A compound according to claim 4, wherein R3 is -(CH2)7CH3, R4 is -CH2CH3, and R5 is -CH2C(O)OCH2CH3.
6. A compound according to claim 4, wherein R3 is -(CH2^CH3, R4 is -CH2CH3, and R5 is -CH3.
7. A compound according to claim 4, wherein R3 is -(CH2)7CH3, R4 is -CH2CH3, and R5 is -CH2(C6H4).
8. A compound according to claim 4, wherein R3 is -(CH2)7CH3, R4 is -CH2CH3, and R5 is -CH2-CH=CH2.
9. A compound according to claim 4, wherein R3 is -(CH2)7CH3, R4 is -CH2CH3, and R5 is -C(O)OCH2CH3.
10. A pharmaceutical composition comprising a pharmaceutical diluent and a compound of formula V:
Figure imgf000036_0001
wherein:
R8 = H, or C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms;
R9 = H, or C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms.
11. a pharmaceutical diluent and a compound of formula VI:
Figure imgf000037_0001
VI
wherein:
R10 = H, or C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms;
R11 = H, or C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms;
R12 = H, or C1-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms, -C(O)R13, where R13 is H, C]-C20 alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms, or -OH, -OR14 or -NR14, where R14 is H, Cj-C2O alkyl, cycloalkyl, alkenyl, aryl, arylalkyl, or alkylaryl, optionally containing halogen atoms.
12. A method oi inducing weight loss in an animal or human subject comprising administering an effective amount of a pharmaceutical composition according to claim 10 to said subject.
13. The method of claim 12, wherein the subject is a human.
14. The method of claim 12, wherein the subject is an animal.
15. A method of inducing weight loss in an animal or human subject comprising administering an effective amount of a pharmaceutical composition according to claim 11 to said subject.
16. The method of claim 12, wherein the subject is a human.
17. The method of claim 12, wherein the subject is an animal.
18. A method of treating cancer in an animal or human subject, comprising administering an effective amount of a pharmaceutical composition according to claim 10 to said subject.
19. The method of claim 18, wherein the subject is a human.
20. The method of claim 18, wherein the subject is an animal.
21. A method of treating cancer in an animal or human subject, comprising administering an effective amount of a pharmaceutical composition according to claim 11 to said subject.
22. The method of claim 21 , wherein the subject is a human.
23. The method of claim 21, wherein the subject is an animal.
24. A method of inhibiting fatty acid synthase activity in an animal or human subject comprising administering an effective amount of a pharmaceutical composition according to claim 3 to said subject.
25. A method of inhibiting growth of invasive microbial cells in an animal or human subject comprising the administration of an effective amount of a pharmaceutical composition according to claim 10 to said subject.
26. The method of claim 25, wherein the subject is a human.
27. The method of claim 25, wherein the subject is an animal.
28. A method of inhibiting growth of invasive microbial cells in an animal or human subject comprising the administration of an effective amount of a pharmaceutical composition according to claim 11 to said subject.
29. The method of claim 28, wherein the subject is a human.
30. The method of claim 28, wherein the subject is an animal.
PCT/US2006/028979 2005-07-26 2006-07-26 Novel compounds, pharmaceutical compositions containing same, and methods of use for same WO2007014247A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/309,419 US20100029761A1 (en) 2005-07-26 2006-07-26 Novel compounds, pharmaceutical compositions containing same, and methods of use for same

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US70231605P 2005-07-26 2005-07-26
US60/702,316 2005-07-26

Publications (2)

Publication Number Publication Date
WO2007014247A2 true WO2007014247A2 (en) 2007-02-01
WO2007014247A3 WO2007014247A3 (en) 2007-11-29

Family

ID=37683930

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/028979 WO2007014247A2 (en) 2005-07-26 2006-07-26 Novel compounds, pharmaceutical compositions containing same, and methods of use for same

Country Status (2)

Country Link
US (1) US20100029761A1 (en)
WO (1) WO2007014247A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8729239B2 (en) 2009-04-09 2014-05-20 Nuclea Biotechnologies, Inc. Antibodies against fatty acid synthase
US11202795B2 (en) 2014-11-20 2021-12-21 Vib Vzw Means and methods for treatment of early-onset Parkinson's disease

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR898946A (en) * 1945-08-07 1945-05-11 Process for the preparation of halogenated derivatives of ketobutyrolactone-carboxylic esters
JP2003163031A (en) * 2001-09-12 2003-06-06 Daicel Chem Ind Ltd ELECTROLYTE, NONAQUEOUS ELECTROCHEMICAL EQUIPMENT AND alpha-SUBSTITUTIONAL OXY-gamma-BUTYROLACTONE DERIVATIVE

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR898946A (en) * 1945-08-07 1945-05-11 Process for the preparation of halogenated derivatives of ketobutyrolactone-carboxylic esters
JP2003163031A (en) * 2001-09-12 2003-06-06 Daicel Chem Ind Ltd ELECTROLYTE, NONAQUEOUS ELECTROCHEMICAL EQUIPMENT AND alpha-SUBSTITUTIONAL OXY-gamma-BUTYROLACTONE DERIVATIVE

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LI ET AL.: 'reactions of arylaldehydes and N-sulfonated imines with Dimethyl acetylenedicarboxylate catalyzed by nitrogen and phosphine lewis bases' ORGANIC LETTERS vol. 5, 2003, pages 4273 - 4276 *
WASSERMAN ET AL.: 'Pyrolysis of aryl glycolic acids. III. The structure of langenbeck's compound' JOURNAL OF THE AMERICAN CHEMICAL SOCIETY vol. 77, 1955, pages 973 - 939 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8729239B2 (en) 2009-04-09 2014-05-20 Nuclea Biotechnologies, Inc. Antibodies against fatty acid synthase
US9732158B2 (en) 2009-04-09 2017-08-15 Nmdx, Llc Antibodies against fatty acid synthase
US11202795B2 (en) 2014-11-20 2021-12-21 Vib Vzw Means and methods for treatment of early-onset Parkinson's disease

Also Published As

Publication number Publication date
WO2007014247A3 (en) 2007-11-29
US20100029761A1 (en) 2010-02-04

Similar Documents

Publication Publication Date Title
CA2767092C (en) Novel compounds, pharmaceutical compositions containing same and methods of use for same
US20090005435A1 (en) Novel Compounds, Pharmaceutical Compositions Containing Same, and Methods of Use for Same
AU2003248810B2 (en) Novel compounds, pharmaceutical compositions containing same, and methods of use for same
US20110288052A1 (en) Novel compounds, pharmaceutical compositions containing same, and methods of use for same
US20100168176A1 (en) Novel compounds, pharmaceutical compositions containing same, and methods of use for same
US20100029752A1 (en) Novel compounds, pharmaceutical compositions containing same, and methods of use for same
WO2007014247A2 (en) Novel compounds, pharmaceutical compositions containing same, and methods of use for same

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 06788520

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 12309419

Country of ref document: US