CA2115994A1 - Protein kinase c inhibition and novel compound balanol - Google Patents

Protein kinase c inhibition and novel compound balanol

Info

Publication number
CA2115994A1
CA2115994A1 CA002115994A CA2115994A CA2115994A1 CA 2115994 A1 CA2115994 A1 CA 2115994A1 CA 002115994 A CA002115994 A CA 002115994A CA 2115994 A CA2115994 A CA 2115994A CA 2115994 A1 CA2115994 A1 CA 2115994A1
Authority
CA
Canada
Prior art keywords
protein kinase
compound
approximately
pharmaceutically acceptable
composition
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
CA002115994A
Other languages
French (fr)
Inventor
Yali F. Hallock
William P. Janzen
Lawrence M. Ballas
Palaniappan Kulanthivel
Christie Boros
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Eli Lilly and Co
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2115994A1 publication Critical patent/CA2115994A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D223/00Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom
    • C07D223/02Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom not condensed with other rings
    • C07D223/06Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom not condensed with other rings 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
    • C07D223/12Nitrogen atoms not forming part of a nitro radical
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Landscapes

  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

2115994 9303730 PCTABS00020 Balanol, which is derived from fungi of the genus Verticillium, especially the fungus Verticillium balanoides, is provided. Balanol, when substantially pure, is soluble in dimethyl sulfoxide, water, and methanol; insoluble in ethyl acetate and chloroform; gives a positive color reaction with polymolybdic acid, ninhydrin reagent, and ferric chloride; is negative with Dragendorff's reagent and Iodoplatinate spray; has an Rf value of approximately 0.58 with silica gel thin layer chromatography with n-butanol/acetic acid/water at a ratio of 4:1:1 respectively; and has a molecular weight of approximately 550. Compounds of the invention are useful for inhibiting protein kinase C and treating conditions related to, or affected by inhibition of protein kinase C, particularly cancer tumors, inflammatory disease, reperfusion injury, and cardiac dysfunctions related to reperfusion injury.

Description

W093/~3730 PCT/US92/07124
2 1 1 5 9 9 ~

PROTBIN ~INA~ C IN~IBITION
AND NOVEL CONPOUND B~LaNOL
FIELD OF THg INVENTTON
The present învention relates to diagnosis and S treatment for inflammatory, cardiovascular and neoplastic diseases. More particularly, the present invention relates to novel compounds derived from fungi of the genus Verticillium, especially the fungus Verticlllium balanoides (DreschlerJ
Do~wsett et al., useful for inhibiting activity of the enzyme 10 protein kinase C. A preferred composition has been normal Balanol.

BACXGRO'U~D OF THE INVE~TION
Protein kinase C is a family of calcium stimulant and phospholipid-dependent ssrine/threonine-specific protein lS kinases which play an important role in cellular growth control, regulation, and differentiation. Protein kinase C is also fundamental to the processes involved in tumorigenicity, since it is the major high-affinity receptor for several classes tumor promoters as well as for endogenous cellular 20 diacylglycerols. These tumor promoters also stimulate protein kinase C catalysis. Direct activation of protein kinase C by tumor promoting phorbol esters has been reported. See Castagna et al. J. Biol. ~h~m., 257:7847 (1982). The mechanisms of protein kinase C action have been described in U.S. Patent 25 4,816,450 issued March 28, 1989 to Bell et al., the disclosure of which is specifically incorporated as fully set forth herein. Protein kinase C is activated by diacylglycerol (DAG), a neutral lipid, and when activated will transfer the y-211599~ - 2 -phosphate of MgATP to a serine or threonine residue on a substrate protein. ~
Since the activation of protein kinase C has been implicated in several human disease processes, including cancer 5 tumors, inflammation, and reperfusion injury, inhibition of protein kinase C should be of great therapeutic value in treating these conditions. Certain protein kinase C inhibitors have been reported to potentiate the antitumor activity of cis-platin both in vitro and in vivo, Grunicke et al., Adv. Enzyme 10 Regul. 28: 201 (1989); and German Offenlegungsschrift DE
3827974. In addition, it has been suggested that protein kinase C would be a potential target for therapeutic design because of its central role in cell growth. See Tritton, T.R. and Hickman, J.A. Cancer Cells 2: 95-102 (1990).
Certain protein kinase C inhibitors have been demonstrated to block platelet aggregation and release of neutrophil activating agents such as platelet activating factor, PAF. See Schachtele et al., Biochem. Biophy. Res.
Commun. 151:542 (1988); Hannun et al., J. Biol. Chem. 262:13620 (1987); and Yamada et al., Biochqm. Pharmacol. 37: 1161 (1988).
Protein kinase C inhibitors have also been shown to inhibit neutrophil activation, and chemotactic migration. See McIntyre et al., J. Biol C~em. 262:15730 (1987); Lambreth et al., J.
Biol. Chem. 263:3818 (1988); Pittet et al., J. Biol. Chem.
25 262:10072 ~1987); and Gaudry et al., Immunology 63:71~ (1988).
Neutrophil degranulation and release of proteolytic enzymes and reactive oxygen intermediates has also been shown. See Wilson et al., J. Biol. Chem. 261:12616 (1986); Fujita et al., Biochem. P~larmac~l. 35:4555 (1986); Berkow et al., J. Leukoc., 30 Biol. 41:441 (1987); Salzer et al., Bioch~m. Biophys. Res.
Comm~n. 148:747 (1987); Kramer et al., J. Biol. Chem. 262:5876 (1988); and Dewald et al., Biochem. J. 264:879 (1989).
It is apparent that inhibitors of protein kinase C
have the potential for blocking all three of the most 35 significant mechanisms of pathogenesis associated with myocardial reperfusion injury, and should thus have a decided wo s3/0373n PCr/USg2/07124 211599~

therapeutic advantage. Additionally, the inhibitory effect of protein kinase C inhibitors on keratinocytes, and on the oxidative burst in neutrophils will lead to an anti-inflammatory effect. Inflammation and reperfusion injury, 5 particularly pertaining to cardiac injury, are common conditions for which there exists no definitive treatment despite extensive research, and appropriate treatments for these conditions are needed.
German Offenlegungsschrift DE 3827974 Al discloses lo preparations comprising a protein kinase C inhibitor such as in combination with a lipid, a lipid analog, a cytostatic agent or phospholipase inhibitor useful for cancer therapy.
Natural products that inhibit protein kinase C have been reported. See Tamaoki et al., Bio/Technology 8:732 (1988). For example, saturosporine, Nakano et al., J.
Antibiotics 40:706-708 (1987), an alkaloid metabolite, and its 7-hydroxy congener, Takahashi et al., J. Antibiotics~ 42:571-576 (1989), isolated from Streptomyces st~urosporeus, have shown protein kinase C inhibitory activity at nanomolar 20 concentrations. Hypericin and pseudohypericin, both from plants of Hypericum, are also reported as protein kinase C
inhibitors. See Lavie et al~, Proc. Natl. Acad~ Sci. USA
86:5963-s967 (1989). These two compounds also have potent anti-retroviral activity. Calphostins A to I, secondary 25 metabolites isolated from Caldosporium cladosporides, represent another group of protein kinase C inhibitors, with Calphostin C as the most active compound in this series. See Xobayashi et al., J. Antibiotics 42:1470-1474 (1989).
Thus, protein kinase C has been implicated in several 30 human disease processes, including cancer tumors, inflammation, and reper`fusion injury. There remains, however, a long-felt need for efficacious inhibitors of protein kinase C for therapeutic use.

8UMMARY OF ~ INVEN~ION
The present invention provides novel protein kinase C inhibitor isolated from fungi of -the genus Verticillium, W093/03730 PCT/US92/071~4 211S9~ 4 _ preferably the fungus Verticillium balanoides. A preferred, novel compound of the invention has been named Balanol. The novel compounds of the invention are potent protein kinase C
inhibitors, inhibiting protein kinase C activity at nanomolar 5 concentrations.
When substantially pure, the preferred compound of the invention, Balanol, is soluble in dimethyl sulfoxide, water, and methanol; insoluble in ethyl acetate and chloroform; gives a positive color reaction with polymolydbic acid, ninhydrin lO reagent, and ferric chloride; is negative with Dragendorff's reagent and Iodoplatinate spray; has an R~ value of approximately 0.58 with silica gel thin layer chromatography with n-butanol/acetic acid/water at a ratio of 4:l:l respectively; and has a molecular weight of approximately 550.
lS Balanol is believed to have the following formula:

HO~

0~ H~

~S :
`-- `H

The compounds of the present invention are further useful for treating conditions related to or affected by inhibition of protein kinase C activity, particularly 20 hyperproligenative diseases such as cancers, inflammatory disease, myocardial reperfusion injury, and cardiac dysfunctions related to reperfusion injury. Inhibition of protein kinase C activity can lead to inhibition of growth of tumor cells and can thereby produce an anti-tumor effect.
25 Further, inhibition of protein kinase C activity can also lead i !3 9 ~
to inhibition of the oxidative burst in neutrophils, platelet aggregation, and keratinocyte proliferation, leading to an anti-inflammatory effect. The inhibitory activities of the compounds of the invention against platelet aggregation, 5 neutrophil activation, and neutrophil release demonstrate its usefulness in treating reperfusion injury, particularly myocardial reperfusion injury.
This invention is more particularly pointed out in the appended claims and is described in its preferred embodiments 10 in the following description.

BRIBF DE8CRIPTION OF T~E DRAWING8 Figure 1 illustrates an 1H NMR spectrum of Balanol using a 300 MHz dual proton-carbon probe of 5mm.
Figure 2 illustrates a C-13 NMR spectrum of Balanol 15 using a 300 MHz dual proton-carbon probe of Smm.

DETAILED DE8CRIPTION OF ~E INVENTION
The present invention provides a novel protein kinase C inhibitor3 especially Balanol isolated from fungi of the genus Verticillium, preferably from the fungus Verticillium 20 balanoides. Verticill~um balanoides has been described by Dowsett et al. ( 1982) Mycologia 74: 687-690. The fungus used for the isolation of the compound of the invention was recovered from a yellow rhizomorph in Pinus palustris needle litter located in a U.S. Forest Service P. palustris forest 25 near Hoffman, North Carolina. The compound of the invention may also be present in other Verticillium species, or variants, mutants, or recombinants of Vert~cillium balanoides, or in other organisms, however, at the present time the preferred source of~ the compounds of the invention is Verticillium 30 balanoides. Vertic~llium balano~des was deposited in the American Type Culture Collection, Rockville, Maryland, 20852 on July 19, 1991 and has accession number 74082. Balanol can be isolated from the aforementioned fungus using the method described herein in the Examples.

211~99 ~ - 6 -Balanol has been characterized; it is believed that the compound has the following physico-chemical characteristics. The compound generally comprises a liqht yellow amorphous solid that is soluble in dimethyl sulfoxide (DMSO), water, and methanol, but is insoluble in ethyl acetate and chloroform. The~compound gives positive color reactivities with phosphomolybdic acid (PMA) reagent (20% by weight solution in ethyl alcohol), ninhydrin reagent, and ferric chloride, but is negative with Dragendorff's reagent and Iodoplatinate spray lO reagent. The Rf value of the compound is approximately 0.58 using 0.25 mm E Merck silica gel 60F 254 thin layer chromatography (TLC) plate developed with n-butanol/acetic acid/water in a ratio of 4:l:l respectively. The molecular weight of the compound is approximately 550. Other physical 15 ckaracteristics of Balanol are disclosed in Example 3.
The invention relates to the compound having the properties described above together with pharmaceutically acceptable salts of such a compound. The invention relates to pharmaceutical preparations which comprise a pharmaceutically 20 acceptable carrier in combination with either a compound having the properties described above or pharmaceutically acceptable salts of such a compound. Moreover, it is believed that modifications of the Balanol nucleus will also be likely to find utility in therapeutics and as a protein kinase C
25 inhibitor.
Applicants have performed proton nuclear resonance spectroscopy and carbon nuclear resonance spectroscopy on samples of Balanol; the data are reported in Table 4 and Table 5. This data suggest to the inventors that Balanol can be 30 represented by the following formula:

W093/03730 PCT/~S92/07124 , 211S99~

.2 HO ~ N 4, 5 ~ ) 2 6--1 ~H

It is to be understood that the foregoing structure is not yet a cèrtainty and that Balanol is to be identified in the alternative by the structure and but its physical properties set forth above.
It can be appreciated by one having ordinary skill in the art that optical isomers of this structure are possible and that some optical isomers may h~ve more activity than others.
Furthermore, while the above formula is believed to an accurate representation of the structure of the compound of the present lO invention, other isomers are poæsible.
Balanol is useful for treating conditions related to, or affected by inhibition of protein kinase C activity, particularly cancer tumors, inflammatory disease, reperfusion injury, and cardiac dysfunctions related to reperfusion injury.
15 Accordingly, the invention provides methods and pharmaceutical compositions for inhibiting protein kinase C activity.
Preferred methods comprise contacting mammalian tissues and/or fluids with an inhibitory amount of the compound of tbe invention. The pharmaceutical compositions of the invention 20 comprise the compound of the invention preferably in a pharmaceutically acceptable carrier or diluent and in a protein kinase C inhibitory amount.
The invention also provides methods of inhibiting an oxidative burst in neutrophils which comprise contacting a . ~ ~

211~994 - 8 -neutrophil with a protein kinase C inhibitory amount of the compound of the invention, or contacting the neutrophil with an amount of the compound of the invention effective to inhibit such oxidative burst.
The invention further provides methods for treating - inflammation which comprise administering to a mammal suffering from inflammation a protein kinase C inhibitory amount of the compound of the invention, or administering to the mammal an amount of the compound of the invention effective to inhibit inflammation.
This invention additionally provides methods for inhibiting growth of mammalian tumor cells which comprise contacting a mammalian tumor with a protein kinase C inhibitory amount of the compound of the invention, or contacting the 15 tumor cell with an amount of the compound of the invention effective to inhibit growth of the tumor.
Another embodiment of the invention provides methods of inhibiting mammalian keratinocyte cell proliferation which comprise administering to a mammalian keratinocyte a protein 20 kinase C inhibitory amount of the compound of the invention, or administering to the keratinocyte an amount of the compound effective to inhibit proliferation of the keratinocyte.
Cancer is a disease characterized in part by uncontrolled cell growth. Protein kinase C is directly 25 involved in cellular growth control and is believed to be involved in tumor formation. Protein kinase C is the major, if not exclusive, intracellular receptor of phorbol esters which are very potent tumor promoters. Phorbol esters and other tumor promoters bind to and activate protein kinase C. Since 30 diacylglycerol (DAG) and phorbol esters interact at the same site, DAG's have been suggested to be the "endogenous phorbol ~ esters" by analogy with the opiate receptor where the ;~ conservation of a high affihity receptor implied the existence of an endogenous analogue. DAG has been shown to increase the 35 affinity of protein kinase C for Ca 2 and phospholipid and thus activates protein kinase C at cellular levels of these - essential cofactors.

.

~ W093/03730 PCT/US92/07124 g Extracellular signals including hormones, growth factors, and neurotransmitters are known to stimulate phosphatidylinositol turnover resulting in the generation of IP3 and DAG. Structures of 40 distinct oncogenes of viral and 5 cellular origin have revealed that oncogenes encode altered forms of normal cellular proteins. Several of the gene products appear related to growth factors or other elements involved in transmembrane signalling. These oncogene products appear to function by altering the level of critical second 10 messengers. Cells transformed with the oncogenes ras, sis, erbB, ~bl, and src have been shown to contain elevated levels of DAG which is then believed to activate protein kinase C.
Indeed, studies on ras transformed cells have shown protein kinase C activation to be concomitant with elevation of DAG.
~ Phorbol esters, such as phorbol myristate acetate (PMA), have complex effects on cells including effects on membrane function, mit~genesis, differentiation, and gene expression. Synthetic diacylglycerols mimic many of the effects of PMA in vitro and inhibitors of protein kinase C have 20 been shown to block PMA-induced effects on cells. Thus, protein kinase ^ may mediate the actions of certain oncogenes, such as ras, which cause intracellular increases in DAG and concomitant increases in protein kinase C. In addition, activation of protein kinase C leads to the expression of c-25 myc, c-fos, c-cis, c-fms, nuclear protooncogenes important in cell transformation. Overexpression of protein kinase C in NIH
3T3 cells causes altered growth regulation and enhanced tumorigenicity and in rat fibroblasts leads to anchorage-independent growth in soft agar. In these experiments, 30 overexpression of protein kinase C in these cells resulted in tumor formation in animals receiving transplanted cells.
Several studies have shown increased expression of protein kinase C in certain tumor types such as breast and lung carcinomas. Activated protein kinase C has also been detected in human colon carcinomas although increased expression on the gene level was not seen. Topoisomerases are directly modulated by protein kinase C as substrates for the enzyme and protein W093/03730 PCT/USg2/07124 :
211599~ - lO -kinase C inhibitors have been shown to potentiate the action of chemotherapy drugs such as cis-platin. Other compounds which have been identified specifically as inhibitors of protein kinase C have shown early promise as therapeutic agents in 5 inhibiting tumor growth in animal models.
Animal studies have shown that perhaps 50% or more of ischemic-related myocardial damage can be attributed to polymorphonuclear leukocytes (neutrophils) which accumulate at the site of occlusion. Damage from the accumulated neutrophils 10 may be due to the release of proteolytic enzymes from the activated neutrophils or the release of reactive oxygen intermediates (ROI). Much of the "no reflow" phenomenon associated with myocardial ischemia is~attributed to myocardial capillary plugging. The plugging of capillaries has been 15 attributed to both aggregated platelets and aggregated neutrophils. Although both cell types are aggregated during the ischemic event, the relative contribution of each to capillary plugging has not yet been established. It is accepted that ~he damage by neutrophils to myocardial tissue 20 proceeds through a cascade of events, one of the earliest being the bonding of activated neutrophils to damaged vascular ; endothelium. However, the binding of the neutrophils is significantly enhanced by their activation and this an even earlier event is the generation of molecules, such as 25 cytokines, and chemotactic factors, which can function as activation stimuli. These molecules probably originate from damaged and aggregated platelets, from damaged vascular endothelium, or from the oxidation of plasma proteins or lipids by endothelial-derived oxidants.
Strategies for overcoming the deleterious effects of reactive oxygen intermediates have centered on the development of scavengers for the molecules. Superoxide dismutase (SOD) has been shown to be a particularly effective scavenger of superoxide, but suffers from a very short half-life in the 35 blood. Several companies have tackled this problem by creating versions of this enzyme with increased half-lives by techniques such as liposome encapsulation or polyethylene glycol W093/03730 21 1 599 ~ PcT/us92~o7l24 conjugation. Reports on the effectiveness of these new version are mixed. Catalase, a scavenger of hydrogen peroxide, and hydroxyl radical scavengers have also been tested and found to be effective to varying degrees. However, none of the 5 strategies designed to scavenge reactive oxygen intermediates will prevent the aggregation of platelets, the release of chemotactic molecules, the activation and adherence of neutrophils to vascular endothelium, or the release of proteolytic enzymes from activated neutrophils.
An advantage of protein kinase C inhibitors as -~ therapeutics- for reperfusion injury is that they have been demonstrated to 1) block platelet aggregation and release of neutrophil activating agents such as P~AF, 2) block neutrophil activation, chemotactic migration, and adherence to activated 15 or damaged endothelium, and 3) block neutrophil release of proteolytic enzymes and reactive oxygen intermediates. Thus these agents have the capability of blocking all three of the most significant mechanisms of pathogenesis associated with reperfusion iniury and should thus have a decided therapeutic 20 advantage.
The compound of the invention may be administered by - any method that produces contact of the active ingredient with the agent's site of action in the body of a mammal or in a body fluid or tissue including but not limited to oral, topical, 25 hypodermal, intravenous, intramuscular and intraparenteral.
The compound may be administered singly or in combination with other compounds, other pharmaceutical compounds, such as chemotherapeutic compounds, or in conjunction with therapies, such as radiation treatment. The compound of the invention is 30 preferably administered as a composition comprising the compound of the invention and a pharmaceutically acceptable ; ,carrier or diluent selected on the basis of ~he selected route of administration and standard pharmaceutical practice.
.The novel compound of the invention is administered -35 to mammals, preferably humans, in therapeutically effective amounts which are effective to inhibit protein kinase C, or to inhibit tumor cell growth, inhibit inflammation of tissue, .
.

W O 93/03730 PC~r/US92/07124 211599~ - 12 -inhibit keratinocyte cell proliferation, inhibit oxidative burst from neutrophils or inhibit platelet aggregation. The dosage administered in any particular instance will depend upon factors such as the pharmacodynamic characteristics of the 5 compound of the invention, its mode and route of administration; age, health, and weight of the recipient;
nature and extent of symptoms; kind of concurrent treatment, frequency of treatment, and the effect desired. It is contemplated that the daily dosage of the compound will be in 10 the range of from about 1 ~g to about 100 mg per kg of body weight, preferably from about 1 ~g to about 1 mg per kg body weight, and more preferably from about 10 ~g to about 1 mg per kg per day, and preferably administered in a single dosage or in divided dosages~ Persons of ordinary skill will be able to 15 determine dosage forms and amounts with only routine experimentation based upon the considerations of this invention. Pharmaceutically acceptable salts of the compound of the invention are also within the scope of the invention.
The compound of the invention may be administered - 20 orally in solid dosage forms, such as capsules, tablets, and powders, or in liquid dosage forms, such as elixirs, syrups, and suspensions. The compound may also be administered parenterally in sterile liquid dosage forms or topically in a carrier. The compound of the invention may be formulated into 25 dosage forms according to standard practices in the field of pharmaceutical preparations. See Remington 's Pharmaceutical Sciences, A. Osol, Mack Publishing Company, Easton, Pennsylvania.
For example, the compound of the invention may be 30 mixed with powdered carriers, such as lactose, sucrose, mannitol, starch, cellulose derivatives, magnesium stearate, and stearic acid for insertion into gelatin capsules, or for forming into tablets. Both tablets and capsules may be manufactured as sustained release products for continuous 35 release of medication over a period of hours. Compressed tablets can be sugar or film coated to mask any unpleasant taste and protect the tablet from tho atmosphere or enteric ` W O 93/03730 PC~r/US92/07124 211599~

coated for selective disintegration in the gastrointestinal tract.
Liquid .dosage forms for oral administration may contain coloring and flavoring to increase patient acceptance, 5 in addition to a pharmaceutically acceptable diluent such as water, buffer or saline solution.
For parenteral administration, the compound of the invention may be mixed with.a suitable carrier or diluent such as water, a oil, saline solution, aqueous dextrose (glucose), 10 and related sugar solutions, and glycols such as propylene glycol or polyethylene glycols. Solutions for parenteral administration contain preferably a water soluble salt of the compound of the invention. Stabilizing agents, antioxidizing agents and preservatives may also be added. Suitable 15 antioxidizing agents include sodiu~ bisulfite, sodium sulfite, and ascorbic acid, citric acid and its salts, and sodium EDTA.
Suitable preservatives include benzalkonium chloride, methyl-or propyl-paraben, and chlorbutanol.
The following examples are illustrative of the present 20 invention and are not intended to limit the scope of the invention.
It should be noted that a known compound, Ophiocordin, is an antifungal antibiotic which, like Balanol, has the formula C2lH22N20~. However, Ophiocordin is a natural product 25 isolated from Cordyceps ophioglossoides which is known to have the structure:

S.'BSTITI I~E SHEET

W O 93/03730 PC~r/US92/07124 ~ ~ OH
~S I I
`H ~

HO~CO2H

As is apparent, Ophiocordin shares structural groups with Balanol, but they are arranged completely differently. In accord with the different structures, NMR data generated by Balanol and Ophiocordin are different.

EXAMPLES
EXANPLE 1 Growt~ of Verticillium balanoides fungus.
The fungus Verticillium balanoides tMYCOsearch, Durham, North Carolina, accession number 25901) was grown on 10 Yeast Extract Peptone Dextrose ~YePD), Malt, Cornmeal, Potato Dextrose (PDA), and Czapek's agar media for 7 days at 21C and 37OC. Fungal colonies of approximately 2.5 mm in diameter were grown on Malt at 21C and exhibited a white to Dresden Brown color according to Color Standard and Color Nomenclature, 15 R.Ridgeway, Washington, D.C. 19122. In reverse, the colonies were Dresden Brown to Cinnamon in color. These colonies had an appressed to tomentose morphology. On Cornmeal, the colonies measured approximately 3.4 mm in diameter having a white color, no reverse color and a felted morphology. Colonies grown on 20 YePD were approximately 5.0 mm in diameter having a white color with no reverse color and with minutely tomentose and slightly furrowed morphology. Growth on PDA yielded colonies of approximately 3.4 mm in diameter having a white to tawny color, - a white to tawny color on the reverse and slightly furrowed W093/03730 PCT/US92/07t24 211599q morphology. Growth on Czapek's medium yielded colonies of approximately 1.4 mm in diameter having a white to primuline yellow color, a primuline yellow reverse and minutely tomentose morphology. The fungi exhibited substantially no growth in any 5 media at 37C. Fungal colonies had substantially no odor.
Mycelium comprised straight, septate, branching hyphae of approximately 1 to 5 ~m in diameter having frequent swellings to approximately 7 ~m in diameter.
Conidiogenous cells were phialidic and micronematous;
10 arose substantially directly from the arial mycelium; were solitary, lateral or terminal; and had dimensions of approximately 10 to 30 ~m by 0.8 to 2.5 ~5~0) ~m in diameter.
These cells further comprised a swollen base of the phialid~s of approximately 7 to 15 ~m in length that tapered to 15 approximately 0.8 ~m in diameter. Conidia were cuneiform, hyaline, smooth-walled and aseptate being approximately 2.5 to
3.5 ~m long and 1.8 to 2.2 ~m wide at the apex; further, being 1.0 to 1.3 ~m wide at the base.
Fungi were recovered by Barry Xatz of MYCOsearch Inc.
20 from a yellow rhizomorph in ~inus palustris needle litter located in a U. S. Forest Service P . pal ustris f orest near Hoffman, North Carolina. Verticil l ium bal anoides has MYCOsearch accession no. 25901.

EXAMP~E 2 F~rment~tion of Verticillium balanoides fungus.
The fungus Verticil l ium bal anoides ~which was deposited in the American Type Culture Collection, Rockville, Maryland 20854 on July 1~, lg91 and has accession number 74082) as described in Example 1 was maintained on malt extract agar which contained 1% malt extract and 1.8% agar. The organism 30 was transferred into a Corning 50 ml conical centrifuge tube containing 6 ml of YePD broth which consists of 1.0% Difco yeast extract, 2.0% Sigma peptone, and 2.0% dextrose (6 ml culture). The 6 ml culture was then incubated for 7 days at 21C while shaking at 200 rpm.
The 6 ml culture ~as then added to a 250 ml ~rlenmeyer flask containing 75 ml cornmeal broth. The cornmeal broth used W093~03730 PCT/US92~07124 ~ilS 9 9 4 - 16 -- consisted of 2.0% organically grown cornmeal, 2.0% tomato paste and 1.0% yeast extract. The culture was incubated for 12 days at 21C under shaking conditions at 200 rpm. Flask contents were then filtered to separate the filtrate from the mycelia 5 mass. t For the preparation of the 10 1 size fermentations, multiple 250 ml Erlenmeyer flasks were employed. The 6 ml culture was first transferred to 75 ml YePD broths and incubated for 7 days. 10 ml of such seed cultures were used to 10 inoculate 75 ml cornmeal broth. After cultivation, the cultures were combined and filtered to separate the mycelia and the filtrate.

EXAMRLE 3 Isol~tion Crude methanol extracts of both mycelia and freeze-15 dried filtrate showed protein kinase C inhibitory activity(ICso) of less than approximately 5 ~g/ml in the protein kinase C inhibition assay explained in detail in Example 5.
Fractionation to isolate the protein kinase C inhibitor of the invention was guided by such enzyme assays. All fractions at 20 each purification step were evaluated and only those which showed improved protein kinase C inhibitory activity were - further fractionated.
The mycelia or freeze-dried filtrate from 10 1 culture was stirred in 1.5 1 of methanol at room temperature and was 25 filtered under vacuum. Such extraction procedures were repeated 4-5 times. The filtrates were combined and concentrated in vacuo to give a dark brown residue which was then dissolved in 200 ml water and extracted with 200 ml n-butanol (n-BuOH) 3 times. In the case of mycelia, it was 30 necessary to wash the water and crude extract mixture with ethyl acetate prior to extraction by n-BuOH. The extraction with n-BuOH was repeated until protein kinase C activity of the water layer dropped to an IC50 of less than approximately 30 ~g/ml. The combined n-BuOH extracts were concentrated in Yacuo 35 at room temperature yielding a dark brown residue which had protein kinase C inhibitory activity with an IC5Q of 5 ~g/ml.

eCTjUS 52Jo7 1~4 21l59~ ~O/US 28S~Pl992 The crude n-BuOH extracts were then fractionated on a gel permeation column (crosc-linked dextran, Sephadex LH-20, Pharmacia, Inc., New Jersey) equilibrated with chloroform/methanol (CHCl3/MeOH) at a ratio of 3:1. The column 5 was eluted with a gradient composition of CHCl3/MeOH
(3:1 followed by 2:1, 1:1 then finally by 100% methanol (MeOH)). The fractions were pooled according to thin layer chromatography (TLC) patterns on Silica gel (E Merck) developed with n-BuOH/HOAc/H20 at a ratio of approximately 4:1:1 10 respectively and visualized by W lamp and ninhydrin spray reagent. The desired protein kinase C inhibitor was present in the last fractions eluted from the column with 100% methanol.
The most active fraction from the above chromatography was further fractionated by rever~e phas~ HPLC using a C
15 column (octadecyldimethylsilyl bonded phase silica gel, Waters microBondapak) under gradient condition~ with acetonitrile (CH3CN) and water. The elution of active component was monitored by W at 254 um. Lyophilization of HPLC fractions yielded pure light yellow solid material.

20 ~SANPL~ ~ Phy~ioo-ch--lc~ Ch~r~ct-ristic~.
The phy~ico-chemical properties of the compound of ~ Example 3 are as follows. The compound appears as a light yellow amorphous solid that is soluble in dimethyl sulfoxide (DMSO), H20, and methanol (MeOH) and insoluble in ethyl acetate (EtOAc). The compound give~ a positive color reaction with phosphomolybdic ~cid reagent (PMA) (20% by weight solution in ethyl alcohol), ninhydrin, and ferric chloride (FeCl3), but is negative with Draqendorff's reagent and Iodoplatinate spray reagent. The R~ value of the compound is approximately 0.58 30 using E Merck silica gel 60F 254 thin layer chromatography ~TLC) plates with n-butanol/acQtic acid/w~ter (n-BuOH/HOAc/H20) at a ratio of 4:1:1 re~pectively in à thin layer chromatographic analysis. Further, the molecular weight of the compound is approximately 550. Mass spectrum analysis using a 35 FAB, VG-Analytical ZAB 2-SF high field mass spectrometer showed (M+H) ion 551. The ultraviolet spectrum (Shimadzu) of the S'..!E~STITUTE Sh'E~

2~81599~ ~~tls 2 8S~P 1~92 compound when neutral (H20) and acidic (0.1 N HCl) has a maximum at approximately 256 nm and an extinction coefficient of approximately 22,807; and when ba~ic (0.1 N NaOH) has a maximum at approximately 289 nm and an extinction coefficient s of approximately 37,368. ~ NMR using a 300 MHz Varian Gemini dual proton-carbon probe 5 mm showed peaks at approximately - (MeOH-d~ 7.40 (lH, d, 8.7 Hz), 7.06 (lH, d, 8.1 Hz), 6.7 (lH, t~ 7.9 Hz), 6.70 tlH, s), 6~5 (2H, dd, 7.9 Hz, 8.0 Hz), 5.08 (lH, dt, 3.4, 5.0 Hz), 4.38 (lH, m), 4.13 (lH, m), 2.7-10 3.05 (4H, m), and 1.55-1.95 (4H, m).
An example of the instrument settings for the 300 MHz Varian Gemini dual proton-carbon probe of 5 mm in a proton NMR
analysis i8 illustrated in Table 1. The analysis utilized pulse sequence S2Pul, and CD30D a~ a solvent. The lH NMR
15 spectru~ of the compound of the invention using a 300 MHz dual proton-carbon probe of 5mm i~ depicted in Figure 1.

ACQ~I8IT~ON D~C ~ V~
TN 1.000 DN 1.000 20 SW 4500.5 DO -450.0 AT 1.778 DM NNN
NP 16000 DHP 1.0 PW 8.0 DLP 20 Pl 0 HOMO N
25 Dl 0 D2 0 P~OCJ88INa CT 128 DI~PLaY
30 PW90 21.2 SP -38.5 FB 2250 WP 2470.3 DP Y RFL 557.6 INS 1.000 An example of the instrument settings for the 300 MHz Varian Gemini dual proton-carbon probe of 5 mm in a C-13 NMR
analysi~ i8 illustrated in Table 2. The analysis utilized CD30D as a solvent. The C-13 NMR spectrum of the compound of SUBSnTUIl'E SHEET

W093/03730 2 1 1 5 9 9 ~ PCT~US92/07124 -- 19 -- .
the invention using a 300 MHz dual proton-car~on probe of 5mm is depicted in Figure 2.

TABI~ 2 ACQ~I8ITION DE~ ~ VT
5 TN 13.000 DN 1.000 SM 18761.7 DO -450.0 AT 0.8~0 DM YYY

PW 8.0 DMF 8500 10 P1 0 DHP 1.0 Dl 0 HOMO N
D2 o TO 0 PRoC~R8IN~
NT 1.00E 9 SE 0.318 15 CT 36352 LB 1.o00 PWso 23.6 PN 32768 PP 21.2 SN 35 20 SS 0 DI8~LAY
IL N SP -lllg.9 IN Y WP 18761.7 AFL 1119.9 TH g INS 1.000 ~SAMPLE S ~rotein ~ina~- C Inhib~tion.
The protein kinase C assay is designed to duplicate the in vivo conditions required for protein kinase C function.
Therefore, pH, salt and cofactor concentrations are similar to 35 physiological levels. Histone Hl (lysine rich) is used in the assay as the phosphorylation acceptor protein because it is readily available and serves as a good substrate for protein kinase C~ The enzyme is prepared from rat brain and is purified to apparent homogeneity as determined by a single band 40 on silver stained sodium dodecyl sulfate (SDS)-polyacrylamide.
In the screening assay, phosphatidylserine (PS) and DAG are co-sonicated to form unilamellar and multilamellar vesicles. The concentration of lipids in the assay are SUE~STlllJTE SHl~E~

W093/03730 PCT~US92/07124 2~994 - 20 -~ suboptimal to maximize the detection potential of the assay for inhibitors. Potential inhibitor compounds are added to the assay in dimethylsulfoxide (DMSO) at three concentrations to give final inhibitor concentrations of 4.3, 43 and 218 ~M, 5 respectively. The assay is started with the addition of enzyme and stopped after 10 min by the addition of 25% trichloroacetic acid (TCA) and 1.0 mg/ml bovine serum albumin (BSA). The radioactive histone product is retained and washed on glass fiber filters that allow the unreacted 32P-ATP to pass through.
10 The amount of phosphorylation is determined by the radioactivity measured in a scintillation counter. Controls are included in every assay to measure background activity in the absence of enzyme, activity in the absence of lipids and the maximum enzy~me activity with saturating levels of the 15 activator lipids. Table 3 shows the components and their concentration used in the protein kinase C assay.

Ass~y Component Concentration HEPES pH 7 . 5 2 0 ~M
MgC12 . - 20 ~LM : :~
CaCl2 100 ,uM
EGTA 95 ,UM
Histone Hl 200 ~g/ml ~
Phosphatidylserine 40 ~g/ml `
25 Diacylglycerol 1.8 ~g/ml Pr~tein Kinase C 0.6 ~g/ml y-- P-ATP 2 0 ,UM
HEPES is N-t2-hydroxyethyl~ piperazine-N'-12-ethanesulfonic acidl and EGTA is Ethylene-30 bis(oxyethylenenitrilo) tetraacetic acid.
Protein kinase C assay results are measured as an ICsovalue, which is the concentration of test compounds needed to inhibit 50% of the protein kinase C activity as compared with levels of protein kinase C activity in controls. The compound 35 of the invention was able to effectively inhibit protein kinase activity.
The compound of the present invention exhibited potent protein kinase C inhibitory activity with an IC50 of 5 ng/ml (9 nM)- -WO 93/03730 211 5 9 9 ~ PCI`/US92/07124 ~: o ~1 ~

a~t~
a D. ~ o O J~
u ~o o ~r 1` ` `
a~ ~ t~ o o~
C~ ~ U~ N
N U~ 0 CO OD CO

D~

CO
E~ ~
, ::
~ ~ ' U
e ~
~: ~ Q :
~:

~J ~
O tO o a~ r o ,1 ~ to N er o~ 1 0 ~ o ~o 1` ~ ~ o ~........................................ 'u t~
.
1` ~ ~: ~
O ~I N 1' 0 ~ r ~ S
U) O U~ O

2~S99~ - 22 -.~

~1 0 ~ ~I H H ~ ~ N --I H H ~ N -1 H H H _I H ~ ~1 H
O ~ .
1 ~
~H

P
O
U
O
U

U
0~
~n P:

'0 13 ~ ~ O u~ N 1~ ~ O 1~ ~ ~ r O ~ 0~ ~ O r~
ZU ~ ~ ~ O O t~ N0~ r O et~ ~ 1~ 0~ 03 H el' Cl~ ~r U) O t~ ~ 1~
~-- o u~ ~ o u~ ~ o i O~ ~ ~O O~ CO ~ ~i O O It) In 1~ N N el~ ~D N N H ~ ~ O l0 N O N 11 ~I N H ~ 1~
C~

~i N 1~ U) It~ H N 1~ U~ U~ 0 0~ 0 j N ~

t~ N ~ ~ Ul ~D r`

U) O ' U~ O - U~
H H N N

Claims (12)

What is claimed is:
1. A compound isolatable from Verticillium balanoides having protein kinase C inhibitory activity which compound is soluble in dimethyl sulfoxide, water, and methanol; insoluble in ethyl acetate and chloroform; gives a positive color reaction with phosphomolybdic acid, ninhydrin reagent, and ferric chloride; is negative with Dragendorff's reagent and Iodoplatinate spray; has an Rf value of approximately 0.58 with silica gel thin layer chromatography with n-butanol/acetic acid/water at a ratio of 4:1:1 respectively; has a molecular weight of approximately 550; the ultraviolet spectrum of said compound when neutral (water) and acidic (0.1 N HCl) has a maximum at approximately 256 nm and an extinction coefficient of approximately 22,807; and when basic (0.1 N NaOH) has a maximum at approximately 289 nm and an extinction coefficient of approximately 37,368; or a pharmaceutically acceptable salt thereof.
2. A composition comprising:
a pharmaceutically acceptable carrier or diluent and, a therapeutically effective amount of the compound of claim 1 or a pharmaceutically acceptable salt thereof.
3. A method of inhibiting protein kinase C comprising contacting a protein kinase C with an inhibitory amount of the compound of claim 1.
4. A method of inhibiting protein kinase C comprising contacting a protein kinase C with an inhibitory amount of the compound of claim 2.
5. The use of the composition of claim 1 in the manufacture of a medicament for inhibiting protein kinase C.
6. The use of the composition of claim 1 in the manufacture of a medicament for inhibiting growth of mammalian tumor cells.
7. A compound using the formula:

or a pharmaceutically acceptable salt thereof.
B. A composition comprising:
a pharmaceutically acceptable carrier or diluent and, a therapeutically effective amount of the compound of claim 6 or a pharmaceutically acceptable salt thereof.
9. A method of inhibiting protein kinase C comprising contacting a protein kinase C with an inhibitory amount of the compound of claim 8.
10. A method of inhibiting protein kinase C comprising contacting a protein kinase C with an inhibitory amount of the compound of claim 8.
11. The use of the composition of claim 6 in the manufacture of a medicament for inhibiting protein kinase C.
12. The use of the composition of claim 6 in the manufacture of a medicament for inhibiting growth of mammalian tumor cells.
CA002115994A 1991-08-22 1992-08-21 Protein kinase c inhibition and novel compound balanol Abandoned CA2115994A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US74863291A 1991-08-22 1991-08-22
US748,632 1991-08-22

Publications (1)

Publication Number Publication Date
CA2115994A1 true CA2115994A1 (en) 1993-03-04

Family

ID=25010269

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002115994A Abandoned CA2115994A1 (en) 1991-08-22 1992-08-21 Protein kinase c inhibition and novel compound balanol

Country Status (5)

Country Link
EP (1) EP0664706A1 (en)
JP (1) JPH06510280A (en)
AU (1) AU2504192A (en)
CA (1) CA2115994A1 (en)
WO (1) WO1993003730A1 (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH09503994A (en) * 1993-03-03 1997-04-22 イーライ・リリー・アンド・カンパニー Balanoid
AU686691B2 (en) * 1994-01-12 1998-02-12 F. Hoffmann-La Roche Ag Novel azepanes and homologs thereof
US5583221A (en) * 1994-05-04 1996-12-10 Eli Lilly And Company Substituted fused and bridged bicyclic compounds as therapeutic agents
TW339325B (en) * 1995-07-05 1998-09-01 Hoffmann La Roche Novel azepane derivatives, process for the preparation thereof and pharmaceutical composition containing the same
JP2000504672A (en) * 1996-02-09 2000-04-18 フィテラ シンビオン エイピーエス Balanol analogues
US5902882A (en) * 1996-04-17 1999-05-11 Hoffmann-La Roche Inc. Assymetric synthesis of azepines
US6914140B1 (en) 1996-04-17 2005-07-05 Hoffmann-La Roche Inc. Asymmetric synthesis process
US6232299B1 (en) * 1996-05-01 2001-05-15 Eli Lilly And Company Use of protein kinase C inhibitors to enhance the clinical efficacy of oncolytic agents and radiation therapy
WO2006102267A2 (en) * 2005-03-21 2006-09-28 The Trustees Of Columbia University In The City Of New York New neuronal pain pathway
US8846742B2 (en) 2006-02-14 2014-09-30 The Trustees Of Columbia University In The City Of New York Neuronal pain pathway modulators

Also Published As

Publication number Publication date
JPH06510280A (en) 1994-11-17
EP0664706A4 (en) 1995-06-14
AU2504192A (en) 1993-03-16
EP0664706A1 (en) 1995-08-02
WO1993003730A1 (en) 1993-03-04

Similar Documents

Publication Publication Date Title
US6214340B1 (en) Physiologically active substance sulphostin, process for producing the same, and use thereof
US7335679B2 (en) Wortmannin analogs and methods of using same
CA2115994A1 (en) Protein kinase c inhibition and novel compound balanol
WO2003024183A2 (en) Wortmannin analogs and methods of using same
PT94555B (en) METHOD FOR PRODUCING AN EQUINOXANDIN B DERIVATIVE, USEFUL AS ANTIBIOTIC
JP4769959B2 (en) Antitumor agent
EP1548026A1 (en) Novel depsipeptide compound
JPH09176083A (en) New compound f-12509a
US5550148A (en) PAF synthesis modulators
KR100516105B1 (en) 13-deoxyanthracycline derivatives and processes for preparing them
US7470714B2 (en) GM-95-containing antitumor effect potentiator, combined antitumor preparation and antitumor agent
US5801254A (en) Arteminolide derivatives and process for the manufacture thereof
JP3786461B2 (en) New physiologically active substance
GB2296496A (en) Pharmaceutical pregnane derivatives
JPH10182627A (en) New compound s-15183a and s-15183b
US7528169B2 (en) Substance having antitumor/anti-inflammatory activity
EP0322748A1 (en) Antibiotic NK86-0279, process for production of the same and application of the same
JP3733163B2 (en) Compound TAN-2177, its production and use
KR100663188B1 (en) Novel heptatrienoic acid substituted bicyclic ketone derivative and pharmaceutical compositions comprising the same
WO2005003076A1 (en) Novel heptatrienoic acid substitued bicyclic ketone derivative and pharmaceutical compositions comprising the same
KR100327043B1 (en) Grb2-shc binding inhibitory compounds, process for isolating them from a microorganism and composition comprising same for inhibiting grb2-shc binding
JP4452787B2 (en) New fermentation products
JP2005272357A (en) Anticancer agent
JP2002114741A (en) New compound f-16053a
JP2004275060A (en) Hyaluronic acid-cd44 binding inhibitor and method for producing the same

Legal Events

Date Code Title Description
FZDE Dead