US20030195168A1 - Therapeutic agents - III - Google Patents

Therapeutic agents - III Download PDF

Info

Publication number
US20030195168A1
US20030195168A1 US10/315,319 US31531902A US2003195168A1 US 20030195168 A1 US20030195168 A1 US 20030195168A1 US 31531902 A US31531902 A US 31531902A US 2003195168 A1 US2003195168 A1 US 2003195168A1
Authority
US
United States
Prior art keywords
euphorbia
derivative
sonr
onr
sor
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
US10/315,319
Inventor
James Aylward
Peter Parsons
Andreas Suhrbier
Kathleen Turner
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.)
Peplin Research Pty Ltd
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
Assigned to PEPLIN RESEARCH PTY LTD reassignment PEPLIN RESEARCH PTY LTD ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SUHRBIER, ANDREAS, TURNER, KATHLEEN, AYLWARD, JAMES HARRISON, PARSONS, PETER GORDON
Publication of US20030195168A1 publication Critical patent/US20030195168A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/455Nicotinic acids, e.g. niacin; Derivatives thereof, e.g. esters, amides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/47Euphorbiaceae (Spurge family), e.g. Ricinus (castorbean)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/10Anti-acne agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/02Local antiseptics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/10Anthelmintics
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • 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
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/52Esters of acyclic unsaturated carboxylic acids having the esterified carboxyl group bound to an acyclic carbon atom
    • C07C69/533Monocarboxylic acid esters having only one carbon-to-carbon double bond
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A90/00Technologies having an indirect contribution to adaptation to climate change
    • Y02A90/10Information and communication technologies [ICT] supporting adaptation to climate change, e.g. for weather forecasting or climate simulation

Definitions

  • the present invention relates generally to chemical agents useful in the treatment and prophylaxis of protein kinase C (PKC) related conditions in mammals, including humans and primates, non-mammalian animals and avian species. More particularly, the present invention provides a chemical agent of the macrocyclic diterpene family obtainable from a member of the Euphorbiaceac family of plants or botanical or horticultural relatives thereof or derivatives or chemical analogues or chemically synthetic forms of the agents for use in the treatment or prophylaxis of PKC-related conditions in mammalian, animal and avian subjects. The subject chemical agents are also useful for modulating expression of genetic sequences including promotion and other regulatory sequences.
  • PKC protein kinase C
  • the present invention further contemplates a method for the prophylaxis and/or treatment in mammalian, animal or avian subjects with PKC-related conditions by the topical or systemic administration of a macrocyclic diterpene obtainable from a member of the Euphorbiaceae family of plants or their botanical or horticultural derivatives or a derivative, chemical analogue or chemically synthetic form of the agent.
  • the chemical agent of the present invention may be in the form of a purified compound, mixture of compounds, a precursor form of one or more of the compounds capable of chemical transformation into a therapeutically and/or genetically active agent or in the form of a chemical fraction, sub-fraction, preparation or extract of the plant.
  • Natural product screening is a term applied to the screening of natural environments for bioactive molecules. Particularly sought after bioactive molecules are those having potential as useful therapeutic agents. Natural environments include plants, microorganisms, coral and marine animals. The search for potential therapeutic agents for the treatment of cancer and infection by pathogenic organisms remains an important focus.
  • the Euphorbiaceae family of plants covers a wide variety of plants including weeds of Euphorbia species. There have been a variety of inconclusive reports on the potential effects of the sap of these plants on a variety of conditions as well as promoting tumorigenesis and causing skin and ocular irritation.
  • Euphorbia pilulifera L (synonyms E. hirta L., E. capitata Lam. ), whose common names include pill-bearing spurge, snakeweed, cat's hair, Queensland asthma weed and flowery-headed spurge.
  • the plant is widely distributed in tropical countries, including India, and in Northern Australia, including Queensland.
  • a recent report describes selective cytotoxicity of a number of tiglilane diterpene esters from the latex of Euphorbia poisonii , a highly toxic plant found in Northern Nigeria, which is used as a garden pesticide.
  • One of these compounds has a selective cytotoxicity for the human kidney carcinoma cell line A-498 more than 10,000 times greater than that of adriamycin (Fatope et al., 1996).
  • Euphorbia hirta plants and extracts thereof have been considered for a variety of purposes, including tumor therapy (EP 0 330 094), AIDS-related complex and AIDS (HU-208790) and increasing immunity and as an anti-fungoid agent for treatment of open wounds (DE-4102054).
  • the inventors have identified chemical agents and fractions comprising these agents which are useful in the treatment and prophylaxis of PKC-related conditions in an animal, mammal or avian species.
  • the instant agents are also useful in modulating and in particular stimulating or otherwise promoting expression or function of nucleotide sequences such as promoter or other regulatory sequences which is useful in gene therapy, genetic therapy, genetic augmentation and gene replacement therapies as well as promoting expression of genetic material into an expression product.
  • the present invention is predicated in part on the identification of chemical agents and fractions comprising same from plants of the Euphorbiaceae family which are useful in the treatment and prophylaxis of PKC-related conditions in an animal, mammal incluidng a human and avian species.
  • the inventors have identified that the chemical agents of the present invention are capable of modulating protein kinase C (PKC) activity thus providing a basis for the treatment of conditions where PKC activity is required to be up-regulated or down-regulated.
  • PKC protein kinase C
  • the instant chemical agents further modulate and more particularly activate or otherwise promote expression of genetic sequences and/or operation of promoter or other regulatory sequences. The latter is useful for genetic therapies including gene therapy.
  • one aspect of the present invention contemplates a method for the treatment or prophylaxis of a PKC-related condition or disorder in a subject, said method comprising the administration to said subject of a symptom-ameliorating effective amount of a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or chemical analogue thereof which chemical agent is a macrocyclic diterpene selected from compounds of the ingenane, pepluane and jatrophane families and which chemical agent or derivative or chemical analogue is represented by any one of the general formulae (I)-(V) as defined herein and which chemical agent or derivative or chemical analogue thereof is capable of modulating PKC activity, PKC-gene expression or PKC enzyme turnover and wherein said chemical agent or its derivatives or chemical analogues is administered for a time and under conditions sufficient to ameliorate one or more symptoms associated with a PKC-related condition or disorder.
  • a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or
  • Another aspect of the present invention provide a method for the treatment or prophylaxis of a PKC-related condition or disorder in a subject, said method comprising the administration to said subject of a symptom-ameliorating effective amount of a macrocyclic diterpene, or a chemical fraction comprising same from a plant of the family Euphorbiaceae or a derivative or chemical analogue of said macrocyclic diterpene having the structures as defined above wherein said macrocyclic diterpene or its derivative or chemical analogue modulates PKC activity, synthesis or enzyme turnover, said administration being for a time and under conditions sufficient to ameliorate one or more symptoms of associated with a PKC-related condition or disorder.
  • Yet another aspect of the invention encompasses a method of modulating the expression of a genetic sequence for the treatment or prophylaxis of a condition or disorder in a subject, said genetic sequence being under the control of a promoter whose activity is modulated by a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or chemical analogue thereof which chemical agent is a macrocyclic diterpene selected from compounds of the ingenane, pepluane and jatrophane families and which chemical agent or derivative or chemical analogue is represented by any one of the general formulae (I)-(V) as defined herein and which chemical agent or derivative or chemical analogue thereof is capable of modulating PKC activity, PKC-gene expression or PKC enzyme turnover, said method comprising administering to said subject an expression facilitating amount of said chemical agent or derivative or chemical analogue thereof for a time and under conditions sufficient to facilitate the expression of said genetic sequence to thereby ameliorate one or more symptoms associated with said condition or disorder.
  • a chemical agent obtainable from
  • Still another aspect in the invention envisions a method of modulating the expression of a genetic sequence by the administration of a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or chemical analogue thereof which chemical agent is a macrocyclic diterpene selected from compounds of the ingenane, pepluane and jatrophane familes and which chemical agent or derivative or chemical analogue is represented by any one of the general formulae (I)-(V) as defined herein and which chemical agent or derivative or chemical analogue thereof is capable of modulating PKC activity, PKC-gene expression or PKC enzyme turnover and wherein said chemical agent or its derivatives or chemical analogues is administered for a time and under conditions sufficient to facilitate the expression of said genetic sequence.
  • a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or chemical analogue thereof which chemical agent is a macrocyclic diterpene selected from compounds of the ingenane, pepluane and jatrophane fa
  • Still yet another aspect of the present invention features a method of stimulating the activation or function of a promoter by the administration of a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or chemical analogue thereof which chemical agent is a macrocyclic diterpene selected from compounds of the ingenane, pepluane and jatrophane families and which chemical agent or derivative or chemical analogue is represented by any one of the general formulae (I)-(V) as defined herein and which chemical agent or derivative or chemical analogue thereof is capable of modulating PKC activity, PKC-gene expression or PKC enzyme turnover and wherein said chemical agent or its derivatives or chemical analogues is administered for a time and under conditions sufficient to stimulate the activation or function of said promoter.
  • a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or chemical analogue thereof which chemical agent is a macrocyclic diterpene selected from compounds of the ingenane, pepluane and jatrophane families
  • Still yet another aspect of the present invention contemplates a method for the treatment or prophylaxis of a PKC-related condition in a subject, said method comprising the administration to said subject of a symptom-ameliorating or immunopotentiating effective amount of a macrocyclic diterpene obtainable from a Euphorbiaceae plant or its botanical or horticultural relative, said macrocyclic diterpene being selected from an ingenane, pepluane or jatrophane, or a derivative or chemical analogue thereof, having the structure represented by any one of the general formulae (I)-(V) as defined herein and which chemical agent or derivative or chemical analogue thereof is capable of modulating PKC activity, PKC-gene expression or PKC enzyme turnover and wherein said chemical agent or its derivatives or chemical analogues is administered for a time and under conditions sufficient to ameliorate one or more symptoms associated with said biological entity and wherein said chemical agent exhibits a potency of agent (P A ) of >10, wherein
  • FIG. 1 shows the activation of PKC, using a fluorescent peptide assay (“PepTag” non-radioactive protein kinase kit, Promega).
  • Lane 1 PKC and substrate alone; lane 2, plus positive control activator; lane 3, plus 100 ng/ml TPA; lane 4, plus 0.1 ng/ml TPA; lane 5, plus 0.01 ng/ml TPA; lane 6, plus 0.001 ng/ml TPA; lane 7, ether extract of E. peplus sap in DEM, diluted 1 in 5; lane 8, aqueous layer from ether extraction, diluted 1/25; lane 9, crude sap diluted 1/25; lane 10, DME alone.
  • PepTag fluorescent peptide assay
  • FIG. 2 shows the activation of PKC by E. peplus fractions. Lanes 1 and 2, same as FIG. 1; lane 3, 2 mg/ml fraction H; lane 4, 2 mg/ml ingenanes.
  • FIG. 3 is photographic representation showing the results of a PKC assay using rat brain PKC.
  • Lane 1 negative control; lane 2, positive control; lane 3, empty; lane 4, PEP001 (1/125 dilution), lane 5, PEP001 (1/500 dilution) and lane 6, TPA (20 ⁇ g).
  • FIG. 4 is a photographic representation showing the activation of PKC in MM96L cells expressing PKC fused to green fluorescent protein (GFP).
  • GFP green fluorescent protein
  • FIG. 5 is a photographic representation showing induction of translocation of activated PKCs by the compounds of the instant invention to the cytoplasm, plasma membrane and to the Golgi or similarly located cellular structure.
  • FIG. 6 is a graphical representation showing the induction of translation of the classical and novel PKC isoforms in response to PEP003, PEP005, bryostatin-1 and TPA.
  • FIG. 7 is a graphical representation showing the activation of HIV from U1 cells.
  • FIG. 8 is a graphical representation showing treatment of lytic HIV infection of peripheral blood mononuclear cells (PBMC) with PEP003, PEP004, TPA and ingenol, expressed as p24 production over a 10 day treatment period.
  • PBMC peripheral blood mononuclear cells
  • A Uninfected cells
  • B low titer infected cells
  • C low titer infected cells represented as p24 production versus drug concentration
  • D same as (C) but high titer infection.
  • FIG. 9 is a photographic representation showing the recruitment of neutrophils in the skin induced by PEP001 extract.
  • A Normal skin of nude mouse.
  • B Skin of nude mouse showing infiltration of neutrophils one day after treatment with E. peplus sap.
  • FIG. 10 is a photographic representation showing effect of PEP010 onrecruitment of neutrophils in normal skin of nude mouse and skin overlying subcutaneously implanted B16 melanoma.
  • A 24 hr treatment
  • B 48 hr treatment.
  • FIG. 11 is a graphical representation illustrating the ability of PEP001 to induce the release of superoxide radical, as demonstrated by fluorescence-activated cell sorting.
  • FIG. 12 is a graphical representation showing the effect of pre-treatment of leukocytes with PEP003 on E. coli activity (16 hr incubation), relative to PBS control; depicted as numbers of E. coli cells/ml media.
  • FIG. 13 is a graphical representation showing the effect of pre-treatment of leukocytes with PEP003 on E. coli numbers depicted in terms of turbidity.
  • FIG. 14 is a photographic representation showing production of viral capsid antigen (VCA) in B95-8 (EBV+ Marmoset cell line) after treatment with TPA, PEP003 and PEP004 for 3 and 7 days.
  • VCA viral capsid antigen
  • FIG. 15 is a photographic representation showing production of viral capsid antigen (VCA) in BL74 and Mutu I (Burkitts lymphoma cell lines) after treatment with TPA, PEP003 and PEP004 for 3 and 7 days.
  • VCA viral capsid antigen
  • FIG. 16 is a photographic representation showing production of BZLF1 (the initial transactivator of EBV) after treatment with TPA, PEP003 and PEP004 for 3 and 7 days.
  • FIG. 17 is a graphical representation showing activation of natural killer cell activity, assayed as % specific lysis of K562 cells (a natural killer-sensitive cell line) after pre-treatment of AO2-M melanoma cells with PEP003 and TPA.
  • FIG. 18 is a graphical representation showing survival of Jam cells after treatment with saps from the Euphorbiaceae, expressed as percentage cell survival determined by sulfurhodamine B staining of cells.
  • FIG. 19 is a diagrammatic representation of a system used to carry out the instructions encoded by the storage medium of FIGS. 9 and 10.
  • FIG. 20 is a diagrammatic representation of a cross-section of a magnetic storage medium.
  • FIG. 21 is a diagrammatic representation of a cross-section of an optically readable data storage system.
  • Compounds may be referred to in the subject specification by a compound code. These are defined as below: TABLE OF COMPOUND CODES COMPOUND CODE DESCRIPTION PEP001 Crude sap PEP002 Methanol and ether extract of E.
  • the present invention is predicated in part on the identification of biologically useful properties of chemical agents and chemical fractions comprising these agents obtainable from a member of the Euphorbiaceae family of plants or their botanical or horticultural relatives. These biologically useful properties include their use in the prophylaxis and/or treatment of PKC-related conditions.
  • PKC-related condition includes a PKC-related disorder and comprises medical including psychological conditions associated with aberrant PKC activity including activating low PKC activity or lowering higher than normal levels of PKC as well as conditions associated with the presence of latent virus in a host cell.
  • Examples of PKC disorders include but are not limnited to alcoholism, Alzheimer's disease, asthma, atherosclosis, atopic dermatitis, autoimmune disease, bipolar-disorder, blood disorder, cardiac hypertrophy, depression, diabetes, hypertension, hyperplastic dermatosis, multiple sclerosis, myocardial ischemia, osteoarthritis, psoriasis, rheumatoid arthritis and transplantation.
  • the instant chemical agents are also useful in promoting gene activation and promotion.
  • treatment is used in its broadest sense and includes the prevention of a PKC-related condition or the reversal of a condition to asymptomatic levels or to levels in between as well as facilitating the amelioration of the effects of symptoms of the PKC-related condition.
  • a PKC-related condition occurs when an increase or decrease in PKC promotes or facilitates an effect.
  • treatment further encompasses promoting an elevation in promoter or other regulatory sequence activity and/or structural gene sequence expression.
  • prophylaxis is also used herein in its broadest sense to encompass a reduction in the risk of development of a PKC-related condition.
  • an agent may act to treat a subject prophylactically.
  • the prophylactic administration of an agent may result in the agent becoming involved in the treatment of a pathological condition.
  • Use of the terms “treatment” or “prophylaxis” is not to be taken as limiting the intended result which is to reduce the incidence of a PKC-related disorder or condition and/or to ameliorate the symptoms or risk of development of symptoms caused or facilitated by a PKC-related condition.
  • the present invention is particularly directed to the use of one or more macrocyclic diterpenes from a member of the Euphorbiaceae family of plants or botanical or horticultural relatives of such plants.
  • Reference herein to a member of the Euphorbiaceae family includes reference to species from the genera Acalypha, Acidoton, Actinostemon, Adelia, Adenocline, Adenocrepis, Adenophaedra, Adisca, Agrostistachys, Alchornea, Alchorneopsis, Alcinaeanthus, Alcoceria, Aleurites, Amanoa, Andrachne, Angostyles, Anisophyllum, Antidesma, Aphora, Aporosa, Aporosella, Argythamnia, Astrococcus, Astrogyne, Baccanrea, Baliospermum, Bernardia, Beyeriopsis, Bischofia, Blachia, Blumeodondron, Bonania, Bradleia, Brey
  • the most preferred genus and most suitable for the practice of the present invention is the genus Euphorbia.
  • Particularly useful species of this genus include Euphorbia aaron - rossii, Euphorbia abbreviata, Euphorbia acuta, Euphorbia alatocaulis, Euphorbia albicaulis, Euphorbia algomarginata, Euphorbia aliceae, Euphorbia alta, Euphorbia anacampseros, Euphorbia andromedae, Euphorbia angusta, Euphorbia anthonyi, Euphorbia antiguensis, Euphorbia apocynifolia, Euphorbia arabica, Euphorbia ariensis, Euphorbia arizonica, Euphorbia arkansana, Euphorbia arteagae, Euphorbia arundelana, Euphorbia astroites, Euphorbia atrococca, Euphorbia baselicis, Euphorbia batabanensis, Euphorbia berger
  • Synadenium grantii and Synadenium compactum are particularly preferred species of the genus Synadenium.
  • Particularly preferred species of the genus Monadenium include Monadenium lugardae and Monadenium guentheri.
  • a preferred species of the genus Endadenium is Endadenium gossweileni.
  • Euphorbia peplus is particularly useful in the practice of the present invention.
  • Reference herein to “ Euphorbia peplus ” or its abbreviation “ E. peplus ” includes various varieties, strains, lines, hybrids or derivatives of this plant as well as its botanical or horticultural relatives.
  • the present invention may be practiced using a whole Euphorbiaceae plant or parts thereof including sap or seeds or other reproductive material may be used. Generally, for seeds or reproductive material to be used, a plant or plantlet is first required to be propagated.
  • Reference herein to a Euphorbiaceae plant, a Euphorbia species or E. peplus further encompasses genetically modified plants.
  • Genetically modified plants include trangenic plants or plants in which a trait has been removed or where an endogenous gene sequence has been down-regulated, mutated or otherwise altered including the alteration or introduction of genetic material which exhibits a regulatory effect on a particular gene. Consequently, a plant which exhibits a character not naturally present in a Euphorbiaceae plant or a species of Euphorbia or in E. peplus is nevertheless encompassed by the present invention and is included within the scope of the above-mentioned terms.
  • the macrocyclic diterpenes are generally in extracts of the Euphorbiaceae plants.
  • An extract may comprise, therefore, sap or liquid or semi-liquid material exuded from, or present in, leaves, stem, flowers, seeds, bark or between the bark and the stem. Most preferably, the extract is from sap.
  • the extract may comprise liquid or semi-liquid material located in fractions extracted from sap, leaves, stems, flowers, bark or other plant material of the Euphoriaceae plant.
  • plant material may be subject to physical manipulation to disrupt plant fibres and extracellular matrix material and inter- and intra-tissue extracted into a solvent including an aqueous environment. All such sources of the macrocyclic diterpenes are encompassed by the present invention including macrocyclic diterpenes obtained by synthetic routes.
  • the preferred macrocyclic diterpenes are selected from compounds of the ingenane, pepluane and jatrophane families.
  • a compound is stated to be a member of the ingenane, pepulane or jatrophane families on the basis of chemical structure and/or chemical or physical properties.
  • a compound which is a derivative of an ingenane, pepluane or jatrophane is nevertheless encompassed by the present invention through use of the terms “ingenane”, “pepluane” or “jatrophane” since these terms include derivatives, chemical analogues and chemically synthetic forms of these families of compounds.
  • One particularly preferred derivative is an angeloyl derivative of ingenane.
  • the preferred chemical agent of the present invention is one which exhibits an effect on PKC.
  • Such an effect may be a direct activation or inhibition of PKC activity or a direct effect on the levels of PKC enzyme in a cell or exported from a cell.
  • the effect may be transitory or may involve an initial activation of PKC activity or PKC enzyme synthesis or induction of a functional conformation followed by a down-regulation of PKC activity, enzyme levels or formation of a deactivated conformation. Consequently, an effect on PKC is regarded herein as a modulatory effect and is conveniently determined by consequential events such as resulting from altered signal transduction. For example, activation of latent virus, activation of immune mechanisms or activation of a gene promoter may occur and this is regarded herein as a modulatory effect on PKC.
  • the chemical agents of the present invention may be in purified or isolated form meaning that the preparation is substantially devoid of other compounds or contaminating agents other than diluent, solvent or carrier or isoforms of the agents.
  • the term “chemical agent” includes preparations of two or more compounds either admixed together or co-purified from a particular source.
  • the chemical agent may also be a chemical fraction, extract or other preparation from the Euphorbiaceace plant.
  • a “chemical agent” includes a purified form of one or more compounds or a chemical fraction or extract such as from the sap of a Euphorbiaceace plant, and in particular a species of Euphorbia, and most preferably from E. peplus or botanical or horticultural relatives or variants thereof.
  • one aspect of the present invention contemplates a method for the treatment or prophylaxis of a PKC-related condition or disorder in a subject, said method comprising the administration to said subject of a symptom-ameliorating effective amount of a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or chemical analogue thereof which chemical agent is a macrocyclic diterpene selected from compounds of the ingenane, pepluane and jatrophane families and which chemical agent or derivative or chemical analogue is represented by any one of the general formulae (I)-(V)
  • n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
  • A-T are independently selected from hydrogen, R 1 , R 2 , R 3 , F, Cl, Br, I, CN, OR 1 , SR 1 , NR 1 R 2 , N( ⁇ O) 2 , NR 1 OR 2 , ONR 1 R 2 , SOR 1 , SO 2 R 1 , SO 3 R 1 , SONR 1 R 2 , SO 2 NR 1 R 2 , SO 3 NR 1 R 2 , P(R 1 ) 3 , P( ⁇ O)(R 1 ) 3 , Si(R 1 ) 3 , B(R 1 ) 2 , (C ⁇ X)R 3 or X(C ⁇ X)R 3 where X is selected from sulfur, oxygen and nitrogen;
  • R 1 and R 2 are each independently selected from C 1 -C 20 alkyl (branched and/or straight chained), C 1 -C 20 arylalkyl, C 3 -C 8 cycloalkyl, C 6 -C 14 aryl, C 1 -C 14 heteroaryl, C 1 -C 14 heterocycle, C 2 -C 10 alkenyl (branched and/or straight chained), C 2 -C 10 alkynyl (branched and/or straight chained), C 1 -C 10 heteroarylalkyl, C 1 -C 10 alkoxyalkyl, C 1 -C 10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C 1 -C 10 [CN, OR 1 , SR 1 , NR 1 R 2 , N( ⁇ O) 2 , NR 1 OR 2 , ONR 1 R 2 , SOR 1 , SO 2 R 1 , SO 3 R 1
  • R 3 is selected from R 1 , R 2 , CN, COR 1 , CO 2 R 1 , OR 1 , SR 1 , NR 1 R 2 , N( ⁇ O) 2 , NR 1 OR 2 , ONR 1 R 2 , SOR 1 , SO 2 R 1 , SO 3 R 1 , SONR 1 R 2 , SO 2 NR 1 R 2 , SO 3 NR 1 R 2 , P(R 1 ) 3 , P( ⁇ O)(R 1 ) 3 , Si(R 1 ) 3 , B(R 1 ) 2 ;
  • a connected to B (or C), D (or E, R (or Q), P (or O) or S (or T) is a selection of C 1 -C 8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic rings further substituted by R 3 , (C ⁇ X)R 3 and X(C ⁇ X)R 3 , including epoxides and thioepoxides;
  • J connected to I (or H), G (or F), K (or L), M (or N) or S (or T) is a selection of C 1 -C 8 disubstituted (fused) saturated and unsaturated carbocyclic or heterocyclic rings further substituted by R 3 , (C ⁇ X)R 3 and X(C ⁇ X)R 3 , including epoxides and thioepoxides;
  • D (or E) connected to B (or C) or G (or F); I (or H) connected to G (or F); P (or O) connected to R (or Q) or M (or N); K (or L) connected to N (or M) is a selection of C 1 -C 8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic rings substituted by R 3 , (C ⁇ X)R 3 and X(C ⁇ X)R 3 , including epoxides and thioepoxides;
  • B and C, D and E, R and Q, P and O, I and H, G and F, K and L, M and N or S and T are ⁇ X where X is selected from sulfur, oxygen, nitrogen, NR 1 R 2 , and ⁇ CR 1 R 2
  • n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
  • A′-T′ are independently selected from hydrogen, R 4 , R 5 , R 6 , F, Cl, Br, I, CN, COR 4 , CO 2 R 4 , OR 4 , SR 4 , NR 4 R 5 , CONR 4 R 5 , N( ⁇ O) 2 , NR 4 OR 5 , ONR 4 R 5 , SOR 4 , SO 2 R 4 , SO 3 R 4 , SONR 4 R 5 , S0 2 NR4R 5 , S0 3 NR 4 R 5 , P(R 4 ) 3, P( ⁇ O)(R 4 ) 3 , Si(R 4 ) 3 , B(R 4 ) 2 , (C ⁇ X)R 6 or X(C ⁇ X)R 6 where X is selected from sulfur, oxygen and nitrogen;
  • R 4 and R 5 are each independently selected from C 1 -C 20 alkyl (branched and/or straight chained), C 1 -C 20 arylalkyl, C 3 -C 8 cycloalkyl, C 6 -C 14 aryl, C 1 -C 14 heteroaryl, C 1 -C 14 heterocycle, C 2 -C 10 alkenyl (branched and/or straight chained), C 2 -C 10 alkynyl (branched and/or straight chained), C 1 -C 10 heteroarylalkyl, C 1 -C 10 alkoxyalkyl, C 1 -C 10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C 1 -C 10 [CN, OR 4 , SR 4 , NR 4 R 5 , N( ⁇ O) 2 , NR 4 OR 5 , ONR 4 R 5 , SOR 4 , SO 2 R 4 , SO 3 R 4 , C 1
  • R 6 is selected from R 4 , R 5 , CN, COR 4 , CO 2 R 4 , OR 4 , SR 4 , NR 4 R 5 , N( ⁇ O) 2 , NR 4 OR 5 , ONR 4 R 5 , SOR 4 , SO 2 R 4 , SO 3 R 4 , SONR 4 R 5 , SO 2 NR 4 R 5 , SO 3 NR 4 R 5 , P(R 4 ) 3 , P( ⁇ O)(R 4 ) 3 , Si(R 4 ) 3 , B(R 4 ) 2 ;
  • E′ and R′ or H′ and O′ is a C 2 -C 8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R 6 , including epoxides and thioepoxides;
  • A′, B′ and C′, D′ and F′, G′ and M′, N′ and P′, Q′ and S′, T′ are ⁇ X where X is selected from sulfur, oxygen, nitrogen, NR 4 R 5 , (C ⁇ X)R 6 , X(C ⁇ X)R 6 , and ⁇ CR 7 R 8 ;
  • R 7 and R 8 are each independently selected from R 6 , (C ⁇ X)R 6 and X(C ⁇ X)R 6
  • n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
  • a 1 -T 1 are independently selected from hydrogen, R 9 , R 10 , R 11 , F, Cl, Br, I, CN, OR 9 , SR 9 , NR 9 R 10 , N( ⁇ O) 2 , NR 9 OR 10 , ONR 9 R 10 , SOR 9 , SO 2 R 9 , SO 3 R 9 , SONR 9 R 10 , SO 2 NR 9 R 10 , SO 3 NR 9 R 10 , P(R 9 ) 3 , P( ⁇ O)(R 9 ) 3 , Si(R 9 ) 3 , B(R 9 ) 2 , (C ⁇ X)R 11 or X(C ⁇ X)R 11 where X is selected from sulfur, oxygen and nitrogen;
  • R 9 and R 10 are each independently selected from C 1 -C 20 alkyl (branched and straight chained), C 1 -C 20 arylalkyl, C 3 -C 8 cycloalkyl, C 6 -C 14 aryl, C 1 -C 14 heteroaryl, C 1 -C 14 heterocycle, C 2 -C 10 alkenyl (branched and straight chained), C 2 -C 10 alkynyl (branched and straight chained), C 1 -C 10 heteroarylalkyl, C 1 -C 10 alkoxyalkyl, C 1 -C 10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C 1 -C 10 [CN, OR 9 , SR 9 , NR 9 R 10 , N( ⁇ O) 2 , NR 9 OR 10 , ONR 9 R 10 , SOR 9 , SO 2 R 9 , SO 3 R 9 , SONR 9 R
  • R 11 is selected from R 9 , R 10 , CN, COR 9 , CO 2 R 9 , OR 9 , SR 9 , NR 9 R 10 , N( ⁇ O) 2 , NR 9 OR 10 , ONR 9 R 10 , SOR 9 , SO 2 R 9 , SO 3 R 9 , SONR 9 R 10 , SO 2 NR 9 R 10 , SO 3 NR 9 R 10 , P(R 9 ) 3 , P( ⁇ O)(R 9 ) 3 , Si(R 9 ) 3 , B(R 9 ) 2 ;
  • B 1 and R 1 , E 1 and ⁇ 1 and ⁇ 1 and M 1 are selected from a C 2 -C 8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R 11 , including epoxides and thioepoxides;
  • a 1 , ⁇ and ⁇ , ⁇ and C 1 , D 1 and F 1 , É and G 1 , H 1 and I 1 , J 1 and K 1 , L 1 and N 1 , O 1 and P 1 , Q 1 and S 1 , T 1 are ⁇ X where X is selected from sulfur, oxygen, nitrogen, NR 9 R 10 , including (C ⁇ X)R 11 and X(C ⁇ X)R 11 , and ⁇ CR 12 R 13 ;
  • R 12 and R 13 are independently selected from R 11 , (C ⁇ X)R 11 and X(C ⁇ X)R 11
  • n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
  • a 2 -X 2 are independently selected from hydrogen, R 14 , R 15 , R 16 , F, Cl, Br, I, CN, OR 14 , SR 14 , NR 14 R 15 , N( ⁇ O) 2 , NR 14 OR 15 , ONR 14 R 15 , SOR 14 , SO 2 R 14 , SO 3 R 14 , SONR 14 R 15 , SO 2 NR 14 R 15 , SO 3 NR 14 R 15 , P(R 14 ) 3 , P( ⁇ O)(R 14 ) 3 , Si(R 14 ) 3 , B(R 14 ), (C ⁇ Y)R 16 or Y(C ⁇ Y)R 16 where Y is selected from sulfur, oxygen and nitrogen;
  • R 14 and R 15 are each independently selected from C 1 -C 20 alkyl (branched and/or straight chained), C 1 -C 20 arylalkyl, C 3 -C 8 cycloalkyl, C 6 -C 14 aryl, C 1 -C 14 heteroaryl, C 1 -C 14 heterocycle, C 2 -C 10 alkenyl (branched and/or straight chained), C 2 -C 10 alkynyl (branched and/or straight chained), C 1 -C 10 heteroarylalkyl, C 1 -C 10 alkoxyalkyl, C 1 -C 10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C 1 -C 10 [CN, OR 14 , SR 14 , NR 14 R 10 , N( ⁇ O) 2 , NR 14 OR 15 , ONR 14 R 15 , SOR 14 , SO 2 R 14 , SO 3 R 14 , C 1
  • R 16 is selected from R 14 , R 15 , CN, COR 14 , CO 2 R 15 , OR 14 , SR 14 , NR 14 R 15 , N( ⁇ O) 2 , NR 14 OR 15 , ONR 14 R 15 , SOR 14 , SO 2 R 14 , SO 3 R 14 , SONR 14 R 15 , SO 2 NR 14 R 15 , SO 3 NR 14 R 15 , P(R 14 ) 3 , P( ⁇ O)(R 14 ) 3 , Si(R 14 ) 3 , B(R 14 ) 2 ;
  • E 2 and V 2 , H 2 and S 2 , and I 2 and P 2 are C 2 -C 8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R 16 , including epoxides and thioepoxides;
  • a 2 , B 2 ; C 2 , D 2 ; F 2 , G 2 ; J 2 , K 2 ; L 2 , M 2 ; N 2 , O 2 ; Q 2 , R 2 ; U 2 , T 2 and X 2 , W 2 are ⁇ Y where Y is selected from sulfur, oxygen, nitrogen, NR 14 R 15 and ⁇ CR 17 R 18 ;
  • R 17 and R 18 are independently selected from R 16 , (C ⁇ Y)R 16 and Y(C ⁇ Y)R 16
  • n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
  • a 3 -Z 3 are independently selected from hydrogen, R 19 , R 20 , R 21 , F, Cl, Br, I, CN, OR 19 , SR 19 , NR 19 R 20 , N( ⁇ O) 2 , NR 19 OR 20 , ONR 19 R 20 , SOR 19 , SO 2 R 19 , SO 3 R 19 , SONR 19 R 20 , SO 2 NR 19 R 20 , SO 3 NR 19 R 20 , P(R, 9 ) 3 , P(O)(R 19 ) 3 , Si(R 19 ) 3 , B(R 19 ) 2 , (C ⁇ )R 21 or ⁇ (C ⁇ )R 21 where ⁇ is sullur, oxygen and nitrogen;
  • R 19 and R 20 are each independently selected from C 1 -C 20 alkyl (branched and/or straight chained), C 1 -C 20 arylalkyl, C 3 -C 8 cycloalkyl, C 6 -C 14 aryl, C 1 -C 14 heteroaryl, C 1 -C 14 heterocycle, C 2 -C 10 alkenyl (branched and/or straight chained), C 2 -C 10 alkynyl (branched and/or straight chained), C 1 -C 10 heteroarylalkyl, C 1 -C 10 alkoxyalkyl, C 1 -C 10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C 1 -C 10 [CN, OR 19 , SR 19 , NR 19 R 20 , N( ⁇ O) 2 , NR 19 OR 20 , ONR 19 R 20 , SOR 19 , SO 2 R 19 , SO 3 R 19
  • R 21 is selected from R 19 , R 20 , CN, COR 19 , CO 2 R 19 , OR 19 , SR 19 , NR 19 R 20 , N( ⁇ O) 2 , NR 19 OR 20 , ONR 19 R 20 , SOR 19 , SO 2 R 19 , SO 3 R 19 , SONR 19 R 20 , SO 2 NR 19 R 20 , SO 3 NR 19 R 20 , P(R 19 ) 3 , P( ⁇ O)(R 19 ) 3 , Si(R 19 ) 3 , B(R 19 ) 2 ;
  • D 3 connected to X 3 is a C 2 -C 8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R 21 , including epoxides and thioepoxides;
  • W 3 (or V 3 ) connected to U 3 (or T 3 ) or X 3 ; X 3 connected to Y 3 (or Z 3 ) are C 1 -C 8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R 21 , (C ⁇ )R 21 and ⁇ (C ⁇ )R 21 , including epoxides and thioepoxides;
  • a 3 , ⁇ 3 ; B 3 , C 3 ; E 3 , F 3 ; G 3 , H 3 ; I 3 , J 3 ; K 3 , L 3 ; M 3 , N 3 ; O 3 , ⁇ 3 ; Q 3 , P 3 , S 3 , R 3 , U 3 , T 3 , W 3 , V 3 , and Z 3 , Y 3 are ⁇ where ⁇ is selected from sulfur, oxygen, nitrogen, NR 19 R 20 , and ⁇ CR 22 R 23 ; and
  • R 22 and R 23 are selected from R 21 , (C ⁇ )R 21 and ⁇ (C ⁇ )R 21 ;
  • R 24 , R 25 and R 26 are independently selected from hydrogen, R 27 , R 28 , F, Cl, Br, I, CN, OR 27 , SR 27 , NR 27 R 28 , N( ⁇ O) 2 , NR 27 OR 28 , ONR 27 R 28 , SOR 27 , SO 2 R 27 , SO 3 R 27 , SONR 27 R 28 , SO 2 NR 27 R 28 , SO 3 NR 27 R 28 , P(R 27 ) 3 , P( ⁇ O)(R 27 ) 3 , Si(R 27 ) 3 , B(R 27 ) 2 , (C ⁇ X)R 29 or X(C ⁇ X)R 29 where X is selected from sulfur, oxygen and nitrogen;
  • R 27 and R 28 are each independently selected from C 1 -C 20 alkyl (branched and/or straight chained), C 1 -C 20 arylalkyl, C 3 -C 8 cycloalkyl, C 6 -C 14 aryl, C 1 -C 14 heteroaryl, C 1 -C 14 heterocycle, C 2 -C 10 alkenyl (branched and/or straight chained), C 2 -C 10 alkynyl (branched and/or straight chained), C 1 -C 10 heteroarylalkyl, C 1 -C 10 alkoxyalkyl, C 1 -C 10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C 1 -C 10 [CN, OR 27 , SR 27 , NR 27 R 28 , N( ⁇ O) 2 , NR 27 OR 28 , ONR 27 R 28 , SOR 27 , SO 2 R 27 , SO 3 R 27 , C 1
  • R 29 is selected from R 27 , R 28 , CN, COR 27 , CO 2 R 27 , OR 27 , SR 27 , NR 27 R 28 , N( ⁇ O) 2 , NR 27 OR 28 , ONR 27 R 28 , SOR 27 , S0 2 R 27 , SO 3 R 27 , SONR 27 R 28 , SO 2 NR 27 R 28 , SO 3 NR 27 R 28 , P(R 27 ) 3 , P( ⁇ O)(R 27 ) 3 , Si(R 27 ) 3 , B(R 27 ) 2 .
  • R 24 is hydrogen, OAcetyl or OH.
  • R 25 and R 26 are OH.
  • alkyl refers to linear or branched chains.
  • haloalkyl refers to an alkyl group substituted by at least one halogen.
  • haloalkoxy refers to an alkoxy group substituted by at least one halogen.
  • halogen refers to fluorine, chlorine, bromine and iodine.
  • aryl refers to aromatic carbocyclic ring systems such as phenyl or naphthyl, anthracenyl, especially phenyl;
  • aryl is C 6 -C 14 with mono, di- and tri- substitution containing F, Cl, Br, I, NO 2 , CF 3 , CN, OR 1 , COR 1 , CO 2 R 1 , NHR 1 , NR 1 R 2 , NR 1 OR 2 , ONR 1 R 2 , SOR 1 , SO 2 R 1 , SO 3 R 1 , SONR 1 R 2 , SO 2 NR 1 R 2 , SO 3 NR 1 R 2 , P(R 1 ) 3 , P( ⁇ O)(R 1 ) 3 , Si(R 1 ) 3 , B(R 1 ) 2 , wherein R 1 and R 2 are defined above
  • heterocycle refers to a saturated, unsaturated, or aromatic carbocyclic group having a single ring, multiple fused rings (for example, bicyclic, tricyclic, or other similar bridged ring systems or substituents), or multiple condensed rings, and having at least one heteroatom such as nitrogen, oxygen, or sulfur within at least one of the rings.
  • This term also includes “heteroaryl” which refers to a heterocycle in which at least one ring is aromatic. Any heterocyclic or heteroaryl group can be unsubstituted or optionally substituted with one or more groups, as defined above.
  • bi- or tricyclic heteroaryl moieties may comprise at least one ring, which is either completely, or partially, saturated.
  • Suitable heteroaryl moieties include, but are not limited to oxazolyl, thiazaoyl, thienyl, furyl, 1-isobenzofuranyl, 3H-pyrrolyl, 2H-pyrrolyl, N-pyrrolyl, imidazolyl, pyrazolyl, isothiazolyl, isooxazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyradazinyl, indolizinyl, isoindolyl, indoyl, indolyl, purinyl, phthalazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,3-oxadiazoyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-o
  • Preferred PKC-related conditions include alcoholism, Alzheimer's disease, asthma, atherosclosis, atopic dermatitis, autoimmune disease, bipolar-disorder, blood disorder, cardiac hypertrophy, depression, diabetes, hypertension, hyperplastic dermatosis, multiple sclerosis, myocardial ischemia, osteoarthritis, psoriasis, rheumatoid arthritis, transplantation and a latent virus.
  • the instant chemical agents are also useful in promoting gene activation and promotion.
  • the subject chemical agents are also useful in modulating and more particularly promoting or activating gene expression or the expression or operation of a promoter or other regulatory sequence.
  • This aspect is useful for gene therapy, gene augmentation or gene replacement therapy or the provision of expression products (e.g. peptides, polypeptides, proteins, antisense RNA, sense RNA) by endogenous genes or exogenously introduced DNA or RNA.
  • the subject compounds are also useful in activating latent viruses.
  • viruses is used in its broadest sense to include viruses of the families adenoviruses, papovaviruses, herpesviruses: simplex, varicella-zoster, Epstein-Barr, CMV, pox viruses: smallpox, vaccinia, hepatitis B, rhinoviruses, hepatitis A, poliovirus, rubellavirus, hepatitis C, arboviruses, rabiesvirus, influenzaviruses A and B, measlesvirus, mumpsvirus, HIV, HTLV I and II.
  • viruses are HIV, EBV and CMV.
  • another aspect of the present invention provide a method for the treatment or prophylaxis of a PKC-related condition or disorder in a subject, said method comprising the administration to said subject of a symptom-ameliorating effective amount of a macrocyclic diterpene, or a chemical fraction comprising same from a plant of the family Euphorbiaceae or a derivative or chemical analogue of said macrocyclic diterpene having the structures as defined above wherein said macrocyclic diterpene or its derivative or chemical analogue modulates PKC activity, synthesis or enzyme turnover, said administration being for a time and under conditions sufficient to ameliorate one or more symptoms of associated with a PKC-related condition or disorder.
  • This aspect of the present invention is particularly useful in the treatment of latent virus infection.
  • latent virus includes reference to a virus or more particularly a virus genome or part thereof which has integrated into the genome of a cell.
  • the subject chemical agents are capable of activating a latent virus thereby causing the virus to undergo replication and at least partial assembly.
  • a mechanism within the host or within the cells of the host is then induced to assist in the eradication of the virus.
  • the present invention extends to both the direct effect of the chemical agent on the virus as well as promoting the immune system to direct same against the virus.
  • another aspect of the present invention contemplates a method for the treatment or prophylaxis of a PKC-related condition or disorder relating to a latent virus in a subject, said method comprising the administration to said subject of a virus-activating effective amount of a macrocyclic diterpene, or a chemical fraction comprising same from a plant of the family Euphorbiaceae or a derivative or chemical analogue of said macrocyclic diterpene having the structures as defined above wherein said macrocyclic diterpene or its derivative or chemical analogue modulates PKC activity, synthesis or enzyme turnover, said administration being for a time and under conditions sufficient to activate said virus and to combination therapies with antiviral agents.
  • the activated virus is then destroyed or removed by the host's own immune system and/or by the effects of the agents themselves.
  • the method further comprises the simultaneous, sequential or separate administration of an ancillary agent that destroys or attenuates a replicating virus, in combination with the macrocyclic diterpene or chemical fraction.
  • an ancillary agent that destroys or attenuates a replicating virus
  • the invention contemplates combination therapies in which the macrocyclic diterpene or chemical fraction activates a latent virus to thereby cause the virus to undergo replication and the ancillary agent eradicates the replicating virus.
  • the invention encompasses a method for the treatment or prophylaxis of a PKC-related condition or disorder relating to a latent virus in a subject, said method comprising simultaneously, sequentially or separately administrating to said subject a virus-activating effective amount of a macrocyclic diterpene, or a chemical fraction comprising same from a plant of the family Euphorbiaceae or a derivative or chemical analogue of said macrocyclic diterpene having the structures as defined above wherein said macrocyclic diterpene or its derivative or chemical analogue modulates PKC activity, synthesis or enzyme turnover, together with a virus-destroying or -attenuating effective amount of an ancillary agent which destroys or attenuates a replicating form of said virus, said administration being for a time and under conditions sufficient for said macrocyclic diterpene or chemical fraction to activate said virus to thereby cause the virus to undergo replication and for said ancillary agent to destroy or attenuate said replicating virus.
  • the latent virus is HIV.
  • a range of HIV specific agents may be used for the destruction or attenuation of this virus, including, for example, those described in Matsuhita et al., 2000.
  • Preferred HIV specific ancillary agents include, for example, nucleoside analogues such as combivir, epivir, hivid, retrovir, videx, zerat and zygen, non-nucleoside agents such as rescriptor, sustiva and viramune, adjunctive anti-retrovirals such as hydrea and droxa, and protease inhibitors such as agenerase, fortovase, crixivan, invirase, norvir and virasept.
  • the latent virus is EBV.
  • EBV specific ancillary agents are selected from ganciclovir (GVC) or 3′-azido-3′deoxythymidine (AZT).
  • the latent virus is CMV.
  • a preferred CMV specific ancillary agent is cidofovir.
  • Particularly useful compounds include 5,8,9,10,14-pentaacetoxy-3-benzoyloxy- 15-hydroxypepluane (pepluane), derivatives of said pepluane, jatrophanes of Conformation II including 2,3,5,7,15-pentaacetoxy-9-nicotinoyloxy-14-oxojatropha-6(17),11E-diene kjatrophane 1), derivatives of said jatrophane 1,2,5,7,8,9,14-hexaacetoxy-3-benzoyloxy-15-hydroxy-jatropha-6(17),11E-diene (jatrophane 2), derivatives of said jatrophane 2, 2,5,14-triacetoxy-3-benzoyloxy-8,15-dihydroxy-7-isobutyroyloxy-9-nicotinoyloxy-jatropha-6(17),11E-diene (jatrophane 3), derivatives of said jatrophane 3, 2,5,9,14-tetraacetoxy
  • angeloyl substituted ingenanes or derivatives thereof such as ingenol-3-angelate, 20-hydroxy-ingenol-3-angelate, 20-O-acetyl-ingenol-3-angelate, or derivatives of said angelates, or pharmaceutically acceptable salts of these.
  • Still a further aspect of the present invention contemplates a method of assessing the suitability of a chemical agent from Euphorbiaceae for the practice of the present invention.
  • Numerical values are assigned to chemical agents including fractions comprising the chemical agents as set forth, for example, in Table A: TABLE A Feature Value An ability to modulate PKC activity or effect +1 An ability to induce bipolar dendritic activity +1 An ability to displace phorbol dibutyrate from +1 binding to PKC An ability to induce respiratory burst in leucocytes +1 An ability to stimulate phagocytosis in peripheral +1 blood mononuclear cells Derived from a member of the Euphorbiaeae family +1 Derived from E. peplus +3 Water extractible from the sap of Euphorbia sp. +2 An ability to activate latent virus +4 A lower tumor promotion activity than TPA/PMA +2
  • the value for each feature is referred to as the Index Value (I V ).
  • Preferred compounds are selected from the list comprising:
  • ingenol-3-angelate derivatives of ingenol-3-angelate.
  • the biological entity is a microorganism, virus, yeast, fungus, insect, arachnid or Plasmodium sp.
  • Reference herein to a subject includes a human, primate, livestock animal (e.g. sheep, cow, horse, pig, goat, donkey), laboratory test animal (e.g. mouse, rat, guinea pig, hamster), companion animal (e.g. dog, cat) or avian species such as poultry birds (e.g. chicken, ducks, turkeys, geese) or game birds (e.g. arid ducks, pheasants).
  • livestock animal e.g. sheep, cow, horse, pig, goat, donkey
  • laboratory test animal e.g. mouse, rat, guinea pig, hamster
  • companion animal e.g. dog, cat
  • avian species such as poultry birds (e.g. chicken, ducks, turkeys, geese) or game birds (e.g. arid ducks, pheasants).
  • the preferred subject is a human or primate or laboratory test animal.
  • the most preferred subject is a human.
  • the assessment of the suitability of a compound or group of compounds for the practice of the present invention is suitably facilitated with the assistance of a computer programmed with software, which inter alia adds index values (I V ) for at least two features associated with the compound(s) to provide a potency value (P A ) corresponding to the effectiveness of the compound(s) for treating or preventing infection or colonization or presence of a biological entity in a subject.
  • the compound features can be selected from:
  • index values for such features are stored in a machine-readable storage medium, which is capable of processing the data to provide a potency value for a compound or group of compounds of interest.
  • the invention contemplates a computer program product for assessing the likely usefulness of a candidate compound or group of compounds for treating or preventing a PKC-related condition or disorder in a subject, said product comprising:
  • the computer program product comprises code that assigns an index value for each feature of a compound or group of compounds.
  • index values are assigned as set forth in Table A above.
  • the invention extends to a computer for assessing the likely usefulness of a candidate compound or group of compounds for treating or preventing a PKC-related condition or disorder in a subject, wherein said computer comprises:
  • a machine-readable data storage medium comprising a data storage material encoded with machine-readable data, wherein said machine-readable data comprise index values for at least two features associated with said compound(s), wherein said features are selected from:
  • a central-processing unit coupled to said working memory and to said machine-readable data storage medium, for processing said machine readable data to provide a sum of said index values corresponding to a potency value for said compound(s);
  • FIG. 8 shows a system 10 including a computer 11 comprising a central processing unit (“CPU”) 20 , a working memory 22 which may be, e.g. RAM (random-access memory) or “core” memory, mass storage memory 24 (such as one or more disk drives or CD-ROM drives), one or more cathode-ray tube (“CRT”) display terminals 26 , one or more keyboards 28 , one or more input lines 30 , and one or more output lines 40 , all of which are interconnected by a conventional bidirectional system bus 50 .
  • CPU central processing unit
  • working memory 22 which may be, e.g. RAM (random-access memory) or “core” memory
  • mass storage memory 24 such as one or more disk drives or CD-ROM drives
  • CRT cathode-ray tube
  • Input hardware 36 coupled to computer 11 by input lines 30 , may be implemented in a variety of ways.
  • machine-readable data of this invention may be inputted via the use of a modem or modems 32 connected by a telephone line or dedicated data line 34 .
  • the input hardware 36 may comprise CD.
  • ROM drives or disk drives 24 in conjunction with display terminal 26 , keyboard 28 may also be used as an input device.
  • Output hardware 46 coupled to computer 11 by output lines 40 , may similarly be implemented by conventional devices.
  • output hardware 46 may include CRT display terminal 26 for displaying a synthetic polynucleotide sequence or a synthetic polypeptide sequence as described herein.
  • Output hardware might also include a printer 42 , so that hard copy output may be produced, or a disk drive 24 , to store system output for later use.
  • CPU 20 coordinates the use of the various input and output devices 36 , 46 coordinates data accesses from mass storage 24 and accesses to and from working memory 22 , and determines the sequence of data processing steps.
  • a number of programs may be used to process the machine readable data of this invention. Exemplary programs may use for example the following steps:
  • FIG. 9 shows a cross section of a magnetic data storage medium 100 which can be encoded with machine readable data, or set of instructions, for designing a synthetic molecule of the invention, which can be carried out by a system such as system 10 of FIG. 10.
  • Medium 100 can be a conventional floppy diskette or hard disk, having a suitable substrate 101 , which may be conventional, and a suitable coating 102 , which may be conventional, on one or both sides, containing magnetic domains (not visible) whose polarity or orientation can be altered magnetically.
  • Medium 100 may also have an opening (not shown) for receiving the spindle of a disk drive or other data storage device 24 .
  • the magnetic domains of coating 102 of medium 100 are polarized or oriented so as to encode in manner which may be conventional, machine readable data such as that described herein, for execution by a system such as system 10 of FIG. 8.
  • FIG. 10 shows a cross section of an optically readable data storage medium 110 which also can be encoded with such a machine-readable data, or set of instructions, for designing a synthetic molecule of the invention, which can be carried out by a system such as system 10 of FIG. 8.
  • Medium 110 can be a conventional compact disk read only memory (CD-ROM) or a rewritable medium such as a magneto-optical disk, which is optically readable and magneto-optically writable.
  • Medium 100 preferably has a suitable substrate 111 , which may be conventional, and a suitable coating 112 , which may be conventional, usually of one side of substrate 111 .
  • coating 112 is reflective and is impressed with a plurality of pits 113 to encode the machine-readable data.
  • the arrangement of pits is read by reflecting laser light off the surface of coating 112 .
  • a protective coating 114 which preferably is substantially transparent, is provided on top of coating 112 .
  • coating 112 has no pits 113 , but has a plurality of magnetic domains whose polarity or orientation can be changed magnetically when heated above a certain temperature, as by a laser (not shown).
  • the orientation of the domains can be read by measuring the polarisation of laser light reflected from coating 112 .
  • the arrangement of the domains encodes the data as described above.
  • the present invention further extends to pharmaceutical compositions useful in treating a PKC-related disorder.
  • the chemical agents of the invention can be used as actives for the treatment or prophylaxis of a condition associated with the presence of a PKC-related disorder in a subject.
  • the chemical agents can be administered to a patient either by themselves, or in pharmaceutical compositions where they are mixed with a suitable pharmaceutically acceptable carrier.
  • the invention also provides a composition for treatment and/or prophylaxis of a PKC-related condition or disorder in a subject, comprising one or more chemical agents of the invention, together with a pharmaceutically acceptable carrier and/or diluent.
  • chemical agents may be formulated and administered systemically or locally. Techniques for formulation and administration may be found in “Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, Pa., latest edition. Suitable routes may, for example, include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections.
  • the chemical agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. Intra-muscular and subcutaneous injection is appropriate, for example, for administration of immunomodulatory compositions and vaccines.
  • the chemical agents can be formulated readily using pharmaceutically acceptable carriers well known in the art into dosages suitable for oral administration.
  • Such carriers enable the compounds of the invention to be formulated in dosage forms such as tablets, pills, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • These carriers may be selected from sugars, starches, cellulose and its derivatives, malt, gelatine, talc, calcium sulphate, vegetable oils, synthetic oils, polyols, alginic acid, phosphate buffered solutions, emulsifiers, isotonic saline, and pyrogen-free water.
  • compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve their intended purpose.
  • the dose of agent administered to a patient should be sufficient to effect a beneficial response in the patient over time such as a reduction in the symptoms associated with the presence of a biological entity or part thereof or toxin or venom therefrom or a genetic event caused thereby in a subject.
  • the quantity of the agent(s) to be administered may depend on the subject to be treated inclusive of the age, sex, weight and general health condition thereof. In this regard, precise amounts of the agent(s) for administration will depend on the judgement of the practitioner.
  • the physician may evaluate tissue or fluid levels of the biological entity, and progression of the disorder.
  • tissue or fluid levels of the biological entity may be evaluated.
  • those of skill in the art may readily determine suitable dosages of the chemical agents of the invention.
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • compositions for oral use can be obtained by combining the active compounds with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinyl-pyrrolidone (PVP).
  • PVP polyvinyl-pyrrolidone
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Such compositions may be prepared by any of the methods of pharmacy but all methods include the step of bringing into association one or more chemical agents as described above with the carrier which constitutes one or more necessary ingredients.
  • the pharmaceutical compositions of the present invention may be manufactured in a manner that is itself known, e.g. by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilising processes.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added.
  • Dosage forms of the chemical agents of the invention may also include injecting or implanting controlled releasing devices designed specifically for this purpose or other forms of implants modified to act additionally in this fashion.
  • Controlled release of an agent of the invention may be effected by coating the same, for example, with hydrophobic polymers including acrylic resins, waxes, higher aliphatic alcohols, polylactic and polyglycolic acids and certain cellulose derivatives such as hydroxypropyhnethyl cellulose.
  • controlled release may be effected by using other polymer matrices, liposomes and/or microspheres.
  • Chemical agents of the invention may be provided as salts with pharmaceutically compatible counterions.
  • Pharmaceutically compatible salts may be formed with many acids, including but not limited to hydrochloric, sulphuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents that are the corresponding free base forms.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC50 as determined in cell culture (e.g. the concentration of a test agent, which achieves a half-maximal inhibition of infection or colonization or presence of a biological entity). Such information can be used to more accurately determine useful doses in humans.
  • Toxicity and therapeutic efficacy of such chemical agents can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g. for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50.
  • Compounds that exhibit large therapeutic indices are preferred.
  • the data obtained from these cell culture assays and animal studies can be used in formulating a range of dosages for use in humans.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition (see for example Fingl et al., 1975, in “The Pharmacological Basis of Therapeutics”, Ch. 1 p1).
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active agent which are sufficient to maintain symptom-ameliorating effects.
  • Usual patient dosages for systemic administration range from 1-2000 mg/day, commonly from 1-250 mg/day, and typically from 10-150 mg/day. Stated in terms of patient body weight, usual dosages range from 0.02-25 mg/kg/day, commonly from 0.02-3 mg/kg/day, typically from 0.2-1.5 mg/kg/day. Stated in terms of patient body surface areas, usual dosages range from 0.5-1200 mg/m 2 /day, commonly from 0.5-150 mg/m 2 /day, typically from 5-100 mg/m 2 /day.
  • the liposomes will be targeted to and taken up selectively by the tissue.
  • the effective local concentration of the agent may not be related to plasma concentration.
  • compositions containing between 0.001-5% or more chemical agent are generally suitable. Regions for topical administration include the skin surface and also mucous membrane tissues of the vagina, rectum, nose, mouth, and throat. Compositions for topical administration via the skin and mucous membranes should not give rise to signs of irritation, such as swelling or redness.
  • the topical composition may include a pharmaceutically acceptable carrier adapted for topical administration.
  • the composition may take the form of a suspension, solution, ointment, lotion, sexual lubricant, cream, foam, aerosol, spray, suppository, implant, inhalant, tablet, capsule, dry powder, syrup, balm or lozenge, for example. Methods for preparing such compositions are well known in the pharmaceutical industry.
  • the topical composition is administered topically to a subject, e.g. by the direct laying on or spreading of the composition on the epidermal or epithelial tissue of the subject, or transdermally via a “patch”.
  • compositions include, for example, lotions, creams, solutions, gels and solids.
  • Suitable carriers for topical administration preferably remain in place on the skin as a continuous film, and resist being removed by perspiration or immersion in water.
  • the carrier is organic in nature and capable of having dispersed or dissolved therein a chemical agent of the invention.
  • the carrier may include pharmaceutically-acceptable emolients, emulsifiers, thickening agents, solvents and the like.
  • the invention also features a process for separating macrocyclic diterpenes from a biomass containing same, said process comprising contacting the biomass with an aqueous solvent for a time and under conditions sufficient to extract the macrocyclic diterpenes into said solvent.
  • the aqueous solvent is preferably water.
  • the biomass is derived from a plant, which is preferably a member of the Euphorbiaceae family of plants or botanical or horticultural relatives of such plants. Matter from the plant (e.g. foliage, stems, roots, seeds, bark, etc.) is preferably cut, macerated or mulched to increase the surface area of the plant matter for aqueous extraction of the macrocyclic diterpenes.
  • the process preferably further comprises adsorbing the macrocyclic diterpenes to a non-ionic adsorbent, which is suitably a non-ionic porous synthetic adsorbent.
  • a non-ionic porous synthetic adsorbent that can be used for the purposes of the present invention include, but are not restricted to, aromatic copolymers mainly composed of styrene and divinylbenzene, and methacrylic copolymers mainly composed of monomethacrylate and dimethacrylate.
  • non-ionic porous synthetic adsorbents which comprise, as the basic structure, aromatic copolymers mainly composed of styrene and divinylbenzene include, for example, Diaion HP10, HP20, HP21, HP30, HP40, HP50, SP850, and SP205 (trade names: Mitsubishi Chemical Corp.), and Amberlite XAD-2, XAD4, (trade names: Rohm and Haas Co.).
  • non-ionic porous synthetic adsorbent which comprise, as the basic structure, methacrylic copolymer mainly composed of monomethacrylate and dimethacrylate are Diaion HP2MG, Amberlite XAD-7, XAD-8 and XAD-16 and others.
  • the process further comprises eluting macrocyclic diterpenes from the non-ionic adsorbent with water and water-soluble organic solvent(s).
  • the treatment may be conducted by a batch method using water and water-soluble organic solvent(s) which dissolve macrocyclic diterpenes, or may also be conducted continuously or in batch using a column chromatography method.
  • Examples of a water-soluble organic solvent which may be used in the present invention are alcohols such as methanol, ethanol, n-propyl alcohol, isopropyl alcohol, and tert-butanol, ethers such as dioxane and tetrahydrofuran, ketones such as acetone, amides such as dimethylformamide, sulfur-containing compounds such as dimethylsulfoxide. Two or more of such organic solvents may be mixed for use.
  • solvents less soluble in water for example, alcohols such as n-butanol, esters such as methyl formate and methyl acetate, and ketones such as methyl ethyl ketone may also be used to the extent that it does not separate during development.
  • Particularly preferred water-soluble organic solvents are alcohols, in particular, methanol, ethanol, propyl alcohol, and the like.
  • different kinds of solvent may also be used sequentially for development.
  • Macrocyclic diterpenes can be further purified using media and techniques which separate compounds on the basis of molecular size and/or polarity.
  • the macrocyclic diterpenes are separated using Sephadex HL-20 resin and preferably using water and water-soluble organic solvent(s) for development.
  • the present invention contemplates the use of the chemical agents of the invention for modulating the expression of genes under the control of promoters whose activity is modulated by said chemical agents.
  • the chemical agents of the present invention find particular utility in modulating and preferably in promoting or activating the expression of gene therapy reagents (e.g. peptides, polypeptides, proteins, antisense RNA, sense RNA) under the control of such modulatable promoters.
  • gene therapy refers generally to the therapeutic replacement, augmenting or silencing of an endogenous gene activity, which is typically effected by gene transfer.
  • Gene transfer or “gene delivery” as used herein refer to methods or systems for reliably inserting exogenous nucleic acids, typically DNA, into host cells. Such methods can result in transient expression of non-integrated transferred nucleic acids, extrachromosomal replication and expression of transferred replicons (e.g. episomes), or integration of transferred genetic material into the genomic DNA of host cells.
  • transferred replicons e.g. episomes
  • a number of systems have been developed for gene transfer into mammalian cells. See, e.g., U.S. Pat. No. 5,399,346.
  • exogenous means any nucleic acid that is introduced into an organism or cell.
  • Promoters suitable for the present invention include viral and non-viral promoter, which are preferably inducible.
  • Non-limiting examples of inducible non-viral promoters include metallothionein promoters, and metallothionein-related promoters.
  • Non-limiting examples of inducible viral promoters effective for use in gene transfer vectors of the present invention include, but are not limited to, mouse mammary tumour virus promoters, CMV (cytomegalovirus), SV40 (simian virus 40), RSV (Rous sarcoma virus), HSV (herpes simplex virus), EBV (Epstein-Barr virus), retroviral promoters and adenoviral promoters.
  • vector any genetic element, such as a plasmid, phage, transposon, cosmid, chromosome, virus, virion, etc., which is capable of replication when associated with the proper control elements and which can transfer gene sequences between cells.
  • vector includes cloning and expression vehicles, as well as viral vectors.
  • Preferred gene therapy vectors of the present invention are viral vectors.
  • a viral-based vector comprises: (1) nucleic acid of, or corresponding to at least a portion of, the genome of a virus, which portion is capable of directing the expression of a DNA sequence; and (2) a DNA sequence encoding an expression product (e.g. polypeptide or transcript), operably linked to the viral nucleic acid and capable of being expressed as a functional gene product in the target cell.
  • the recombinant viral vectors of the present invention may be derived from a variety of viral nucleic acids known to one skilled in the art, e.g. the genomes of adenovirus, adeno-associated virus, HSV, alphavirus, vaccinia virus, and other viruses, including RNA and DNA viruses.
  • Particularly preferred viral vectors are those that can accept foreign genetic material and can infect an extended range of cells.
  • Adenoviruses do not integrate their genetic material into the host genome and therefore do not require host replication for gene expression, making them ideally suited for rapid, efficient, heterologous gene expression.
  • Techniques for preparing replication-defective infective viruses are well known in the art.
  • a viral delivery system including the virion is free of undesirable contaminants, such as defective interfering viral particles or endotoxins and other pyrogens such that it will not cause any inappropriate reactions in the cell, animal or individual receiving the adenoviral vector construct.
  • a preferred means of purifying the vector involves the use of buoyant density gradients, such as cesium chloride gradient centrifugation.
  • Adenovirus is a particularly preferred gene therapy vector
  • Particularly preferred Adenoviral vector of the present invention include commercially available retroviral expression vectors including but not limited to pLAPSN, pLEGFP-C1, pLEGFP-N1, pLHCX, pLIB, pLNCX, pLNCX2, pLNHX, pLPCX, pLXIN, pLXRN, pLXSN, pMSCVhyg, pMSCVneo, pMSCVpuro, pSIR, and pVSV-G.
  • the present invention further contemplates the use of Adeno-associated virus (AAV) is vector system for use in the present invention. Details concerning the generation and use of recombinant AAV vectors are described in U.S. Pat. Nos. 5,139,941 and 4,797,368, each incorporated herein by reference.
  • the present invention further contemplates gene therapy using retroviral vectors such as an engineered variant of the Moloney murine leukemia virus.
  • the viral vector may be derived from other viral vectors such as vaccinia virus, Sindbis virus, cytomegalovirus and herpes simplex virus.
  • the present invention further contemplates the use of gene therapy vectors derived from an RNA virus such as but not limited to the following: members of the family Reoviridae, including the genus Orthoreovirus (multiple serotypes of both mammalian and avian retroviruses), the genus Orbivirus (Bluetongue virus, Eugenangee virus, Kemerovo virus, African horse sickness virus, and Colorado Tick Fever virus), the genus Rotavirus (human rotavirus, Kansas calf diarrhea virus, murine rotavirus, simian rotavirus, bovine or ovine rotavirus, avian rotavirus); the family Retrovirideae including the genus Lentivirus (Human Immunodeficiency Virus), the family Picomaviridae, including the genus Enterovirus (poliovirus, Coxsackie virus A and B, enteric cytopathic human orphan (ECHO) viruses, hepatitis A virus, Sim
  • the family Bunyaviridae including the genus Bunyvirus (Bunyamwera and related viruses, California encephalitis group viruses), the genus Phlebovirus (Sandfly fever Sicilian virus, Rift Valley fever virus), the genus Nairovirus (Crimean-Congo hemorrhagic fever virus, Kenya sheep disease virus), and the genus Uukuvirus (Uukuniemi and related viruses); the family Orthomyxoviridae, including the genus Influenza virus (Influenza virus type A, many human subtype
  • the family Bunyaviridae including the genus Bunyvirus (Bunyamwera and related viruses, California encephalitis group viruses), the genus Phlebovirus (Sandfly fever Sicilian virus, Rift Valley fever virus), the genus Nairovirus (Crimean-Congo hemorrhagic fever virus, Kenya sheep disease virus), and the genus Uukuvirus (Uukuniemi and related viruses); the family Orthomyxoviridae, including the genus Influenza virus (Influenza virus type A, many human subtype
  • the present invention further envisions the use of viral vectors derived from the following DNA viruses including but not limited to the family Poxviridae, including the genus Orthopoxvirus (Variola major, Variola minor, Monkey pox Vaccinia, Cowpox, Buffalopox, Rabbitpox, Ectromelia), the genus Leporipoxvirus (Myxoma, Fibroma), the genus Avipoxvirus (Fowlpox, other avian poxvirus), the genus Capripoxvirus (sheeppox, goatpox), the genus Suipoxvirus (Swinepox), the genus Parapoxvirus (contagious postular dermatitis virus, pseudocowpox, bovine papular stomatitis virus); the family Iridoviridae (African swine fever virus, Frog viruses 2 and 3, Lymphocystis virus of fish); the family Herpesviridae
  • the present invention is further directed to the use of defective hepatitis B viruses as gene therapy reagent.
  • the gene therapy vector can be HSV.
  • DNA is delivered to an animal as an expression construct.
  • the expression construct In order to effect expression of a gene construct, the expression construct must be delivered into a cell.
  • the preferred mechanism for DNA delivery is via viral infection, where the expression construct is encapsidated in an infectious viral particle.
  • the expression construct may consist only of naked recombinant DNA or plasmids.
  • Transfection of the construct may be performed by any of the methods mentioned which physically or chemically permeabilize the cell membrane. One or other of these techniques may be successfully adapted for in vivo or ex vivo use,
  • the expression construct may be transfected as a liposome.
  • an expression construct complexed with Lipofectamine may be used to facilitate non-viral transfection.
  • the introduction of a vector into a cell of interest may be effected by methods known to one skilled in the art, such as electroporation, DEAE Dextran, cationic liposome fusion, protoplast fusion, DNA-coated microprojectile bombardment, injection with recombinant replication-defective viruses, homologous recombination, and naked DNA transfer by, for example, intravesical instillation. It will be appreciated by those skilled in the art that any of these methods of gene transfer may be combined.
  • PKC Activation Kinase Activity of PKC as Measured by Enzyme Assay
  • PKC assays crude sap (PEP001) and the PEP004 fraction was ether extracted twice to produce an ether-soluble fraction enriched in diterpenes, namely, ingenanes, jatrophanes and pepluanes. The remaining water soluble fraction was also used.
  • An ingenane fraction was prepared from the ether-soluble extract by TLC as described in International Patent Application No. PCT/AU98/00656.
  • PKC protein kinase C
  • the conventional and novel protein kinase C (PKC) isoforms in their unstimulated state, are inactive as kinases.
  • the C1 domain of these PKCs contains an autoinhibitory, pseudosubstrate site that binds to the substrate site (C4 domain) and inactivates the kinase functionality of the protein.
  • Activation of PKC results from binding of diacylglycerol (DAG) to the C1 domain, which, via multiple phosphorylation events and conformational changes to the protein, ultimately leads to the release of PKC autoinhibition.
  • DAG diacylglycerol
  • TPA and other related compounds have been shown to bind to the C1 domain of various PKC isoforms and presumably by similar means as DAG, lead to their activation.
  • kinase activity of rat brain PKC was determined using the PeptagTM Non-Radioactive Protein Kinase Kit (Promega). Using agarose gel electrophoresis the technique visualises the opposing electrostatic charge of a fluorescently labeled peptide (PLSRTLSVAAK) compared to the phosphorylated version of the same peptide.
  • PLSRTLSVAAK fluorescently labeled peptide
  • FIG. 1 The results of an assay of PKC with the fluorescent substrate (PepTag) are shown in FIG. 1.
  • the reaction mixture was separated by gel electrophoresis, showing migration of the unreacted substrate (a) to the anode (top), and the product (b), which is more negatively charged because of phosphorylation by PKC, moving towards the cathode (bottom).
  • the positive control activator (phosphatidyl serine) supplied by the manufacturer (lane 2) showed strong activation compared with PKC and substrate alone (lane 1).
  • Various dilutions of TPA also showed activation of PKC (lanes 3, 4 and 5).
  • FIG. 2 shows the results of testing fractionated materials simultaneously with negative (lane 1) and positive controls (lane 2).
  • Fraction H mixture of jatrophanes and pepluanes
  • lane 3 shows a low activity
  • arrow moving away from the unreacted substrate.
  • a similar result was found for the ingenane fraction (lane 4).
  • the kinase activity of the PKC sample was assessed before stimulation (Negative Control) and after stimulation with PEP001, phosphatidyl serine (an acid-lipid, known to activate PKC, provided by Promega; Positive Control) and TPA (20 ⁇ g/mL).
  • PEP001 phosphatidyl serine
  • TPA 20 ⁇ g/mL
  • PEP001 activates PKC to a similar level as phosphatidyl serine (200 ⁇ g/mL) and to a greater level than TPA (20 ⁇ g/mL). From this experiment, it is clear that the PEP001 activates PKC.
  • Activation of PKC can also be demonstrated by a simple fluorescence microscopy-based assay.
  • PKC Upon activation, PKC is known to translocate from the cytoplasm to the plasma membrane of the cell.
  • GFP green fluorescent protein
  • EGFP enhanced GFP
  • activation of the PKC can be detected by the movement of diffuse cytoplasmic GFP to a ring of fluorescence associated with the plasma membrane.
  • crude E. peplus extract has been shown to activate PKC ⁇ and PKC ⁇ .
  • MM96L cells were first transfected using a commercially-available kit (Qiagen Effectine Transfection Kit) with a PKC-GFP expression vector (Clontech; http://www.clontech.com/gfp/) and allowed to produce the PKC-GFP protein for 24 hr. The cells were then treated with crude E. peplus extract and TPA and observed under a fluorescent microscope (488 nm excitation). Two controls were used—no DNA, which allows for the identification of non-transfected cells, and no drugs, which allows for the calculation of transfection efficiency and the identification of transfected cells without PKC activation. pPKC ⁇ -EGFP and pPKC ⁇ -EGFP were tested, and crude E.
  • FIGS. 4A and 4B respectively show expression of PKCP in the absence of any drug and after exposure to crude E. peplus extract for 2 hr.
  • EGFP-PKC isoforms (Clontech) were available for this experiment, enabling the screening of the three predominant PKC families (i.e. classical, novel and atypical PKCs).
  • the members of the various PKC families are ⁇ , ⁇ , and ⁇ (classical), ⁇ (novel) and ⁇ (atypical).
  • HeLa cells were plated out in a 24-well plate containing coverslips and transfected with PKC isoforms fused to EGFP, using a commercially available effectine-transfection kit (QIAGEN, Pty. Ltd.). Cells were exposed to the transfection reagents for 16-24 hr. Subsequently, transfected cells were treated for one hour with TPA (100 ng/mL), bryostatin-1 (5 pg/mL), PEP003 (2.25 ⁇ g/mL; 5 ⁇ M) or PEP005 (670 ⁇ g/mL) 1.5 ⁇ M). Following treatment, cells were fixed on coverslips and mounted on glass slides.
  • TPA 100 ng/mL
  • bryostatin-1 5 pg/mL
  • PEP003 2.25 ⁇ g/mL; 5 ⁇ M
  • PEP005 670 ⁇ g/mL
  • the slides were subsequently examined visually by fluorescence microscopy, photographed, and over 150 cells were counted/treatment/PKC isoform. Counted cells were classified according to the localisation of the PKC-EGFP fluorescence as either cytoplasmic or plasma membrane using ImageProTM 4.1 (FIG. 5). Several cells also showed localisation to the Golgi, or similarly located cellular structure (FIG. 5). The number of these cells was also counted. Results are presented as an average and standard deviation of percentages of cells (Table 1).
  • results presented in FIG. 6 show that PKC ⁇ , ⁇ and ⁇ are translocated from the cytoplasm to the plasma membrane in response to treatment with PEP003, PEP005 and TPA but not with bryostatin-1.
  • the diacylglycerol-independent PKC ⁇ is not translocated in response to any treatment.
  • PKC ⁇ is translocated in response to PEP003, TPA and bryostatin-1, however, PEP005 does not induce any change in the isoenzymes localization.
  • the results also show that treatment of PKC ⁇ and ⁇ transfected cells with TPA, PEP003 and PEP005 leads to an increase in the number of cells displaying Golgi-like fluorescence.
  • PKC ⁇ transfected HeLa cells treated with TPA also show an increase in Golgi-Tike fluorescence.
  • treatment with PEP005 and bryostatin-1 decreases the number of cells with PKC ⁇ concentrated in the Golgi.
  • the number of PKC ⁇ transfected HeLa cells with Golgi-like localization is increased in response to all treatments.
  • a competition assay was performed to determine whether the diterpene esters of the instant invention bind to the phorbol ester binding site of PKC. This competition assay showed that 23 ⁇ g/mL PEP003 displaced >90% [3H]-phorbol dibutyrate from binding to rat brain homogenate, used as a source of PKC (Gonzalez et al., 1999). This binding was not blocked by co-incubation with bisindolylmaleimide. These results show that PEP003 binds to the phorbol ester binding site of PKC, and bisindolylmaleimide does not.
  • Anti-cancer agents have been widely investigated as potential anti-HIV agents.
  • PKC activators have been shown to activate latent retroviruses.
  • PMA has been shown to activate latent HIV in monocytes (Tobiume et al., 1998).
  • PMA is a known tumor promoter.
  • a latently HIV-1 infected cell line (U1) derived from the promonocytic cell line U937 after infection with HIV-1 LAI strain, was used in these experiments. In the absence of activation, no or little virus (measured as p24 production) is produced by the U1 cell line.
  • Phorbol esters are known to activate virus production from these cells (Tobiume et al., 1998) and so TPA/PMA was used as a positive control in these experiments.
  • U1 cells were cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum, 10 5 cells/mL were cultured for 20 hr in the presence and absence of various concentrations of either the phorbol ester TPA or crude E. peplus sap (PEP001) or PEP004 (HI) derived therefrom.
  • Supernatants were collected and viral replication monitored by determination of the amounts of HIV p24 gag protein in the culture supernatants by ELISA, using a NEN Life Science HIV-1 p24 ELISA kit. p24 values were calculated from OD values using a standard curve.
  • TPA the crude sap (PEP001) from E. peplus and the PEP004 fraction all activated HIV from U1 cells, as illustrated in FIG. 7.
  • the crude sap (PEP001) was 50 times less active than TPA.
  • the PEP004 fraction was 1000 times less active than TPA.
  • HIV human immunodeficiency virus
  • retrovirus a retrovirus
  • PBMC Peripheral blood mononuclear cells
  • the activated PBMC were infected with 10 g (Low Titre) and 100 ng (High Titre) of CA-p24 equivalents of the HIV-strain pNL4-3. Cells were infected for two hr after which, the virus was removed and the cells were washed with culture media.
  • FIGS. 8 A- 8 D show that PEP003 reduced virus replication kinetics in a dose-dependent manner.
  • PEP003 at concentrations of 500 nM, 50 nM and 5 nM reduced the replication rate by approximately 99.9%, 95% and 47%, respectively, relative to the untreated, infected cells.
  • PEP004 at dilutions of 1 ⁇ 10 4 and 1 ⁇ 10 5 reduced the replication rate by approximately 66% and 15%, respectively.
  • Viral load seemed to alter these results slightly, as higher initial inoculums of virus reduced the total inhibition of PEP003 at 500 nM or 50 nM to approximately 97% (t-test; p ⁇ 0.001) or 88% (t-test; p ⁇ 0.074), respectively.
  • the control compounds Ingenol (2.8 ⁇ M) and TPA (80 nM or 8 nM) reduced HIV-1 replication rates by approximately 35%, 98% and 38%, respectively.
  • Viruses and viral promoters especially adenovirus and CMV are used to deliver gene therapy in a range of human disease conditions. Gene expression and, hence, therapeutic effect will be enhanced if the promoters driving their transcription can be activated further by an agent.
  • Virus replication (enumerated as the number of stained cells following successive incubations with adenovirus antibody, peroxidase-conjugated protein A and peroxidase substrate) was increased by 344% with 67 ng/mL PEP005, 256% with 295 ng/mL PEP006, 248% with 226 ng/mL PEP008 and 147% with 67.5 ng/mL PEP010.
  • the CMV promoter is commonly used to activate the transcription of genes in constructs transfected into a variety of cells, due to its strong transcriptional activity in a variety of human cell types.
  • the ability of TPA to increase this activity has been demonstrated in cells undergoing non-productive infection with an adenovirus construct (Christenson et al., 1999), thus raising the possibility of increasing the production of a therapeutic protein encoded by a similar construct.
  • Human melanoma cells (MM96L; 50,000 per microtiter well) were treated with TPA or dilutions of crude E. peplus sap, infected with a 1/20 dilution of a pool of adenovirus-5 expressing ⁇ -galactosidase driven by the CMV promoter. After incubation for 20 hr, the wells were washed with 3 ⁇ with PBS, 50 ⁇ L of chlorophenol red galactoside (GPRG) substrate solution added and the absorbance at 540 nm read after 90 min. The inventors found TPA (100 ng/mL) and crude E. peplus sap (diluted 1 in 10,000) both induced the CMV promoter activity by >3-fold.
  • TPA 100 ng/mL
  • crude E. peplus sap diluted 1 in 10,000
  • Neutrophils represent about 70% of peripheral white blood cells in humans and play a pivotal role in inflammation and the innate defense against disease (Mollinedo, 1999). Upon activation, neutrophils release superoxide radicals and granules containing a variety of enzymes and other compounds. These secretions are able to destroy invading pathogens, but also result in inflammation and associated tissue damage.
  • E. peplus sap causes accumulation of neutrophils at the site of application, showing that E. peplus sap is capable of recruiting neutrophils.
  • a mixture of active diterpenes obtained as an ether extract from E. peplus sap was applied (2 ⁇ L of 100 mg/mL in ethanol) on the skin of a nu/nu mouse. After 24 hr, the animal was sacrificed and the skin fixed in 10% formalin for sectioning and hematoxylin/eosin staining. As shown in FIGS. 9A and 9B, control skin showed normal skin structure with few infiltrating monocytes. The treated skin showed large numbers of infiltrating neutrophils, characterized by their polymorphic nuclei. There was no evidence of gross damage to the skin.
  • Basal cell carcinoma is the most common cancer in the Caucasian population, with the highest annual incidence globally having been recorded in Australia (Miller et al., 1994, Marks et al., 1993).
  • New developments have begun looking at treating non-melanoma skin cancer (NMSC) using topical therapies. The essence of this therapy may rely upon the induction of an inflammatory response with infiltration of leucocytes, in particular neutrophils.
  • mice To assess whether the compounds of the invention induce neutrophil infiltration, an experiment was designed on C57BL/6J mice. Twenty-four mice were divided into six groups of four mice per group. In three of these groups the mice had a B16 melanoma injected s.c. (2 sites per mouse, 5 ⁇ 10 5 cells/site), that was left to grow for 8 days to approximate tumor sizes of 5-8 mm in diameter. A single application of one of all three compounds was then applied to the tumor or to normal skin. Each compound was applied on two groups of mice, one with tumor and 1 without tumor.
  • the three compounds were PEP010 (2 ⁇ L; 150 mM) in 10 ⁇ L of isopropanol gel (isopropyl alcohol 25% (w/w), propyl alcohol 25% (w/w)) (vehicle), PEP009 (2 ⁇ L of stock) in 10 ⁇ L of vehicle or vehicle alone as a control.
  • PEP010 (2 ⁇ L; 150 mM) in 10 ⁇ L of isopropanol gel (isopropyl alcohol 25% (w/w), propyl alcohol 25% (w/w)) (vehicle), PEP009 (2 ⁇ L of stock) in 10 ⁇ L of vehicle or vehicle alone as a control.
  • One mouse from each group was then sacrificed at either 4 hr, 24 hr, 48 hr or 144 hr post single application of compound and then tissue excised and sections prepared for histology.
  • Monocytes/macrophages are blood-borne and tissue cells which are usually activated by T lymphocytes and antibodies. Upon activation, they are able to phagocytose pathogens, release superoxide radicals and are an important source of cytokines.
  • Crude E. peplus extract was shown to be able to induce the release of superoxide radicals by use of a fluorescence-activated cell sorting (FACS)-based method, in which superoxide radicals are detected by the dye dihydroethidium.
  • FACS fluorescence-activated cell sorting
  • phagocytic activity was activated by E. peplus , as shown by increased uptake of nitroblue tetrazolium and adherence to plastic was increased by E. peplus ; this is believed to indicate activation and differentiation of macrophages.
  • PBMC Human peripheral blood mononuclear cells prepared by standard Ficoll separation comprise approximately 5% monocytes. PBMC were incubated with dihydroethidium, a reduced form of the dye which becomes fluorescent when oxidized by a respiratory burst, then treated in 10% FCS-RPMI 1640 at 37° C. for 15 min with crude E. peplus extract diluted 1/1000 or 100 ng/mL TPA and analyzed by flow cytometry using conventional methods (Handbook of Flow Cytometry Methods, p. 151) The mean channel numbers for fluorescence were 618 (controls) and 818 ( E. peplus extract diluted 1/1000). These results, illustrated in FIGS. 11A and 11B, show that the E.
  • peplus extract induced intracellular oxidation of the dye, typical of a respiratory burst. Phagocytic activity was determined by a conventional method (Hudson and Hay, Practical Immunology, 3 rd edition, p. 74). Cells were treated in 10% FCS-RPMI 1640 at 37° C. for 20 min with introblue tetrazolium (NBT) and crude E. peplus extract (PEP001) diluted 1/1000 or 100 ng/mL TPA, followd by counting the number of blue-stained cells in a haemocytometer. The average of three fields gave figures of ⁇ 2% (controls), 10% (TPA) and 8.7% ( E. peplus sap) cells stained blue. This demonstrates induction of phagocytic activity, part of the normal response to infectious agents, by E. peplus sap, as shown by uptake by cells of the blue NBT precipitate.
  • DCFH-DA 2′,7′-dichlorofluorescein diacetate
  • H 2 O 2 oxidizes the non-fluorescent probe (DCFH-DA) to a fluorescent probe that can then be detected by a flow cytometer.
  • PBMC Peripheral blood mononuclear cells
  • the cells were then incubated with DCFH-DA (1 ⁇ L/mL of 20 mM stock) for 15 minutes to allow it to be taken up and trapped by hydrolysis with cellular esterases. The cells were then stimulated by test compounds for 15 min at 37° C. Controls included in the experiment were unloaded control (cells with no DCFH-DA) and loaded control (cells with DCFH-DA, but no stimulation). These were used to monitor the non-specific oxidation of unstimulated cells. The cells were then analyzed on the flow cytometer (excitation at 488 nm, emission at 525 ⁇ 20 nm), gating each sample for individual cell populations-granulocytes, monocytes and lymphocytes (Table 3).
  • Phagocytosis by peripheral blood mononuclear cells was assayed (Steinkamp et al., 1982) using 1 ⁇ m FluoresbriteTM yellow-green fluorescent latex spheres (Polysciences, Inc., Warrington, Pa.).
  • a sample of whole, heparinized blood was treated with drug and 5 ⁇ 10E7 fluorescent latex beads in 10 ⁇ L of PBS added per mL of suspension.
  • Cells were incubated and maintained in suspension for 30 min by means of a shaker platform at 37° C. The stirnulated and non-stimulated samples were then lysed to isolate PBMCs.
  • the PBMCs were run on the flow cytometer measuring FITC (excitation at 488 nm, emission at 525 ⁇ 20 nm), gated for fluorescence (phagocytosed spheres) and light scatter (cell size).
  • viruses including alphaviruses
  • innate antiviral activities which are often mediated by the activation of interferon ⁇ / ⁇ responses (Antalis et al., 1998).
  • antiviral activities inhibit the ability of cells to support viral replication.
  • viral infections including those caused by Ross River virus
  • viral replication results in virus-induced cytophathic effect (CPE) or cell death.
  • CPE virus-induced cytophathic effect
  • Treatment of human fibroblast cells with E. peplus ingenanes was shown to activate antiviral activity and prevented CPE induced by an alphavirus infection.
  • GAS group A streptococcus
  • trep throat and impetigo (superficial skin infection)
  • RF rheumatic fever
  • RHD rheumatic heart disease
  • AGN acute glomerulonephritis
  • mice were treated with 50 ⁇ L of PEP003 (500 nM), PEP004 (1:100 dilution from stock) or control (PBS/10% acetone), 24 hr prior to and thereafter i.p. challenge with live GAS.
  • PEP003 500 nM
  • PEP004 1:100 dilution from stock
  • control PBS/10% acetone
  • mice Two different strains of mice (Quackenbush and B10.BR) and four different GAS strains (NS-1, PL-1, 88/30 and M1) were used. Mice were monitored for two weeks post-challenge and the percentage survival of mice determined.
  • Percentage survival in Quackenbush mice challenged with PL-1 GAS was 70% (PEP003), 60% (PEP004) and 40% (control) (Table 5).
  • Control mice that had been given the same successive treatment of PEP003 and PEP004 (but not challenged) to rule out any potential adverse side effects of the compounds were then also challenged with PL-1; survival was 40%, 80%, and 20% for PEP003, PEP004 and controls, respectively (Table 6).
  • survival was 50% for PEP003 and controls, and 80% for PEP004 (Table 5).
  • FIGS. 12 and 13 show that treatment of leucocytes with PEP003 results in a significant reduction in bacterial numbers.
  • Ringworm is a subcutaneous mycosis or dermatophytosis caused by fungi of the species Trichophyton, Microsporum and Epidermophyton, in which the infection is confined to the keratinous structures of the body.
  • a two week old ringworm lesion, determined to be Trichophyton mentagrophytes var. mentagrophytes by culture, on the volar surface of the forearm of an adult male human was treated with a single topical application of crude E. peplus extract and was shown to resolve after seven days. Resolution of such lesions in the absence of treatment does occur, but is considered extremely rare.
  • PEP004 had no effect on VCA production in any of the cell lines used. However, PEP003 did induce high levels of VCA in both Burkitt's lymphoma cell lines (MutuI and BL74), but only at 10 5 concentration (FIG. 15). Similar results were obtained when the cell lines were assayed for induction of BZLF1, the initial transactivator of EBV replication (FIG. 16). The results show that PEP003 was capable of activating EBV in Burkitt's lymphoma cell lines, but appeared to have little effect on LCLs.
  • both TPA and PEP003 can modulate gene expression in EBV transformed tumor cells at the doses used; (2) while PEP003 induced VCA in MutuI cells TPA did not, indicating different modes of action; (3) surprisingly, there was no apparent effect of PEP003 on lymphoblastoid cells, indicating potential for activating latent herpesvirus in tumors without affecting the normal infection.
  • Melanomas and other cancers can be killed by both specific (T cell-mediated) and non-specific (natural killer cell and other mechanisms) arms of the immune response. These killer cells can be generated in vitro by stimulating peripheral blood T cells from selected melanoma patients with melanoma cells derived from the same patient (“autologous”). Natural killer cells can be recognized by their lysis of the natural killer-sensitive cell line K562. It has been theorized that some anti-tumor agents alter the susceptibility of melanomas to immune responses.
  • Peripheral blood lymphocytes from patient A02 who has a strong specific T cell response to her own melanoma cells (A02-M), were thawed and stimulated by irradiated A02-M pre-treated overnight at 37° C. with (a) PEP003 (2.25 ⁇ g/mL; 50 ⁇ M); (b) TPA (100 ng/mL); or (c) control solvent/buffer, and washed ⁇ 2 before addition to responding lymphocytes (washing ⁇ 2 achieves a residual agent dilution of ⁇ 100,000).
  • NK-sensitive cell line K562
  • All determinations were performed in triplicate, at E:T ratios of 45, 15, 5 and 1.7:1.
  • a standard 5 hour ⁇ 51 Cr release assay was performed.
  • Stimulations were performed in 10% fetal bovine serurn/RPMI-1640.
  • Fresh E. peplus plants (17 kg) were chopped and soaked in 150 litres of water at 4° C. for 20 hr. The water was pumped through 50 and 100 mesh sieves, filtered through 5 and 2 micron filters and then recirculated through a 100 mm diameter column of Amberlite XAD-16 (1.5 kg, conditioned successively with ethyl acetate, methanol and water) at 4° C. (approximately 1.2 L/min) for 72 hr. Adsorption of bioactivity to the resin was found to be virtually complete within 20 hr.
  • the hydrophobic adsorbent polyamide (ICN Biomedical Research Products) was also used to trap the diterpenes from water; it had the advantage of allowing the diterpene esters to be selectively eluted with 50-80% methanol, thus separating them from inactive, hydrophobic compounds, which remained on the column.
  • the following method is based upon the surprising discovery that the stems of E. peplus contain approximately 90% of the bioactive diterpenes and significantly less chlorophyll compared with the leaves.
  • Leukocytes obtained by lysis of human peripheral blood were added to 5000 MM96L human melanoma cells or 7000 neonatal foreskin fibroblasts per microtitre well at effector: target ratios of 1000, 100 and 10:1.
  • Ing9 60 ng/mL was added and after 48 hr incubation the cultures were washed and labelled with [3H]-thymidine for 2 hr.
  • the melanoma cells showed 9% survival with PEP008 whereas the normal fibroblasts had 100% survival.
  • Untreated leukocytes had no effect on cell survival.
  • diterpene esters of the invention activate human peripheral blood leukocytes to produce, in a PKC-dependent manner, phagocytosis and a respiratory burst which are potentially lethal to micro-organisms and other cells.
  • Leukocytes obtained by lysis of human peripheral blood were added to 5000 MM96L human melanoma cells or 7000 neonatal foreskin fibroblasts per microtitre well at effector: target ratios of 1000, 100 and 10:1.
  • the target cells had been treated with 60 ng/mL PEP008 for 20 hr beforehand, and washed and the medium replaced before the leukocytes were added. After 48 hr incubation with the leukocytes the cultures were washed and labelled with [3H]-thymidine for 2 hr.
  • the melanoma cells showed 12% survival with PEP008 whereas the normal fibroblasts had 100% survival. Untreated leukocytes had no effect on cell survival.
  • Topical Composition A for the Treatment of Conditions Affecting Skin e.g. Infections, Skin Cancers
  • Tinctures Compounds of the invention were diluted into acetone, ethanol or isopropanol to the same final bioactivity as the E. peplus latex as measured by bipolar activity in MM96L human melanoma cells (10 million bp units per mL). Samples (2-5 ⁇ L) were applied daily for 3 days to the surface of mouse melanoma B16 tumor 3-5 days after implanting s.c. 1 million cells on the flanks of nude mice. Efficacy, defined as 67% or more sites cured, was obtained for E. peplus sap, PEP005, PEP008 and a mixture of PEP005, PEP006 and PEP008.
  • Topical Composition B for the Treatment of Conditions Affecting Skin e.g. Infections, Skin Cancers
  • Creams and gels A variety of hydrophobic cream bases was found to be ineffective when used to deliver compounds to the skin as described above for the tinctures.
  • E. peplus sap and its terpenoid components activate PKC, with consequent potential to induce a wide range of cellular responses without the high tumor promoting activity of TPA.
  • the carboxypeptidase activity may have application in enhancement of tissue penetration and in antigen processing for optimal immune responses.
  • E. peplus extract induces a set of cellular responses with affects PKC, cell cycle genes and inflammatory mediators, some but by no means all of which are similar to the action of TPA.
  • E. peplus sap and its terpenoid components are useful in the treatment of a variety of infections and as adjuvants for stimulating immune responses.
  • Example 24 The saps of Example 24 were also examined for their cytotoxic effect on the ovarian carcinoma cell line JAM.
  • FIGS. 18A and 18B indicate that, like the sap of E. peplus , the saps of S. grantii, S. compactum, M. lugardae, M. guentheri , and E. gossweileni, at concentrations of 10 ⁇ 4 and above, are effective in killing JAM cells. These results also show that cytotoxicity is inhibited by bisindolylmaleimide, suggesting that this effect is mediated by modulation of PKC.
  • FIG. 18C Inspection of FIG. 18C reveals that the cytotoxic effects of saps derived from M. guentheri and E. gossweileni were blocked in the presence of phorbol dibutyrate, suggesting that the active components of these saps mediate their cytotoxicity by binding to the phorbol ester binding site of PKC.

Abstract

The present invention relates generally to chemical agents useful in the treatment and prophylaxis of protein kinase C (PKC) related conditions in mammals, including humans and primates, non-mammalian animals and avian species. More particularly, the present invention provides a chemical agent of the macrocyclic diterpene family obtainable from a member of the Euphorbiaceae family of plants or botanical or horticultural relatives thereof or derivatives or chermical analogues or chemically synthetic forms of the agents for use in the treatment or prophylaxis of PKC-related conditions in mammalian, animal and avian subjects. The subject chemical agents are also useful for modulating expression of genetic sequences including promotion and other regulatory sequences. The present invention further contemplates a method for the prophylaxis and/or treatment in mammalian, animal or avian subjects with PKC-related conditions by the topical or systemic administration of a macrocyclic diterpene obtainable from a member of the Euphorbiaceac family of plants or their botanical or horticultural derivatives or a derivative, chemical analogue or chemically synthetic form of the agent. The chemical agent of the present invention may be in the form of a purified compound, mixture of compounds, a precursor form of one or more of the compounds capable of chemical transformation into a therapeutically and/or genetically active agent or in the form of a chemical fraction, sub-fraction, preparation or extract of the plant.

Description

    FIELD OF THE INVENTION
  • The present invention relates generally to chemical agents useful in the treatment and prophylaxis of protein kinase C (PKC) related conditions in mammals, including humans and primates, non-mammalian animals and avian species. More particularly, the present invention provides a chemical agent of the macrocyclic diterpene family obtainable from a member of the Euphorbiaceac family of plants or botanical or horticultural relatives thereof or derivatives or chemical analogues or chemically synthetic forms of the agents for use in the treatment or prophylaxis of PKC-related conditions in mammalian, animal and avian subjects. The subject chemical agents are also useful for modulating expression of genetic sequences including promotion and other regulatory sequences. The present invention further contemplates a method for the prophylaxis and/or treatment in mammalian, animal or avian subjects with PKC-related conditions by the topical or systemic administration of a macrocyclic diterpene obtainable from a member of the Euphorbiaceae family of plants or their botanical or horticultural derivatives or a derivative, chemical analogue or chemically synthetic form of the agent. The chemical agent of the present invention may be in the form of a purified compound, mixture of compounds, a precursor form of one or more of the compounds capable of chemical transformation into a therapeutically and/or genetically active agent or in the form of a chemical fraction, sub-fraction, preparation or extract of the plant. [0001]
  • BACKGROUND OF THE INVENTION
  • Bibliographic details of the publications referred to by author in this specification are collected at the end of the description. [0002]
  • Reference to any prior art in this specification is not, and should not be taken as, an acknowledgment or any form of suggestion that this prior art forms part of the common general knowledge in Australia or any other country. [0003]
  • Natural product screening is a term applied to the screening of natural environments for bioactive molecules. Particularly sought after bioactive molecules are those having potential as useful therapeutic agents. Natural environments include plants, microorganisms, coral and marine animals. The search for potential therapeutic agents for the treatment of cancer and infection by pathogenic organisms remains an important focus. [0004]
  • The Euphorbiaceae family of plants covers a wide variety of plants including weeds of Euphorbia species. There have been a variety of inconclusive reports on the potential effects of the sap of these plants on a variety of conditions as well as promoting tumorigenesis and causing skin and ocular irritation. [0005]
  • The most intensively studied species of this group is [0006] Euphorbia pilulifera L (synonyms E. hirta L., E. capitata Lam.), whose common names include pill-bearing spurge, snakeweed, cat's hair, Queensland asthma weed and flowery-headed spurge. The plant is widely distributed in tropical countries, including India, and in Northern Australia, including Queensland.
  • A recent report describes selective cytotoxicity of a number of tiglilane diterpene esters from the latex of [0007] Euphorbia poisonii, a highly toxic plant found in Northern Nigeria, which is used as a garden pesticide. One of these compounds has a selective cytotoxicity for the human kidney carcinoma cell line A-498 more than 10,000 times greater than that of adriamycin (Fatope et al., 1996).
  • [0008] Euphorbia hirta plants and extracts thereof have been considered for a variety of purposes, including tumor therapy (EP 0 330 094), AIDS-related complex and AIDS (HU-208790) and increasing immunity and as an anti-fungoid agent for treatment of open wounds (DE-4102054).
  • Thus, while there are isolated reports of anti-cancer activity of various Euphorbia preparations (see Fatope et al., 1996; Oksuz et al., 1996), not only are the compounds present in at least one Euphorbia species reported to be carcinogenic (Evans and Osman, 1974; Stavric and Stolz, 1976; Hecker, 1970), but at least one species has a skin-irritant and tumor-promoting effect (Gundidz et al., 1993) and another species reduces EBV-specific cellular immunity in Burkitt's lymphoma (Imai, 1994). [0009]
  • In accordance with the present invention, the inventors have identified chemical agents and fractions comprising these agents which are useful in the treatment and prophylaxis of PKC-related conditions in an animal, mammal or avian species. The instant agents are also useful in modulating and in particular stimulating or otherwise promoting expression or function of nucleotide sequences such as promoter or other regulatory sequences which is useful in gene therapy, genetic therapy, genetic augmentation and gene replacement therapies as well as promoting expression of genetic material into an expression product. [0010]
  • SUMMARY OF THE INVENTION
  • Throughout this specification, unless the context requires otherwise, the word “comprise”, or variations such as “comprises” or “comprising”, will be understood to imply the inclusion of a stated element or integer or group of elements or integers but not the exclusion of any other element or integer or group of elements or integers. [0011]
  • The present invention is predicated in part on the identification of chemical agents and fractions comprising same from plants of the Euphorbiaceae family which are useful in the treatment and prophylaxis of PKC-related conditions in an animal, mammal incluidng a human and avian species. The inventors have identified that the chemical agents of the present invention are capable of modulating protein kinase C (PKC) activity thus providing a basis for the treatment of conditions where PKC activity is required to be up-regulated or down-regulated. The instant chemical agents further modulate and more particularly activate or otherwise promote expression of genetic sequences and/or operation of promoter or other regulatory sequences. The latter is useful for genetic therapies including gene therapy. [0012]
  • Accordingly, one aspect of the present invention contemplates a method for the treatment or prophylaxis of a PKC-related condition or disorder in a subject, said method comprising the administration to said subject of a symptom-ameliorating effective amount of a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or chemical analogue thereof which chemical agent is a macrocyclic diterpene selected from compounds of the ingenane, pepluane and jatrophane families and which chemical agent or derivative or chemical analogue is represented by any one of the general formulae (I)-(V) as defined herein and which chemical agent or derivative or chemical analogue thereof is capable of modulating PKC activity, PKC-gene expression or PKC enzyme turnover and wherein said chemical agent or its derivatives or chemical analogues is administered for a time and under conditions sufficient to ameliorate one or more symptoms associated with a PKC-related condition or disorder. [0013]
  • Another aspect of the present invention provide a method for the treatment or prophylaxis of a PKC-related condition or disorder in a subject, said method comprising the administration to said subject of a symptom-ameliorating effective amount of a macrocyclic diterpene, or a chemical fraction comprising same from a plant of the family Euphorbiaceae or a derivative or chemical analogue of said macrocyclic diterpene having the structures as defined above wherein said macrocyclic diterpene or its derivative or chemical analogue modulates PKC activity, synthesis or enzyme turnover, said administration being for a time and under conditions sufficient to ameliorate one or more symptoms of associated with a PKC-related condition or disorder. [0014]
  • Yet another aspect of the invention encompasses a method of modulating the expression of a genetic sequence for the treatment or prophylaxis of a condition or disorder in a subject, said genetic sequence being under the control of a promoter whose activity is modulated by a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or chemical analogue thereof which chemical agent is a macrocyclic diterpene selected from compounds of the ingenane, pepluane and jatrophane families and which chemical agent or derivative or chemical analogue is represented by any one of the general formulae (I)-(V) as defined herein and which chemical agent or derivative or chemical analogue thereof is capable of modulating PKC activity, PKC-gene expression or PKC enzyme turnover, said method comprising administering to said subject an expression facilitating amount of said chemical agent or derivative or chemical analogue thereof for a time and under conditions sufficient to facilitate the expression of said genetic sequence to thereby ameliorate one or more symptoms associated with said condition or disorder. [0015]
  • Still another aspect in the invention envisions a method of modulating the expression of a genetic sequence by the administration of a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or chemical analogue thereof which chemical agent is a macrocyclic diterpene selected from compounds of the ingenane, pepluane and jatrophane familes and which chemical agent or derivative or chemical analogue is represented by any one of the general formulae (I)-(V) as defined herein and which chemical agent or derivative or chemical analogue thereof is capable of modulating PKC activity, PKC-gene expression or PKC enzyme turnover and wherein said chemical agent or its derivatives or chemical analogues is administered for a time and under conditions sufficient to facilitate the expression of said genetic sequence. [0016]
  • Still yet another aspect of the present invention features a method of stimulating the activation or function of a promoter by the administration of a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or chemical analogue thereof which chemical agent is a macrocyclic diterpene selected from compounds of the ingenane, pepluane and jatrophane families and which chemical agent or derivative or chemical analogue is represented by any one of the general formulae (I)-(V) as defined herein and which chemical agent or derivative or chemical analogue thereof is capable of modulating PKC activity, PKC-gene expression or PKC enzyme turnover and wherein said chemical agent or its derivatives or chemical analogues is administered for a time and under conditions sufficient to stimulate the activation or function of said promoter. [0017]
  • Even yet another aspect of the present invention contemplates a method for the treatment or prophylaxis of a PKC-related condition in a subject, said method comprising the administration to said subject of a symptom-ameliorating or immunopotentiating effective amount of a macrocyclic diterpene obtainable from a Euphorbiaceae plant or its botanical or horticultural relative, said macrocyclic diterpene being selected from an ingenane, pepluane or jatrophane, or a derivative or chemical analogue thereof, having the structure represented by any one of the general formulae (I)-(V) as defined herein and which chemical agent or derivative or chemical analogue thereof is capable of modulating PKC activity, PKC-gene expression or PKC enzyme turnover and wherein said chemical agent or its derivatives or chemical analogues is administered for a time and under conditions sufficient to ameliorate one or more symptoms associated with said biological entity and wherein said chemical agent exhibits a potency of agent (P[0018] A) of >10, wherein the PA=ΣIV where IV is a numerical value associated with a particular feature as defined in Table A or pharmaceutically acceptable salts of these, said chemical agent being administered for a time and under conditions sufficient to ameliorate at least one symptom caused by or associated with said PKC-related condition.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 shows the activation of PKC, using a fluorescent peptide assay (“PepTag” non-radioactive protein kinase kit, Promega). [0019] Lane 1, PKC and substrate alone; lane 2, plus positive control activator; lane 3, plus 100 ng/ml TPA; lane 4, plus 0.1 ng/ml TPA; lane 5, plus 0.01 ng/ml TPA; lane 6, plus 0.001 ng/ml TPA; lane 7, ether extract of E. peplus sap in DEM, diluted 1 in 5; lane 8, aqueous layer from ether extraction, diluted 1/25; lane 9, crude sap diluted 1/25; lane 10, DME alone.
  • FIG. 2 shows the activation of PKC by [0020] E. peplus fractions. Lanes 1 and 2, same as FIG. 1; lane 3, 2 mg/ml fraction H; lane 4, 2 mg/ml ingenanes.
  • FIG. 3 is photographic representation showing the results of a PKC assay using rat brain PKC. [0021] Lane 1, negative control; lane 2, positive control; lane 3, empty; lane 4, PEP001 (1/125 dilution), lane 5, PEP001 (1/500 dilution) and lane 6, TPA (20 μg).
  • FIG. 4 is a photographic representation showing the activation of PKC in MM96L cells expressing PKC fused to green fluorescent protein (GFP). (A) PKCβ expressed in the nuclei of MM96L human melanoma PKC MM96L cells in the absence of drug. (B) After treatment with crude [0022] E. peplus extract for 2 hr.
  • FIG. 5 is a photographic representation showing induction of translocation of activated PKCs by the compounds of the instant invention to the cytoplasm, plasma membrane and to the Golgi or similarly located cellular structure. [0023]
  • FIG. 6 is a graphical representation showing the induction of translation of the classical and novel PKC isoforms in response to PEP003, PEP005, bryostatin-1 and TPA. [0024]
  • FIG. 7 is a graphical representation showing the activation of HIV from U1 cells. [0025]
  • FIG. 8 is a graphical representation showing treatment of lytic HIV infection of peripheral blood mononuclear cells (PBMC) with PEP003, PEP004, TPA and ingenol, expressed as p24 production over a 10 day treatment period. (A) Uninfected cells, (B) low titer infected cells, (C) low titer infected cells represented as p24 production versus drug concentration, (D) same as (C) but high titer infection. [0026]
  • FIG. 9 is a photographic representation showing the recruitment of neutrophils in the skin induced by PEP001 extract. (A) Normal skin of nude mouse. (B) Skin of nude mouse showing infiltration of neutrophils one day after treatment with [0027] E. peplus sap.
  • FIG. 10 is a photographic representation showing effect of PEP010 onrecruitment of neutrophils in normal skin of nude mouse and skin overlying subcutaneously implanted B16 melanoma. (A) 24 hr treatment, (B) 48 hr treatment. [0028]
  • FIG. 11 is a graphical representation illustrating the ability of PEP001 to induce the release of superoxide radical, as demonstrated by fluorescence-activated cell sorting. [0029]
  • FIG. 12 is a graphical representation showing the effect of pre-treatment of leukocytes with PEP003 on [0030] E. coli activity (16 hr incubation), relative to PBS control; depicted as numbers of E. coli cells/ml media.
  • FIG. 13 is a graphical representation showing the effect of pre-treatment of leukocytes with PEP003 on [0031] E. coli numbers depicted in terms of turbidity.
  • FIG. 14 is a photographic representation showing production of viral capsid antigen (VCA) in B95-8 (EBV+ Marmoset cell line) after treatment with TPA, PEP003 and PEP004 for 3 and 7 days. [0032]
  • FIG. 15 is a photographic representation showing production of viral capsid antigen (VCA) in BL74 and Mutu I (Burkitts lymphoma cell lines) after treatment with TPA, PEP003 and PEP004 for 3 and 7 days. [0033]
  • FIG. 16 is a photographic representation showing production of BZLF1 (the initial transactivator of EBV) after treatment with TPA, PEP003 and PEP004 for 3 and 7 days. [0034]
  • FIG. 17 is a graphical representation showing activation of natural killer cell activity, assayed as % specific lysis of K562 cells (a natural killer-sensitive cell line) after pre-treatment of AO2-M melanoma cells with PEP003 and TPA. [0035]
  • FIG. 18 is a graphical representation showing survival of Jam cells after treatment with saps from the Euphorbiaceae, expressed as percentage cell survival determined by sulfurhodamine B staining of cells. [0036]
  • FIG. 19 is a diagrammatic representation of a system used to carry out the instructions encoded by the storage medium of FIGS. 9 and 10. [0037]
  • FIG. 20 is a diagrammatic representation of a cross-section of a magnetic storage medium. [0038]
  • FIG. 21 is a diagrammatic representation of a cross-section of an optically readable data storage system.[0039]
  • Compounds may be referred to in the subject specification by a compound code. These are defined as below: [0040]
    TABLE OF COMPOUND CODES
    COMPOUND CODE DESCRIPTION
    PEP001 Crude sap
    PEP002 Methanol and ether extract of E. peplus sap
    prepared according to Example 7 of
    PCT/AU98/00656
    PEP003 Ingenane enriched fraction prepared according
    to Examples 21 and 23
    PEP004 Jatrophane/Pepluane enriched fraction prepared
    according to Example 7 of PCT/AU98/00656
    PEP005 20-hydroxy-ingenol-3-angelate
    PEP006 Ingenol-3-angelate
    PEP008 20-O-acetyl-ingenol-3-angelate
    PEP009 Acetone Extract of XAD prepared according to
    Example 21
    PEP010 Ingenane enriched fraction prepared according to
    Examples 22 and 23
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • The present invention is predicated in part on the identification of biologically useful properties of chemical agents and chemical fractions comprising these agents obtainable from a member of the Euphorbiaceae family of plants or their botanical or horticultural relatives. These biologically useful properties include their use in the prophylaxis and/or treatment of PKC-related conditions. Reference herein to a “PKC-related condition” includes a PKC-related disorder and comprises medical including psychological conditions associated with aberrant PKC activity including activating low PKC activity or lowering higher than normal levels of PKC as well as conditions associated with the presence of latent virus in a host cell. [0041]
  • Examples of PKC disorders include but are not limnited to alcoholism, Alzheimer's disease, asthma, atherosclosis, atopic dermatitis, autoimmune disease, bipolar-disorder, blood disorder, cardiac hypertrophy, depression, diabetes, hypertension, hyperplastic dermatosis, multiple sclerosis, myocardial ischemia, osteoarthritis, psoriasis, rheumatoid arthritis and transplantation. The instant chemical agents are also useful in promoting gene activation and promotion. [0042]
  • The term “treatment” is used in its broadest sense and includes the prevention of a PKC-related condition or the reversal of a condition to asymptomatic levels or to levels in between as well as facilitating the amelioration of the effects of symptoms of the PKC-related condition. A PKC-related condition occurs when an increase or decrease in PKC promotes or facilitates an effect. Furthermore, “treatment” further encompasses promoting an elevation in promoter or other regulatory sequence activity and/or structural gene sequence expression. [0043]
  • The term “prophylaxis” is also used herein in its broadest sense to encompass a reduction in the risk of development of a PKC-related condition. In certain conditions, an agent may act to treat a subject prophylactically. Furthermore, the prophylactic administration of an agent may result in the agent becoming involved in the treatment of a pathological condition. Use of the terms “treatment” or “prophylaxis” is not to be taken as limiting the intended result which is to reduce the incidence of a PKC-related disorder or condition and/or to ameliorate the symptoms or risk of development of symptoms caused or facilitated by a PKC-related condition. [0044]
  • The present invention is particularly directed to the use of one or more macrocyclic diterpenes from a member of the Euphorbiaceae family of plants or botanical or horticultural relatives of such plants. Reference herein to a member of the Euphorbiaceae family includes reference to species from the genera Acalypha, Acidoton, Actinostemon, Adelia, Adenocline, Adenocrepis, Adenophaedra, Adisca, Agrostistachys, Alchornea, Alchorneopsis, Alcinaeanthus, Alcoceria, Aleurites, Amanoa, Andrachne, Angostyles, Anisophyllum, Antidesma, Aphora, Aporosa, Aporosella, Argythamnia, Astrococcus, Astrogyne, Baccanrea, Baliospermum, Bernardia, Beyeriopsis, Bischofia, Blachia, Blumeodondron, Bonania, Bradleia, Breynia, Breyniopsis, Briedelia, Buraeavia, Caperonia, Caryodendron, Celianella, Cephalocroton, Chaenotheca, Chaetocarpus, Chamaesyce, Cheilosa, Chiropetalum, Choriophyllum, Cicca, Chaoxylon, Cleidon, Cleistanthus, Cluytia, Cnesmone, Cnidoscolus, Coccoceras, Codiaeum, Coelodiscus, Conami, Conceveiba, Conceveibastrum, Conceveibum, Corythea, Croizatia, Croton, Crotonopsis, Crozophora, Cubanthus, Cunuria, Dactylostemon, Dalechampia, Dendrocousinsia, Diaspersus, Didymocistus, Dimorphocalyx, Discocarpus, Ditaxis, Dodecastingma, Drypetes, Dysopsis, Elateriospermum, Endadenium, Endospermum, Erismanthus, Erythrocarpus, Erythrochilus, Eumecanthus, Euphorbia, Euphorbiodendron, Excoecaria, Flueggea, Calearia, Garcia, Gavarretia, Gelonium, Giara, Givotia, Glochidion, Clochidionopsis, Glycydendron, Gymnanthes, Gymnosparia, Haematospermum, Hendecandra, Hevea, Hieronima, Hieronyma, Hippocrepandra, Homalanthus, Hymenocardia, Janipha, Jatropha, Julocroton, Lasiocroton, Leiocarpus, Leonardia, Lepidanthus, Leucocroton, Mabea, Macaranga, Mallotus, Manihot, Mappa, Maprounea, Melanthesa, Mercurialis, Mettenia, Micrandra, Microdesmis, Microelus, Microstachy, Maocroton, Monadenium, Mozinna, Neoscortechinia, Omalanthus, Omphalea, Ophellantha, Orbicularia, Ostodes, Oxydectes, Palenga, Pantadenia, Paradrypeptes, Pausandra, Pedilanthus, Pera, Peridium, Petalostigma, Phyllanthus, Picrodendro, Pierardia, Pilinophytum, Pimeleodendron, Piranhea, Platygyna, Plukenetia, Podocalyx, Poinsettia, Poraresia, Prosartema, Pseudanthus, Pycnocoma, Quadrasia, Reverchonia, Richeria, Richeriella, Ricinella, Ricinocarpus, Rottlera, Sagotia, Sanwithia, Sapium, Savia, Sclerocroton, Sebastiana, Securinega, Senefeldera, Senefilderopsis, Serophyton, Siphonia, Spathiostemon, Spixia, Stillingia, Strophioblachia, Synadenium, Tetracoccus, Tetraplandra, Tetrorchidium, Thyrsanthera, Tithymalus, Trageia, Trewia, Trigonostemon, Tyria and Xylophylla. [0045]
  • The most preferred genus and most suitable for the practice of the present invention is the genus Euphorbia. Particularly useful species of this genus include [0046] Euphorbia aaron-rossii, Euphorbia abbreviata, Euphorbia acuta, Euphorbia alatocaulis, Euphorbia albicaulis, Euphorbia algomarginata, Euphorbia aliceae, Euphorbia alta, Euphorbia anacampseros, Euphorbia andromedae, Euphorbia angusta, Euphorbia anthonyi, Euphorbia antiguensis, Euphorbia apocynifolia, Euphorbia arabica, Euphorbia ariensis, Euphorbia arizonica, Euphorbia arkansana, Euphorbia arteagae, Euphorbia arundelana, Euphorbia astroites, Euphorbia atrococca, Euphorbia baselicis, Euphorbia batabanensis, Euphorbia bergeri, Euphorbia bermudiana, Euphorbia bicolor, Euphorbia biformis, Euphorbia bifurcata, Euphorbia bilobata, Euphorbia biramensis, Euphorbia biuncialis, Euphorbia blepharostipula, Euphorbia blodgetti, Euphorbia boerhaavioides, Euphorbia boliviana, Euphorbia bracei, Euphorbia brachiata, Euphorbia brachycera, Euphorbia brandegee, Euphorbia brittonii, Euphorbia caesia, Euphorbia calcicola, Euphorbia campestris, Euphorbia candelabrum, Euphorbia capitellata, Euphorbia carmenensis, Euphorbia carunculata, Euphorbia cayensis, Euphorbia celastroides, Euphorbia chalicophila, Euphorbia chamaerrhodos, Euphorbia chamaesula, Euphorbia chiapensis, Euphorbia chiogenoides, Euphorbia cinerascens, Euphorbia clarionensis, Euphorbia colimae, Euphorbia colorata, Euphorbia commutata, Euphorbia consoquitlae, Euphorbia convolvuloides, Euphorbia corallifera, Euphorbia creberrima, Euiphorbia crenulata, Euphorbia cubensis, Euphorbia cuspidata, Euphorbia cymbiformis, Euphorbia darlingtonii, Euphorbia defoliata, Euphorbia degeneri, Euphorbia deltoidea, Euphorbia dentata, Euphorbia depressa Euphorbia dictyosperma, Euphorbia dictyosperma, Euphorbia dioeca, Euphorbia discoidalis, Euphorbia dorsiventralis, Euphorbia drumondii, Euphorbia duclouxii, Euphorbia dussii, Euphorbia eanophylla, Euphorbia eggersii, Euphorbia eglandulosa, Euphorbia elata, Euphorbia enalla, Eiphorbia eriogonoides, Euphorbia eriophylla, Euphorbia esculaeformis, Euphorbia espirituensis, Euphorbia esula, Euphorbia excisa, Euphorbia exclusa, Euphorbia exstipitata, Euphorbia exstipulata, Euphorbia fendleri, Euphorbia filicaulis, Euphorbia filiformis, Euphorbia florida, Euphorbia fruticulosa, Euphorbia garber, Euphorbia gaumerii, Euphorbia gerardiana, Euphorbia geyeri, Euphorbia glyptosperma, Euphorbia gorgonis, Euphorbia gracilior, Euphorbia gracillima, Euphorbia gradyi, Euphorbia graminea, Euphorbia graminiea Euphorbia grisea, Euphorbia guadalajarana, Euphorbia guanarensis, Euphorbia gymnadenia, Euphorbia haematantha, Euphorbia hedyotoides, Euphorbia heldrichii, Euphorbia helenae, Euphorbia helleri, Euphorbia helwigii, Euphorbia henricksonii, Euphorbia heterophylla, Euphorbia hexagona, Euphorbia hexagonoides, Euphorbia hinkleyorum, Euphorbia hintonii, Euphorbia hirtula, Euphorbia hirta, Euphorbia hooveri, Euphorbia humistrata, Euphorbia hypericifolia, Euphorbia inundata, Euphorbia involuta, Euphorbia jaliscensis, Euphorbia jejuna, Euphorbia johnston, Euphorbia juttae, Euphorbia knuthii, Euphorbia lasiocarpa, Euphorbia lata, Euphorbia latazi, Euphorbia latericolor, Euphorbia laxiflora Euphorbia lecheoides, Euphorbia ledienii, Euphorbia leucophylla, Euphorbia lineata, Euphorbia linguiformis, Euphorbia longecornuta, Euphorbia longepetiolata, Euphorbia longeramosa, Euphorbia longinsulicola, Euphorbia longipila, Euphorbia lupulina, Euphorbia lurida, Euphorbia lycioides, Euphorbia macropodoides, macvaughiana, Euphorbia manca, Euphorbia mandoniana, Euphorbia mangleti, Euphorbia mango, Euphorbia marylandica, Euphorbia mayana, Euphorbia melanadenia, Euphorbia melanocarpa, Euphorbia meridensis, Euphorbia mertonii, Euphorbia mexiae, Euphorbia microcephala, Euphorbia microclada, Euphorbia micromera, Euphorbia misella, Euphorbia missurica, Euphorbia montana, Euphorbia montereyana, Euphorbia multicaulis, Euphorbia multiformis, Euphorbia multinodis, Euphorbia multiseta, Euphorbia muscicola, Euphorbia neomexicana, Euphorbia nephradenia, Euphorbia niqueroana, Euphorbia oaxacana, Euphorbia occidentalis, Euphorbia odontodenia, Euphorbia olivacea, Euphorbia olowaluana, Euphorbia opthalmica, Euphorbia ovata, Euphorbia pachypoda, Euphorbia pachyrhiza, Euphorbia padifolia, Euphorbia palmeri, Euphorbia paludicola, Euphorbia parciflora, Euphorbia parishii, Euphorbia parryi, Euphorbia paxiana, Euphorbia pediculifera, Euphorbia peplidion, Euphorbia peploides, Euphorbia peplus, Euphorbia pergamena, Euphorbia perlignea, Euphorbia petaloidea, Euphorbia petaloidea, Euphorbia petrina, Euphorbia picachensis, Euphorbia pilosula, Eiphorbia pilulifera, Euphorbia pinariona, Euphorbia pinetorum, Euphorbia pionosperma, Euphorbia platysperma, Euphorbia plicata, Euphorbia poeppigii, Euphorbia poliosperma, Euphorbia polycarpa, Euphorbia polycnemoides, Euphorbia polyphylla, Euphorbia portoricensis, Euphorbia portulacoides Euphorbia portulana, Euphorbia preslii, Euphorbia prostrata, Euphorbia pteroneura, Euphorbia pycnanthema, Euphorbia ramosa, Euphorbia rapulum, Euphorbia remyi, Euphorbia r etroscabra, Euphorbia revoluta, Euphorbia rivularis, Euphorbia robusta, Euphorbia romosa, Euphorbia rubida, Euphorbia rubrosperma, Euphorbia rupicola, Euphor bia sanmartensis, Euphorbia saxatilis M. Bieb, Euphorbia schizoloba, Euphorbia sclerocyathium, Euphorbia scopulorum, Euphorbia senilis, Euphorbia serpyllifolia, Euphorbia serrula, Euphorbia setiloba Engelm, Euphorbia sonorae, Euphorbia soobyi, Euphorbia sparsiflora, Euphorbia sphaerosperma, Euphorbia syphilitica, Euphorbia spruceana, Euphorbia subcoerulea, Euphorbia stellata, Euphorbia submammilaris, Euphorbia subpeltata, Euphorbia subpubens, Euphorbia subreniforme, Euphorbia subtrifoliata, Euphorbia succedanea, Euphorbia tamaulipasana, Euphorbia telephioides, Euphorbia tenuissima, Euphorbia tetrapora, Euphorbia tirucalli, Euphorbia tomentella, Euphorbia tomentosa, Euphorbia torralbasii, Euphorbia tovariensis, Euphorbia trachysperma, Euphorbia tricolor, Euphorbia troyana, Euphorbia tuerckheimii, Euphorbia turczaninowii, Euphorbia umbellulata, Euphorbia undulata, Euphorbia vermiformis, Euphorbia versicolor, Euphorbia villifera, Euphorbia violacea, Euphorbia whitei, Euphorbia xanti Engelm, Euphorbia xylopoda Greenm., Euphorbia yayalesia Urb., Euphorbia yungasensis, Euphorbia zeravschanica and Euphorbia zinniiflora.
  • Particularly preferred species of the genus Synadenium include [0047] Synadenium grantii and Synadenium compactum.
  • Particularly preferred species of the genus Monadenium include [0048] Monadenium lugardae and Monadenium guentheri.
  • A preferred species of the genus Endadenium is [0049] Endadenium gossweileni.
  • [0050] Euphorbia peplus is particularly useful in the practice of the present invention. Reference herein to “Euphorbia peplus” or its abbreviation “E. peplus” includes various varieties, strains, lines, hybrids or derivatives of this plant as well as its botanical or horticultural relatives. Furthermnore, the present invention may be practiced using a whole Euphorbiaceae plant or parts thereof including sap or seeds or other reproductive material may be used. Generally, for seeds or reproductive material to be used, a plant or plantlet is first required to be propagated.
  • Reference herein to a Euphorbiaceae plant, a Euphorbia species or [0051] E. peplus further encompasses genetically modified plants. Genetically modified plants include trangenic plants or plants in which a trait has been removed or where an endogenous gene sequence has been down-regulated, mutated or otherwise altered including the alteration or introduction of genetic material which exhibits a regulatory effect on a particular gene. Consequently, a plant which exhibits a character not naturally present in a Euphorbiaceae plant or a species of Euphorbia or in E. peplus is nevertheless encompassed by the present invention and is included within the scope of the above-mentioned terms.
  • The macrocyclic diterpenes are generally in extracts of the Euphorbiaceae plants. An extract may comprise, therefore, sap or liquid or semi-liquid material exuded from, or present in, leaves, stem, flowers, seeds, bark or between the bark and the stem. Most preferably, the extract is from sap. Furthermore, the extract may comprise liquid or semi-liquid material located in fractions extracted from sap, leaves, stems, flowers, bark or other plant material of the Euphoriaceae plant. For example, plant material may be subject to physical manipulation to disrupt plant fibres and extracellular matrix material and inter- and intra-tissue extracted into a solvent including an aqueous environment. All such sources of the macrocyclic diterpenes are encompassed by the present invention including macrocyclic diterpenes obtained by synthetic routes. [0052]
  • The preferred macrocyclic diterpenes are selected from compounds of the ingenane, pepluane and jatrophane families. A compound is stated to be a member of the ingenane, pepulane or jatrophane families on the basis of chemical structure and/or chemical or physical properties. A compound which is a derivative of an ingenane, pepluane or jatrophane is nevertheless encompassed by the present invention through use of the terms “ingenane”, “pepluane” or “jatrophane” since these terms include derivatives, chemical analogues and chemically synthetic forms of these families of compounds. One particularly preferred derivative is an angeloyl derivative of ingenane. [0053]
  • The preferred chemical agent of the present invention is one which exhibits an effect on PKC. Such an effect may be a direct activation or inhibition of PKC activity or a direct effect on the levels of PKC enzyme in a cell or exported from a cell. Furthermore, the effect may be transitory or may involve an initial activation of PKC activity or PKC enzyme synthesis or induction of a functional conformation followed by a down-regulation of PKC activity, enzyme levels or formation of a deactivated conformation. Consequently, an effect on PKC is regarded herein as a modulatory effect and is conveniently determined by consequential events such as resulting from altered signal transduction. For example, activation of latent virus, activation of immune mechanisms or activation of a gene promoter may occur and this is regarded herein as a modulatory effect on PKC. [0054]
  • The chemical agents of the present invention may be in purified or isolated form meaning that the preparation is substantially devoid of other compounds or contaminating agents other than diluent, solvent or carrier or isoforms of the agents. Furthermore, the term “chemical agent” includes preparations of two or more compounds either admixed together or co-purified from a particular source. The chemical agent may also be a chemical fraction, extract or other preparation from the Euphorbiaceace plant. [0055]
  • Consequently, reference herein to a “chemical agent” includes a purified form of one or more compounds or a chemical fraction or extract such as from the sap of a Euphorbiaceace plant, and in particular a species of Euphorbia, and most preferably from [0056] E. peplus or botanical or horticultural relatives or variants thereof.
  • Accordingly, one aspect of the present invention contemplates a method for the treatment or prophylaxis of a PKC-related condition or disorder in a subject, said method comprising the administration to said subject of a symptom-ameliorating effective amount of a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or chemical analogue thereof which chemical agent is a macrocyclic diterpene selected from compounds of the ingenane, pepluane and jatrophane families and which chemical agent or derivative or chemical analogue is represented by any one of the general formulae (I)-(V) [0057]
    Figure US20030195168A1-20031016-C00001
  • wherein: [0058]
  • n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides; [0059]
  • A-T are independently selected from hydrogen, R[0060] 1, R2, R3, F, Cl, Br, I, CN, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2, (C═X)R3 or X(C═X)R3 where X is selected from sulfur, oxygen and nitrogen;
  • R[0061] 1 and R2 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2]alkyl;
  • R[0062] 3 is selected from R1, R2, CN, COR1, CO2R1, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2;
  • A connected to B (or C), D (or E, R (or Q), P (or O) or S (or T) is a selection of C[0063] 1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic rings further substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
  • J connected to I (or H), G (or F), K (or L), M (or N) or S (or T) is a selection of C[0064] 1-C8 disubstituted (fused) saturated and unsaturated carbocyclic or heterocyclic rings further substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
  • D (or E) connected to B (or C) or G (or F); I (or H) connected to G (or F); P (or O) connected to R (or Q) or M (or N); K (or L) connected to N (or M) is a selection of C[0065] 1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic rings substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
  • B and C, D and E, R and Q, P and O, I and H, G and F, K and L, M and N or S and T are ═X where X is selected from sulfur, oxygen, nitrogen, NR[0066] 1R2, and ═CR1R2
    Figure US20030195168A1-20031016-C00002
  • wherein: [0067]
  • n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides; [0068]
  • A′-T′ are independently selected from hydrogen, R[0069] 4, R5, R6, F, Cl, Br, I, CN, COR4, CO2R4, OR4, SR4, NR4R5, CONR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, S02NR4R5, S03NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(R4)2, (C═X)R6 or X(C═X)R6 where X is selected from sulfur, oxygen and nitrogen;
  • R[0070] 4 and R5 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR4, SR4, NR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, SO2NR4R5, SO3NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(P4)2]alkyl;
  • R[0071] 6 is selected from R4, R5, CN, COR4, CO2R4, OR4, SR4, NR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, SO2NR4R5, SO3NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(R4)2;
  • E′ and R′ or H′ and O′ is a C[0072] 2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R6, including epoxides and thioepoxides;
  • O′ connected to M′ (or N′) or Q′ (or P′); R′ connected to Q′ (or P′) or S′ (or T′); S′ (or T′) connected to A′ (or B′); A′ (or B′) connected to C′ (or D′); E′ connected to C′ (or D′) or F′ (or G′); H′ connected to I′; I′ connected to J′; J′ connected to K′; K′ connected to L′; L′ connected to M′ (or N′) are C[0073] 1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R6, (C═X)R6 and X(C═X)R6, including epoxides and thioepoxides;
  • A′, B′ and C′, D′ and F′, G′ and M′, N′ and P′, Q′ and S′, T′ are ═X where X is selected from sulfur, oxygen, nitrogen, NR[0074] 4R5, (C═X)R6, X(C═X)R6, and ═CR7R8;
  • R[0075] 7 and R8 are each independently selected from R6, (C═X)R6 and X(C═X)R6
    Figure US20030195168A1-20031016-C00003
  • wherein: [0076]
  • n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides; [0077]
  • A[0078] 1-T1 are independently selected from hydrogen, R9, R10, R11, F, Cl, Br, I, CN, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2, (C═X)R11 or X(C═X)R11 where X is selected from sulfur, oxygen and nitrogen;
  • R[0079] 9 and R10 are each independently selected from C1-C20 alkyl (branched and straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and straight chained), C2-C10 alkynyl (branched and straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2]alkyl;
  • R[0080] 11 is selected from R9, R10, CN, COR9, CO2R9, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2;
  • B[0081] 1 and R1, E1 and Ö1 and Ë1 and M1 are selected from a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R11, including epoxides and thioepoxides;
  • A[0082] 1 (or Ä1 connected to Á1 (or Ã1) or T1 (or S1) B1 connected to Á1 (or Ã1) or C1 (or D1) E1 connected to Ë1 or C1 (or D1); Ë1 connected to É1 (or F1); G1 (or H1) connected to É1 (or F1) or I1 (or J1); K1 (or L1) connected to I1 (or J1) or M1; M1 connected to O1 (or N1); Ö1 connected O1 (or N1) or P1 (or Q1); R1 connected P1 (or Q1) or S1 (or T1) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R11, (C═X)R11 and X(C═X)R11, including epoxides and thioepoxides;
  • A[0083] 1, Ä and Á, Ã and C1, D1 and F1, É and G1, H1 and I1, J1 and K1, L1 and N1, O1 and P1, Q1 and S1, T1 are ═X where X is selected from sulfur, oxygen, nitrogen, NR9R10, including (C═X)R11 and X(C═X)R11, and ═CR12R13;
  • R[0084] 12 and R13 are independently selected from R11, (C═X)R11 and X(C═X)R11
    Figure US20030195168A1-20031016-C00004
  • wherein: [0085]
  • n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides; [0086]
  • A[0087] 2-X2 are independently selected from hydrogen, R14, R15, R16, F, Cl, Br, I, CN, OR14, SR14, NR14R15, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14), (C═Y)R16 or Y(C═Y)R16 where Y is selected from sulfur, oxygen and nitrogen;
  • R[0088] 14 and R15 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR14, SR14, NR14R10, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14)2]alkyl;
  • R[0089] 16 is selected from R14, R15, CN, COR14, CO2R15, OR14, SR14, NR14R15, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14)2;
  • E[0090] 2 and V2, H2 and S2, and I2 and P2 are C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R16, including epoxides and thioepoxides;
  • A[0091] 2 (or B2) connected to C2 (or D2) or W2 (or X2); E2 connected to C2 (or D2) or F2 (or G2); H2 connected to F2 (or G2) or I2; I2 connected to J2 (or K2); L2 (or M2) connected to J2 (or K2) or N2 (or O2); R2 (or Q2) connected to P2 or S2; V2 connected to U2 (or T2) or W2 (or X2) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R16, (C═Y)R16 and Y(C═Y)R16, including epoxides and thioepoxides;
  • A[0092] 2, B2; C2, D2; F2, G2; J2, K2; L2, M2; N2, O2; Q2, R2; U2, T2 and X2, W2 are ═Y where Y is selected from sulfur, oxygen, nitrogen, NR14R15 and ═CR17R18;
  • R[0093] 17 and R18 are independently selected from R16, (C═Y)R16 and Y(C═Y)R16
    Figure US20030195168A1-20031016-C00005
  • wherein: [0094]
  • n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides; [0095]
  • A[0096] 3-Z3 are independently selected from hydrogen, R19, R20, R21, F, Cl, Br, I, CN, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R,9)3, P(O)(R19)3, Si(R19)3, B(R19)2, (C═Ø)R21 or Ø(C═Ø)R21 where Ø is sullur, oxygen and nitrogen;
  • R[0097] 19 and R20 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R19)3, P(═O)(R19)3, Si(R19)3, B(R19)2]alkyl;
  • R[0098] 21 is selected from R19, R20, CN, COR19, CO2R19, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R19)3, P(═O)(R19)3, Si(R19)3, B(R19)2;
  • D[0099] 3 connected to X3 is a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R21, including epoxides and thioepoxides;
  • A[0100] 3 (or Ä3) connected to B3 (or C3) or E3 (or X); D3 connected to B3 (or C3) or E3 (or F3); G3 (or H3) connected to E3 (or F3) or I3 (or J3); L3 (or K3) connected to I3 (or J3) or M3 (or N3); O3 (or Ö3) connected to N3 (or M3) or P3 (or Q3). S3 (or R3) connected to Q3 (or P3) or U3 (or T3). W3 (or V3) connected to U3 (or T3) or X3; X3 connected to Y3 (or Z3) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R21, (C═Ø)R21 and Ø (C═Ø)R21, including epoxides and thioepoxides;
  • A[0101] 3, Ä3; B3, C3; E3, F3; G3, H3; I3, J3; K3, L3; M3, N3; O3, Ö3; Q3, P3, S3, R3, U3, T3, W3, V3, and Z3, Y3 are ═Ø where Ø is selected from sulfur, oxygen, nitrogen, NR19R20, and ═CR22R23; and
  • R[0102] 22 and R23 are selected from R21, (C═Ø)R21 and Ø(C═Ø)R21;
  • and which chemical agent or derivative or chemical analogue thereof is capable of modulating PKC activity, PKC-gene expression or PKC enzyme turnover and wherein said chemical agent or its derivatives or chemical analogues is administered for a time and under conditions sufficient to ameliorate one or more symptoms associated with a PKC-related condition or disorder. [0103]
  • Especially preferred chemical agents or derivatives or chemical analogues thereof are represented by the general formula (VI): [0104]
    Figure US20030195168A1-20031016-C00006
  • wherein: [0105]
  • R[0106] 24, R25 and R26 are independently selected from hydrogen, R27, R28, F, Cl, Br, I, CN, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, SO2R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2, (C═X)R29 or X(C═X)R29 where X is selected from sulfur, oxygen and nitrogen;
  • R[0107] 27 and R28 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, SO2R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2]alkyl;
  • R[0108] 29 is selected from R27, R28, CN, COR27, CO2R27, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, S02R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2.
  • In a preferred embodiment, R[0109] 24 is hydrogen, OAcetyl or OH.
  • In another preferred embodiment, R[0110] 25 and R26 are OH.
  • As used herein, the term “alkyl” refers to linear or branched chains. The term “haloalkyl” refers to an alkyl group substituted by at least one halogen. Similarly, the term “haloalkoxy” refers to an alkoxy group substituted by at least one halogen. As used herein the term “halogen” refers to fluorine, chlorine, bromine and iodine. [0111]
  • As used herein the term “aryl” refers to aromatic carbocyclic ring systems such as phenyl or naphthyl, anthracenyl, especially phenyl; Suitably, aryl is C[0112] 6-C14 with mono, di- and tri- substitution containing F, Cl, Br, I, NO2, CF3, CN, OR1, COR1, CO2R1, NHR1, NR1R2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2, wherein R1 and R2 are defined above
  • As used herein the terms “heterocycle”, “heterocyclic”, “heterocyclic systems” and the like refer to a saturated, unsaturated, or aromatic carbocyclic group having a single ring, multiple fused rings (for example, bicyclic, tricyclic, or other similar bridged ring systems or substituents), or multiple condensed rings, and having at least one heteroatom such as nitrogen, oxygen, or sulfur within at least one of the rings. This term also includes “heteroaryl” which refers to a heterocycle in which at least one ring is aromatic. Any heterocyclic or heteroaryl group can be unsubstituted or optionally substituted with one or more groups, as defined above. Further, bi- or tricyclic heteroaryl moieties may comprise at least one ring, which is either completely, or partially, saturated. Suitable heteroaryl moieties include, but are not limited to oxazolyl, thiazaoyl, thienyl, furyl, 1-isobenzofuranyl, 3H-pyrrolyl, 2H-pyrrolyl, N-pyrrolyl, imidazolyl, pyrazolyl, isothiazolyl, isooxazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyradazinyl, indolizinyl, isoindolyl, indoyl, indolyl, purinyl, phthalazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,3-oxadiazoyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, 1,2,3,4-oxatriazolyl, 1,2,3,5-oxatriazolyl, 1,3,5-triazinyl, 1,2,4-triazinyl, 1,2,3-triazinyl, azepinyl, oxepinyl, thiepinyl, benzofuranyl, isobenzofuranyl, thionaphthenyl, isothionaphthenyl, indoleninyl, 2-isobenzazolyl, 1,5-pyrindinyl, pyrano[3,4-b]pyrrolyl, isoindazolyl, indoxazinyl, benzoxazolyl, anthranilyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, naphthyridinyl, pyrido[3,4-b]pyridinyl, and pyrido[3,2-b]pyridinyl, pyrido[4,3-b]pyridinyl. [0113]
  • Preferred PKC-related conditions include alcoholism, Alzheimer's disease, asthma, atherosclosis, atopic dermatitis, autoimmune disease, bipolar-disorder, blood disorder, cardiac hypertrophy, depression, diabetes, hypertension, hyperplastic dermatosis, multiple sclerosis, myocardial ischemia, osteoarthritis, psoriasis, rheumatoid arthritis, transplantation and a latent virus. The instant chemical agents are also useful in promoting gene activation and promotion. [0114]
  • The subject chemical agents are also useful in modulating and more particularly promoting or activating gene expression or the expression or operation of a promoter or other regulatory sequence. This aspect is useful for gene therapy, gene augmentation or gene replacement therapy or the provision of expression products (e.g. peptides, polypeptides, proteins, antisense RNA, sense RNA) by endogenous genes or exogenously introduced DNA or RNA. [0115]
  • The subject compounds are also useful in activating latent viruses. [0116]
  • The term “viruses” is used in its broadest sense to include viruses of the families adenoviruses, papovaviruses, herpesviruses: simplex, varicella-zoster, Epstein-Barr, CMV, pox viruses: smallpox, vaccinia, hepatitis B, rhinoviruses, hepatitis A, poliovirus, rubellavirus, hepatitis C, arboviruses, rabiesvirus, influenzaviruses A and B, measlesvirus, mumpsvirus, HIV, HTLV I and II. [0117]
  • Particularly preferred viruses are HIV, EBV and CMV. [0118]
  • Accordingly, another aspect of the present invention provide a method for the treatment or prophylaxis of a PKC-related condition or disorder in a subject, said method comprising the administration to said subject of a symptom-ameliorating effective amount of a macrocyclic diterpene, or a chemical fraction comprising same from a plant of the family Euphorbiaceae or a derivative or chemical analogue of said macrocyclic diterpene having the structures as defined above wherein said macrocyclic diterpene or its derivative or chemical analogue modulates PKC activity, synthesis or enzyme turnover, said administration being for a time and under conditions sufficient to ameliorate one or more symptoms of associated with a PKC-related condition or disorder. [0119]
  • This aspect of the present invention is particularly useful in the treatment of latent virus infection. The term “latent virus” includes reference to a virus or more particularly a virus genome or part thereof which has integrated into the genome of a cell. When in the latent state, it is more difficult for a host's immune system to recognize a virus as a foreign body. In accordance with the present invention, it is proposed that the subject chemical agents are capable of activating a latent virus thereby causing the virus to undergo replication and at least partial assembly. As a result, a mechanism within the host or within the cells of the host is then induced to assist in the eradication of the virus. The present invention extends to both the direct effect of the chemical agent on the virus as well as promoting the immune system to direct same against the virus. [0120]
  • Accordingly, another aspect of the present invention contemplates a method for the treatment or prophylaxis of a PKC-related condition or disorder relating to a latent virus in a subject, said method comprising the administration to said subject of a virus-activating effective amount of a macrocyclic diterpene, or a chemical fraction comprising same from a plant of the family Euphorbiaceae or a derivative or chemical analogue of said macrocyclic diterpene having the structures as defined above wherein said macrocyclic diterpene or its derivative or chemical analogue modulates PKC activity, synthesis or enzyme turnover, said administration being for a time and under conditions sufficient to activate said virus and to combination therapies with antiviral agents. [0121]
  • The activated virus is then destroyed or removed by the host's own immune system and/or by the effects of the agents themselves. [0122]
  • In a preferred embodiment, the method further comprises the simultaneous, sequential or separate administration of an ancillary agent that destroys or attenuates a replicating virus, in combination with the macrocyclic diterpene or chemical fraction. Thus, the invention contemplates combination therapies in which the macrocyclic diterpene or chemical fraction activates a latent virus to thereby cause the virus to undergo replication and the ancillary agent eradicates the replicating virus. [0123]
  • Accordingly, in another aspect, the invention encompasses a method for the treatment or prophylaxis of a PKC-related condition or disorder relating to a latent virus in a subject, said method comprising simultaneously, sequentially or separately administrating to said subject a virus-activating effective amount of a macrocyclic diterpene, or a chemical fraction comprising same from a plant of the family Euphorbiaceae or a derivative or chemical analogue of said macrocyclic diterpene having the structures as defined above wherein said macrocyclic diterpene or its derivative or chemical analogue modulates PKC activity, synthesis or enzyme turnover, together with a virus-destroying or -attenuating effective amount of an ancillary agent which destroys or attenuates a replicating form of said virus, said administration being for a time and under conditions sufficient for said macrocyclic diterpene or chemical fraction to activate said virus to thereby cause the virus to undergo replication and for said ancillary agent to destroy or attenuate said replicating virus. [0124]
  • In one embodiment, the latent virus is HIV. A range of HIV specific agents may be used for the destruction or attenuation of this virus, including, for example, those described in Matsuhita et al., 2000. Preferred HIV specific ancillary agents include, for example, nucleoside analogues such as combivir, epivir, hivid, retrovir, videx, zerat and zygen, non-nucleoside agents such as rescriptor, sustiva and viramune, adjunctive anti-retrovirals such as hydrea and droxa, and protease inhibitors such as agenerase, fortovase, crixivan, invirase, norvir and virasept. [0125]
  • In another embodiment, the latent virus is EBV. Preferred EBV specific ancillary agents are selected from ganciclovir (GVC) or 3′-azido-3′deoxythymidine (AZT). [0126]
  • In yet another embodiment, the latent virus is CMV. A preferred CMV specific ancillary agent is cidofovir. [0127]
  • Particularly useful compounds include 5,8,9,10,14-pentaacetoxy-3-benzoyloxy- 15-hydroxypepluane (pepluane), derivatives of said pepluane, jatrophanes of Conformation II including 2,3,5,7,15-pentaacetoxy-9-nicotinoyloxy-14-oxojatropha-6(17),11E-diene kjatrophane 1), derivatives of said jatrophane 1,2,5,7,8,9,14-hexaacetoxy-3-benzoyloxy-15-hydroxy-jatropha-6(17),11E-diene (jatrophane 2), derivatives of said jatrophane 2, 2,5,14-triacetoxy-3-benzoyloxy-8,15-dihydroxy-7-isobutyroyloxy-9-nicotinoyloxy-jatropha-6(17),11E-diene (jatrophane 3), derivatives of said jatrophane 3, 2,5,9,14-tetraacetoxy-3-benzoyloxy-8,15-dihydroxy-7-isobutyroyloxyjatropha-6(17),11E-diene) (jatrophane 4), derivatives of said jatrophane 4, 2,5,7,14-tetraacetoxy-3-benzoyloxy-8,15-dihydroxy-9-nicotinoyloxyjatropha-6(17), 11E-diene (jatrophane 5), derivatives of said jatrophane 5, 2,5,7,9,14-pentaacetoxy-3-benzoyloxy-8,15-dihydroxyjatropha-6( 17), 11E-diene jatrophane 6), derivatives of said jatrophane 6, or pharmaceutically acceptable salts of these. [0128]
  • Even more particularly preferred compounds are angeloyl substituted ingenanes or derivatives thereof such as ingenol-3-angelate, 20-hydroxy-ingenol-3-angelate, 20-O-acetyl-ingenol-3-angelate, or derivatives of said angelates, or pharmaceutically acceptable salts of these. [0129]
  • Still a further aspect of the present invention contemplates a method of assessing the suitability of a chemical agent from Euphorbiaceae for the practice of the present invention. Numerical values are assigned to chemical agents including fractions comprising the chemical agents as set forth, for example, in Table A: [0130]
    TABLE A
    Feature Value
    An ability to modulate PKC activity or effect +1
    An ability to induce bipolar dendritic activity +1
    An ability to displace phorbol dibutyrate from +1
    binding to PKC
    An ability to induce respiratory burst in leucocytes +1
    An ability to stimulate phagocytosis in peripheral +1
    blood mononuclear cells
    Derived from a member of the Euphorbiaeae family +1
    Derived from E. peplus +3
    Water extractible from the sap of Euphorbia sp. +2
    An ability to activate latent virus +4
    A lower tumor promotion activity than TPA/PMA + 2
  • The value for each feature is referred to as the Index Value (I[0131] V).
  • The sum of I[0132] V, i.e. ΣIV, provides a potency of agent (PA) value and this enables an analytical approach to screening and selecting compounds from Euphorbiaceae useful in the practice of the present invention.
  • In one example, 20-acetyl-ingenol-3 angelate exhibits a P[0133] A=ΣIV=15.
  • Accordingly, another aspect of the present invention contemplates a method for the treatment or prophylaxis of a PKC-related condition in a subject, said method comprising administration to said subject of a symptom-ameliorating effective amount of a macrocyclic diterpene obtainable from a Euphorbiaceae plant or its botanical or horticultural relative, said macrocyclic diterpene being selected from an ingenane, pepluane or jatrophane, or a derivative or chemical analogue thereof, having the structure represented by any one of the general formulae (I)-(V) as defined above and wherein said chemical agent exhibits a potency of agent (P[0134] A) of >10, wherein the PA=ΣIV where IV is a numerical value associated with a particular feature as listed below:
    Feature Value
    An ability to modulate PKC activity or effect +1
    An ability to induce bipolar dendritic activity +1
    An ability to displace phorbol dibutyrate from +1
    binding to PKC
    An ability to induce respiratory burst in leucocytes +1
    An ability to stimulate phagocytosis in peripheral +1
    blood mononuclear cells
    Derived from a member of the Euphorbiaceae family +1
    Derived from E. peplus +3
    Water extractible from the sap of Euphorbia sp. +2
    An ability to activate latent virus +4
    A lower tumor promotion activity than TPA/PMA + 2
  • or pharmaceutically acceptable salts of these, said chemical agent being administered for a time and under conditions sufficient to ameliorate at least one symptom caused by or associated with the biological entity. [0135]
  • Preferred compounds are selected from the list comprising: [0136]
  • 5,8,9,10,14-pentaacetoxy-3-benzoyloxy-15-hydroxypepluane (pepluane); [0137]
  • 2,3,5,7,15-pentaacetoxy-9-nicotinoyloxy-14-oxojatropha-6(17),11E-diene (jatrophane 1); [0138]
  • 2,5,7,8,9,14-hexaacetoxy-3-benzoyloxy-15-hydroxy-jatropha-6(17),11E-diene (jatrophane 2); [0139]
  • 2,5,14-triacetoxy-3-benzoyloxy-8,15-dihydroxy-7-isobutyroyloxy-9-nicotinoyloxy-jatropha-6(17),11E-diene (jatrophane 3); [0140]
  • 2,5,9,14-tetraacetoxy-3-benzoyloxy-8,15-dihydroxy-7-isobutyroyloxy jatropha-6(17),11E-diene) (jatrophane 4); [0141]
  • 2,5,7,14-tetraacetoxy-3-benzoyloxy-8,15-dihydroxy-9-nicotinoyloxy-jatropha-6(17),11E-diene (jatrophane 5); [0142]
  • 2,5,7,9,14-pentaacetoxy-3-benzoyloxy-8,15-dihydroxyjatropha-6(17),11E-diene (jatrophane 6); [0143]
  • 20-O-acetyl-ingenol-3-angelate, derivatives of 20-O-acetyl-ingenol-3-angelate. [0144]
  • 20-hydroxy-ingelol-3-angelate, derivatives of 20-hydroxy-ingenol-3-angelate; and [0145]
  • ingenol-3-angelate, derivatives of ingenol-3-angelate. [0146]
  • Preferably, the biological entity is a microorganism, virus, yeast, fungus, insect, arachnid or Plasmodium sp. [0147]
  • Reference herein to a subject includes a human, primate, livestock animal (e.g. sheep, cow, horse, pig, goat, donkey), laboratory test animal (e.g. mouse, rat, guinea pig, hamster), companion animal (e.g. dog, cat) or avian species such as poultry birds (e.g. chicken, ducks, turkeys, geese) or game birds (e.g. arid ducks, pheasants). [0148]
  • The preferred subject is a human or primate or laboratory test animal. [0149]
  • The most preferred subject is a human. [0150]
  • The ability to assign numerical values to certain characteristics enables data processing means to assess the likely usefulness of a particular compound or group of compounds forming a chemical agent. [0151]
  • The assessment of the suitability of a compound or group of compounds for the practice of the present invention is suitably facilitated with the assistance of a computer programmed with software, which inter alia adds index values (I[0152] V) for at least two features associated with the compound(s) to provide a potency value (PA) corresponding to the effectiveness of the compound(s) for treating or preventing infection or colonization or presence of a biological entity in a subject. The compound features can be selected from:
  • (a) the ability to modulate PKC activity or effect; [0153]
  • (b) the ability to induce bipolar dendritic activity; [0154]
  • (c) the ability to be derived from a member of the Euphorbiaceae family; [0155]
  • (d) the ability to be derived from [0156] E. peplus;
  • (e) the ability to be water extractable from the sap of a Euphorbia species; [0157]
  • (f) the ability to activate latent virus; or [0158]
  • (g) less tumor promoting capacity than TPA or MPA. [0159]
  • Accordingly, in accordance with the present invention, index values for such features are stored in a machine-readable storage medium, which is capable of processing the data to provide a potency value for a compound or group of compounds of interest. [0160]
  • Thus, in another aspect, the invention contemplates a computer program product for assessing the likely usefulness of a candidate compound or group of compounds for treating or preventing a PKC-related condition or disorder in a subject, said product comprising: [0161]
  • (1) code that receives as input index values for at least two features associated with said compound(s), wherein said features are selected from: [0162]
  • (a) the ability to modulate PKC activity or effect; [0163]
  • (b) the ability to induce bipolar dendritic activity; [0164]
  • (c) the ability to be derived from a member of the Euphorbiaceae family; [0165]
  • (d) the ability to be derived from [0166] E. peplus;
  • (e) the ability to be water extractable from the sap of a Euphorbia species; [0167]
  • (f) the ability to activate latent virus; or [0168]
  • (g) less tumor promoting capacity than TPA or MPA; [0169]
  • (2) code that adds said index values to provide a sum corresponding to a potency value for said compound(s); and [0170]
  • (3) a computer readable medium that stores the codes. [0171]
  • In a preferred embodiment, the computer program product comprises code that assigns an index value for each feature of a compound or group of compounds. In an especially preferred embodiment, index values are assigned as set forth in Table A above. [0172]
  • In a related aspect, the invention extends to a computer for assessing the likely usefulness of a candidate compound or group of compounds for treating or preventing a PKC-related condition or disorder in a subject, wherein said computer comprises: [0173]
  • (1) a machine-readable data storage medium comprising a data storage material encoded with machine-readable data, wherein said machine-readable data comprise index values for at least two features associated with said compound(s), wherein said features are selected from: [0174]
  • (a) the ability to modulate PKC activity or effect; [0175]
  • (b) the ability to induce bipolar dendritic activity; [0176]
  • (c) the ability to be derived from a member of the Euphorbiaceae family; [0177]
  • (d) the ability to be derived from [0178] E. peplus;
  • (e) the ability to be water extractable from the sap of a Euphorbia species; [0179]
  • (f) the ability to activate latent virus; or [0180]
  • (g) less tumor promoting capacity than TPA or MPA; [0181]
  • (2) a working memory for storing instructions for processing said machine-readable data; [0182]
  • (3) a central-processing unit coupled to said working memory and to said machine-readable data storage medium, for processing said machine readable data to provide a sum of said index values corresponding to a potency value for said compound(s); and [0183]
  • (4) an output hardware coupled to said central processing unit, for receiving said potency value. [0184]
  • A version of these embodiments is presented in FIG. 8, which shows a [0185] system 10 including a computer 11 comprising a central processing unit (“CPU”) 20, a working memory 22 which may be, e.g. RAM (random-access memory) or “core” memory, mass storage memory 24 (such as one or more disk drives or CD-ROM drives), one or more cathode-ray tube (“CRT”) display terminals 26, one or more keyboards 28, one or more input lines 30, and one or more output lines 40, all of which are interconnected by a conventional bidirectional system bus 50.
  • [0186] Input hardware 36, coupled to computer 11 by input lines 30, may be implemented in a variety of ways. For example, machine-readable data of this invention may be inputted via the use of a modem or modems 32 connected by a telephone line or dedicated data line 34. Alternatively or additionally, the input hardware 36 may comprise CD. Alternatively, ROM drives or disk drives 24 in conjunction with display terminal 26, keyboard 28 may also be used as an input device.
  • Output hardware [0187] 46, coupled to computer 11 by output lines 40, may similarly be implemented by conventional devices. By way of example, output hardware 46 may include CRT display terminal 26 for displaying a synthetic polynucleotide sequence or a synthetic polypeptide sequence as described herein. Output hardware might also include a printer 42, so that hard copy output may be produced, or a disk drive 24, to store system output for later use.
  • In operation, [0188] CPU 20 coordinates the use of the various input and output devices 36, 46 coordinates data accesses from mass storage 24 and accesses to and from working memory 22, and determines the sequence of data processing steps. A number of programs may be used to process the machine readable data of this invention. Exemplary programs may use for example the following steps:
  • (1) inputting input index values for at least two features associated with said compound(s), wherein said features are selected from: [0189]
  • (a) the ability to modulate PKC activity or effect; [0190]
  • (b) the ability to induce bipolar dendritic activity; [0191]
  • (c) the ability to be derived from a member of the Euphorbiaceae family; [0192]
  • (d) the ability to be derived from [0193] E. peplus;
  • (e) the ability to be water extractable from the sap of a Euphorbia species; [0194]
  • (f) the ability to activate latent virus; or [0195]
  • (g) less tumor promoting capacity than TPA or MPA; and [0196]
  • (2) adding the index values for said features to provide a potency value for said compound(s); and (3) outputting said potency value. [0197]
  • FIG. 9 shows a cross section of a magnetic [0198] data storage medium 100 which can be encoded with machine readable data, or set of instructions, for designing a synthetic molecule of the invention, which can be carried out by a system such as system 10 of FIG. 10. Medium 100 can be a conventional floppy diskette or hard disk, having a suitable substrate 101, which may be conventional, and a suitable coating 102, which may be conventional, on one or both sides, containing magnetic domains (not visible) whose polarity or orientation can be altered magnetically. Medium 100 may also have an opening (not shown) for receiving the spindle of a disk drive or other data storage device 24. The magnetic domains of coating 102 of medium 100 are polarized or oriented so as to encode in manner which may be conventional, machine readable data such as that described herein, for execution by a system such as system 10 of FIG. 8.
  • FIG. 10 shows a cross section of an optically readable data storage medium [0199] 110 which also can be encoded with such a machine-readable data, or set of instructions, for designing a synthetic molecule of the invention, which can be carried out by a system such as system 10 of FIG. 8. Medium 110 can be a conventional compact disk read only memory (CD-ROM) or a rewritable medium such as a magneto-optical disk, which is optically readable and magneto-optically writable. Medium 100 preferably has a suitable substrate 111, which may be conventional, and a suitable coating 112, which may be conventional, usually of one side of substrate 111.
  • In the case of CD-ROM, as is well known, coating [0200] 112 is reflective and is impressed with a plurality of pits 113 to encode the machine-readable data. The arrangement of pits is read by reflecting laser light off the surface of coating 112. A protective coating 114, which preferably is substantially transparent, is provided on top of coating 112.
  • In the case of a magneto-optical disk, as is well known, coating [0201] 112 has no pits 113, but has a plurality of magnetic domains whose polarity or orientation can be changed magnetically when heated above a certain temperature, as by a laser (not shown). The orientation of the domains can be read by measuring the polarisation of laser light reflected from coating 112. The arrangement of the domains encodes the data as described above.
  • The present invention further extends to pharmaceutical compositions useful in treating a PKC-related disorder. In this regard, the chemical agents of the invention can be used as actives for the treatment or prophylaxis of a condition associated with the presence of a PKC-related disorder in a subject. The chemical agents can be administered to a patient either by themselves, or in pharmaceutical compositions where they are mixed with a suitable pharmaceutically acceptable carrier. [0202]
  • Accordingly, the invention also provides a composition for treatment and/or prophylaxis of a PKC-related condition or disorder in a subject, comprising one or more chemical agents of the invention, together with a pharmaceutically acceptable carrier and/or diluent. [0203]
  • Depending on the specific conditions being treated, chemical agents may be formulated and administered systemically or locally. Techniques for formulation and administration may be found in “Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, Pa., latest edition. Suitable routes may, for example, include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections. For injection, the chemical agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. Intra-muscular and subcutaneous injection is appropriate, for example, for administration of immunomodulatory compositions and vaccines. [0204]
  • The chemical agents can be formulated readily using pharmaceutically acceptable carriers well known in the art into dosages suitable for oral administration. Such carriers enable the compounds of the invention to be formulated in dosage forms such as tablets, pills, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. These carriers may be selected from sugars, starches, cellulose and its derivatives, malt, gelatine, talc, calcium sulphate, vegetable oils, synthetic oils, polyols, alginic acid, phosphate buffered solutions, emulsifiers, isotonic saline, and pyrogen-free water. [0205]
  • Pharmaceutical compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve their intended purpose. The dose of agent administered to a patient should be sufficient to effect a beneficial response in the patient over time such as a reduction in the symptoms associated with the presence of a biological entity or part thereof or toxin or venom therefrom or a genetic event caused thereby in a subject. The quantity of the agent(s) to be administered may depend on the subject to be treated inclusive of the age, sex, weight and general health condition thereof. In this regard, precise amounts of the agent(s) for administration will depend on the judgement of the practitioner. In determining the effective amount of the chemical agent to be administered in the treatment or prophylaxis of a condition associated with the biological entity, the physician may evaluate tissue or fluid levels of the biological entity, and progression of the disorder. In any event, those of skill in the art may readily determine suitable dosages of the chemical agents of the invention. [0206]
  • Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. [0207]
  • Pharmaceutical preparations for oral use can be obtained by combining the active compounds with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinyl-pyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. Such compositions may be prepared by any of the methods of pharmacy but all methods include the step of bringing into association one or more chemical agents as described above with the carrier which constitutes one or more necessary ingredients. In general, the pharmaceutical compositions of the present invention may be manufactured in a manner that is itself known, e.g. by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilising processes. [0208]
  • Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses. [0209]
  • Pharmaceutical compositions which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. [0210]
  • Dosage forms of the chemical agents of the invention may also include injecting or implanting controlled releasing devices designed specifically for this purpose or other forms of implants modified to act additionally in this fashion. Controlled release of an agent of the invention may be effected by coating the same, for example, with hydrophobic polymers including acrylic resins, waxes, higher aliphatic alcohols, polylactic and polyglycolic acids and certain cellulose derivatives such as hydroxypropyhnethyl cellulose. In addition, controlled release may be effected by using other polymer matrices, liposomes and/or microspheres. [0211]
  • Chemical agents of the invention may be provided as salts with pharmaceutically compatible counterions. Pharmaceutically compatible salts may be formed with many acids, including but not limited to hydrochloric, sulphuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents that are the corresponding free base forms. [0212]
  • For any chemical agent used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. For example, a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC50 as determined in cell culture (e.g. the concentration of a test agent, which achieves a half-maximal inhibition of infection or colonization or presence of a biological entity). Such information can be used to more accurately determine useful doses in humans. [0213]
  • Toxicity and therapeutic efficacy of such chemical agents can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g. for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds that exhibit large therapeutic indices are preferred. The data obtained from these cell culture assays and animal studies can be used in formulating a range of dosages for use in humans. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition (see for example Fingl et al., 1975, in “The Pharmacological Basis of Therapeutics”, Ch. 1 p1). [0214]
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active agent which are sufficient to maintain symptom-ameliorating effects. Usual patient dosages for systemic administration range from 1-2000 mg/day, commonly from 1-250 mg/day, and typically from 10-150 mg/day. Stated in terms of patient body weight, usual dosages range from 0.02-25 mg/kg/day, commonly from 0.02-3 mg/kg/day, typically from 0.2-1.5 mg/kg/day. Stated in terms of patient body surface areas, usual dosages range from 0.5-1200 mg/m[0215] 2/day, commonly from 0.5-150 mg/m2/day, typically from 5-100 mg/m2/day.
  • Alternately, one may administer the compound in a local rather than systemic manner, for example, via injection of the compound directly into a tissue, often in a depot or sustained release formulation. Furthermore, one may administer the drug in a targeted drug delivery system, for example, in a liposome coated with tissue-specific antibody. The liposomes will be targeted to and taken up selectively by the tissue. In cases of local administration or selective uptake, the effective local concentration of the agent may not be related to plasma concentration. [0216]
  • The chemical agents of the invention can also be delivered topically. For topical administration, a composition containing between 0.001-5% or more chemical agent is generally suitable. Regions for topical administration include the skin surface and also mucous membrane tissues of the vagina, rectum, nose, mouth, and throat. Compositions for topical administration via the skin and mucous membranes should not give rise to signs of irritation, such as swelling or redness. [0217]
  • The topical composition may include a pharmaceutically acceptable carrier adapted for topical administration. Thus, the composition may take the form of a suspension, solution, ointment, lotion, sexual lubricant, cream, foam, aerosol, spray, suppository, implant, inhalant, tablet, capsule, dry powder, syrup, balm or lozenge, for example. Methods for preparing such compositions are well known in the pharmaceutical industry. [0218]
  • In one embodiment, the topical composition is administered topically to a subject, e.g. by the direct laying on or spreading of the composition on the epidermal or epithelial tissue of the subject, or transdermally via a “patch”. Such compositions include, for example, lotions, creams, solutions, gels and solids. Suitable carriers for topical administration preferably remain in place on the skin as a continuous film, and resist being removed by perspiration or immersion in water. Generally, the carrier is organic in nature and capable of having dispersed or dissolved therein a chemical agent of the invention. The carrier may include pharmaceutically-acceptable emolients, emulsifiers, thickening agents, solvents and the like. [0219]
  • The invention also features a process for separating macrocyclic diterpenes from a biomass containing same, said process comprising contacting the biomass with an aqueous solvent for a time and under conditions sufficient to extract the macrocyclic diterpenes into said solvent. [0220]
  • The aqueous solvent is preferably water. [0221]
  • Suitably, the biomass is derived from a plant, which is preferably a member of the Euphorbiaceae family of plants or botanical or horticultural relatives of such plants. Matter from the plant (e.g. foliage, stems, roots, seeds, bark, etc.) is preferably cut, macerated or mulched to increase the surface area of the plant matter for aqueous extraction of the macrocyclic diterpenes. [0222]
  • The process preferably further comprises adsorbing the macrocyclic diterpenes to a non-ionic adsorbent, which is suitably a non-ionic porous synthetic adsorbent. Among the non-ionic porous synthetic adsorbents that can be used for the purposes of the present invention include, but are not restricted to, aromatic copolymers mainly composed of styrene and divinylbenzene, and methacrylic copolymers mainly composed of monomethacrylate and dimethacrylate. Such non-ionic porous synthetic adsorbents which comprise, as the basic structure, aromatic copolymers mainly composed of styrene and divinylbenzene include, for example, Diaion HP10, HP20, HP21, HP30, HP40, HP50, SP850, and SP205 (trade names: Mitsubishi Chemical Corp.), and Amberlite XAD-2, XAD4, (trade names: Rohm and Haas Co.). Examples of non-ionic porous synthetic adsorbent which comprise, as the basic structure, methacrylic copolymer mainly composed of monomethacrylate and dimethacrylate are Diaion HP2MG, Amberlite XAD-7, XAD-8 and XAD-16 and others. [0223]
  • Preferably, the process further comprises eluting macrocyclic diterpenes from the non-ionic adsorbent with water and water-soluble organic solvent(s). [0224]
  • The treatment may be conducted by a batch method using water and water-soluble organic solvent(s) which dissolve macrocyclic diterpenes, or may also be conducted continuously or in batch using a column chromatography method. [0225]
  • Examples of a water-soluble organic solvent which may be used in the present invention are alcohols such as methanol, ethanol, n-propyl alcohol, isopropyl alcohol, and tert-butanol, ethers such as dioxane and tetrahydrofuran, ketones such as acetone, amides such as dimethylformamide, sulfur-containing compounds such as dimethylsulfoxide. Two or more of such organic solvents may be mixed for use. In addition, solvents less soluble in water, for example, alcohols such as n-butanol, esters such as methyl formate and methyl acetate, and ketones such as methyl ethyl ketone may also be used to the extent that it does not separate during development. Particularly preferred water-soluble organic solvents are alcohols, in particular, methanol, ethanol, propyl alcohol, and the like. Furthermore, different kinds of solvent may also be used sequentially for development. [0226]
  • Macrocyclic diterpenes can be further purified using media and techniques which separate compounds on the basis of molecular size and/or polarity. In a preferred embodiment of this type, the macrocyclic diterpenes are separated using Sephadex HL-20 resin and preferably using water and water-soluble organic solvent(s) for development. [0227]
  • The present invention contemplates the use of the chemical agents of the invention for modulating the expression of genes under the control of promoters whose activity is modulated by said chemical agents. Thus, in one embodiment, the chemical agents of the present invention find particular utility in modulating and preferably in promoting or activating the expression of gene therapy reagents (e.g. peptides, polypeptides, proteins, antisense RNA, sense RNA) under the control of such modulatable promoters. [0228]
  • The term “gene therapy” refers generally to the therapeutic replacement, augmenting or silencing of an endogenous gene activity, which is typically effected by gene transfer. [0229]
  • “Gene transfer” or “gene delivery” as used herein refer to methods or systems for reliably inserting exogenous nucleic acids, typically DNA, into host cells. Such methods can result in transient expression of non-integrated transferred nucleic acids, extrachromosomal replication and expression of transferred replicons (e.g. episomes), or integration of transferred genetic material into the genomic DNA of host cells. A number of systems have been developed for gene transfer into mammalian cells. See, e.g., U.S. Pat. No. 5,399,346. [0230]
  • As used herein, “exogenous” means any nucleic acid that is introduced into an organism or cell. [0231]
  • Promoters suitable for the present invention include viral and non-viral promoter, which are preferably inducible. Non-limiting examples of inducible non-viral promoters include metallothionein promoters, and metallothionein-related promoters. Non-limiting examples of inducible viral promoters effective for use in gene transfer vectors of the present invention include, but are not limited to, mouse mammary tumour virus promoters, CMV (cytomegalovirus), SV40 (simian virus 40), RSV (Rous sarcoma virus), HSV (herpes simplex virus), EBV (Epstein-Barr virus), retroviral promoters and adenoviral promoters. [0232]
  • By “vector” is meant any genetic element, such as a plasmid, phage, transposon, cosmid, chromosome, virus, virion, etc., which is capable of replication when associated with the proper control elements and which can transfer gene sequences between cells. Thus, the term includes cloning and expression vehicles, as well as viral vectors. [0233]
  • The ability of certain viruses to infect cells or enter cells via receptor-mediated endocytosis, and to integrate into host cell genome and express viral genes stably and efficiently have made them attractive candidates for the transfer of foreign genes into mammalian cells. Preferred gene therapy vectors of the present invention are viral vectors. [0234]
  • A viral-based vector comprises: (1) nucleic acid of, or corresponding to at least a portion of, the genome of a virus, which portion is capable of directing the expression of a DNA sequence; and (2) a DNA sequence encoding an expression product (e.g. polypeptide or transcript), operably linked to the viral nucleic acid and capable of being expressed as a functional gene product in the target cell. The recombinant viral vectors of the present invention may be derived from a variety of viral nucleic acids known to one skilled in the art, e.g. the genomes of adenovirus, adeno-associated virus, HSV, alphavirus, vaccinia virus, and other viruses, including RNA and DNA viruses. [0235]
  • Particularly preferred viral vectors are those that can accept foreign genetic material and can infect an extended range of cells. Adenoviruses do not integrate their genetic material into the host genome and therefore do not require host replication for gene expression, making them ideally suited for rapid, efficient, heterologous gene expression. Techniques for preparing replication-defective infective viruses are well known in the art. Preferably a viral delivery system including the virion is free of undesirable contaminants, such as defective interfering viral particles or endotoxins and other pyrogens such that it will not cause any inappropriate reactions in the cell, animal or individual receiving the adenoviral vector construct. A preferred means of purifying the vector involves the use of buoyant density gradients, such as cesium chloride gradient centrifugation. [0236]
  • Adenovirus is a particularly preferred gene therapy vector Particularly preferred Adenoviral vector of the present invention include commercially available retroviral expression vectors including but not limited to pLAPSN, pLEGFP-C1, pLEGFP-N1, pLHCX, pLIB, pLNCX, pLNCX2, pLNHX, pLPCX, pLXIN, pLXRN, pLXSN, pMSCVhyg, pMSCVneo, pMSCVpuro, pSIR, and pVSV-G. [0237]
  • The present invention further contemplates the use of Adeno-associated virus (AAV) is vector system for use in the present invention. Details concerning the generation and use of recombinant AAV vectors are described in U.S. Pat. Nos. 5,139,941 and 4,797,368, each incorporated herein by reference. The present invention further contemplates gene therapy using retroviral vectors such as an engineered variant of the Moloney murine leukemia virus. [0238]
  • In another embodiment of the present invention, the viral vector may be derived from other viral vectors such as vaccinia virus, sindbis virus, cytomegalovirus and herpes simplex virus. [0239]
  • The present invention further contemplates the use of gene therapy vectors derived from an RNA virus such as but not limited to the following: members of the family Reoviridae, including the genus Orthoreovirus (multiple serotypes of both mammalian and avian retroviruses), the genus Orbivirus (Bluetongue virus, Eugenangee virus, Kemerovo virus, African horse sickness virus, and Colorado Tick Fever virus), the genus Rotavirus (human rotavirus, Nebraska calf diarrhea virus, murine rotavirus, simian rotavirus, bovine or ovine rotavirus, avian rotavirus); the family Retrovirideae including the genus Lentivirus (Human Immunodeficiency Virus), the family Picomaviridae, including the genus Enterovirus (poliovirus, Coxsackie virus A and B, enteric cytopathic human orphan (ECHO) viruses, hepatitis A virus, Simian enteroviruses, Murine encephalomyelitis (ME) viruses, Poliovirus muris, Bovine enteroviruses, Porcine enteroviruses, the genus Cardiovirus (Encephalomyocarditis virus (EMC), Mengovirus), the genus Rhinovirus (Human rhinoviruses including at least 113 subtypes; other rhinoviruses), the genus Apthovirus (Foot and Mouth disease (FMDV); the family Calciviridae, including Vesicular exanthema of swine virus, San Miguel sea lion virus, Feline picornavirus and Norwalk virus; the family Togaviridae, including the genus Alphavirus (Eastern equine encephalitis virus, Semliki forest virus, Sindbis virus, Chikungunya virus, O'Nyong-Nyong virus, Ross river virus, Venezuelan equine encephalitis virus, Western equine encephalitis virus), the genus Flavirius (Mosquito borne yellow fever virus, Dengue virus, Japanese encephalitis virus, St. Louis encephalitis virus, Murray Valley encephalitis virus, West Nile virus, Kunjin virus, Central European tick borne virus, Far Eastern tick borne virus, Kyasanur forest virus, Louping III virus, Powassan virus, Omsk hemorrhagic fever virus), the genus Rubivirus (Rubella virus), the genus Pestivirus (Mucosal disease virus, Hog cholera virus, Border disease virus); the family Bunyaviridae, including the genus Bunyvirus (Bunyamwera and related viruses, California encephalitis group viruses), the genus Phlebovirus (Sandfly fever Sicilian virus, Rift Valley fever virus), the genus Nairovirus (Crimean-Congo hemorrhagic fever virus, Nairobi sheep disease virus), and the genus Uukuvirus (Uukuniemi and related viruses); the family Orthomyxoviridae, including the genus Influenza virus (Influenza virus type A, many human subtypes); Swine influenza virus, and Avian and Equine Influenza viruses; influenza type B (many human subtypes), and influenza type C (possible separate genus); the family paramyxoviridae, including the genus Paramyxovirus (Parainfluenza virus type 1, Sendai virus, Hemadsorption virus, Parainfluenza viruses types 2 to 5, Newcastle Disease Virus, Mumps virus), the genus Morbillivirus (Measles virus, subacute sclerosing panencephalitis virus, distemper virus, Rinderpest virus), the genus Pneumovirus (respiratory syncytial virus (RSV), Bovine respiratory syncytial virus and Pneumonia virus of mice); forest virus, Sindbis virus, Chikungunya virus, O'Nyong-Nyong virus, Ross river virus, Venezuelan equine encephalitis virus, Western equine encephalitis virus), the genus Flavirius (Mosquito borne yellow fever virus, Dengue virus, Japanese encephalitis virus, St. Louis encephalitis virus, Murray Valley encephalitis virus, West Nile virus, Kunjin virus, Central European tick borne virus, Far Eastern tick borne virus, Kyasanur forest virus, Louping III virus, Powassan virus, Omsk hemorrhagic fever virus), the genus Rubivirus (Rubella virus), the genus Pestivirus (Mucosal disease virus, Hog cholera virus, Border disease virus); the family Bunyaviridae, including the genus Bunyvirus (Bunyamwera and related viruses, California encephalitis group viruses), the genus Phlebovirus (Sandfly fever Sicilian virus, Rift Valley fever virus), the genus Nairovirus (Crimean-Congo hemorrhagic fever virus, Nairobi sheep disease virus), and the genus Uukuvirus (Uukuniemi and related viruses); the family Orthomyxoviridae, including the genus Influenza virus (Influenza virus type A, many human subtypes); Swine influenza virus, and Avian and Equine Influenza viruses; influenza type B (many human subtypes), and influenza type C (possible separate genus); the family paramyxoviridae, including the genus Paramyxovirus (Parainfluenza virus type 1, Sendai virus, Hemadsorption virus, Parainfluenza viruses types 2 to 5, Newcastle Disease Virus, Mumps virus), the genus Morbillivirus (Measles virus, subacute sclerosing panencephalitis virus, distemper virus, Rinderpest virus), the genus Pneumovirus (respiratory syncytial virus (RSV), Bovine respiratory syncytial virus and Pneumonia virus of mice); the family Rhabdoviridae, including the genus Vesiculovirus (VSV), Chandipura virus, Flanders-Hart Park virus), the genus Lyssavirus (Rabies virus), fish Rhabdoviruses, and two probable Rhabdoviruses (Marburg virus and Ebola virus); the family Arenaviridae, including Lymphocytic choriomeningitis virus (LCM), Tacaribe virus complex, and Lassa virus; the family Coronoaviridae, including Infectious Bronchitis Virus (IBV), Mouse Hepatitis virus, Human enteric corona virus, and Feline infectious peritonitis (Feline coronavirus). [0240]
  • The present invention further envisions the use of viral vectors derived from the following DNA viruses including but not limited to the family Poxviridae, including the genus Orthopoxvirus (Variola major, Variola minor, Monkey pox Vaccinia, Cowpox, Buffalopox, Rabbitpox, Ectromelia), the genus Leporipoxvirus (Myxoma, Fibroma), the genus Avipoxvirus (Fowlpox, other avian poxvirus), the genus Capripoxvirus (sheeppox, goatpox), the genus Suipoxvirus (Swinepox), the genus Parapoxvirus (contagious postular dermatitis virus, pseudocowpox, bovine papular stomatitis virus); the family Iridoviridae (African swine fever virus, Frog viruses 2 and 3, Lymphocystis virus of fish); the family Herpesviridae, including the alpha-Herpesviruses (Herpes Simplex Types 1 and 2, Varicella-Zoster, Equine abortion virus, Equine herpes virus 2 and 3, pseudorabies virus, infectious bovine keratoconjunctivitis virus, infectious bovine rhinotracheitis virus, feline rhinotracheitis virus, infectious laryngotracheitis virus) the Beta-herpesviruses (Human cytomegalovirus and cytomegaloviruses of swine, monkeys and rodents); the gamma-herpesviruses (Epstein-Barr virus (EBV), Marek's disease virus, Herpes saimiri, Herpesvirus ateles, Herpesvirus sylvilagus, guinea pig herpes virus, Lucke tumor virus); the family Adenoviridae, including the genus Mastadenovirus (Human subgroups A,B,C,D,E and ungrouped; simian adenoviruses (at least 23 serotypes), infectious canine hepatitis, and adenoviruses of cattle, pigs, sheep, frogs and many other species, the genus Aviadenovirus (Avian adenoviruses); and non-cultivatable adenoviruses; the family Papoviridae, including the genus Papillomavirus (Human papilloma viruses, bovine papilloma viruses, Shope rabbit papilloma virus, and various pathogenic papilloma viruses of other species), the genus Polyomavirus (polyomavirus, Simian vacuolating agent (SV-40), Rabbit vacuolating agent (RKV), K virus, BK virus, JC virus, and other primate polyoma viruses such as Lymphotrophic papilloma virus); the family Parvoviridae including the genus Adeno-associated viruses, the genus Parvovirus (Feline panleukopenia virus, bovine parvovirus, canine parvovirus, Aleutian mink disease virus, etc.). Finally, DNA viruses may include viruses which do not fit into the above families such as Kuru and Creutzfeldt-Jacob disease viruses and chronic infectious neuropathic agents (CHINA virus). [0241]
  • The present invention is further directed to the use of defective hepatitis B viruses as gene therapy reagent. In a related aspect of the present invention, the gene therapy vector can be HSV. In various embodiments of the invention, DNA is delivered to an animal as an expression construct. In order to effect expression of a gene construct, the expression construct must be delivered into a cell. As described herein, the preferred mechanism for DNA delivery is via viral infection, where the expression construct is encapsidated in an infectious viral particle. However, several non-viral methods for the transfer of expression constructs into cells also are contemplated by the present invention. In one embodiment of the present invention, the expression construct may consist only of naked recombinant DNA or plasmids. Transfection of the construct may be performed by any of the methods mentioned which physically or chemically permeabilize the cell membrane. One or other of these techniques may be successfully adapted for in vivo or ex vivo use, In a further embodiment of the invention, the expression construct may be transfected as a liposome. Also it is further contemplated that an expression construct complexed with Lipofectamine (Gibco BRL) may be used to facilitate non-viral transfection. [0242]
  • The introduction of a vector into a cell of interest may be effected by methods known to one skilled in the art, such as electroporation, DEAE Dextran, cationic liposome fusion, protoplast fusion, DNA-coated microprojectile bombardment, injection with recombinant replication-defective viruses, homologous recombination, and naked DNA transfer by, for example, intravesical instillation. It will be appreciated by those skilled in the art that any of these methods of gene transfer may be combined. [0243]
  • The present invention is further described by the following non-limiting Examples. [0244]
  • EXAMPLE 1 PKC Activation: Kinase Activity of PKC as Measured by Enzyme Assay
  • Preparation of Chemical Fractions from [0245] E. peplus
  • Sap from [0246] E. peplus plants was collected, stored at −20° C., thawed and stored at 4° C. for 1 week prior to use. The H fraction was prepared from frozen sap by thin layer chromatography (TLC) as described in International Patent Application No. PCT/AU98/00656 and was stored as dried silica-associated material at 4° C. This material was enriched in jatrophanes and pepluanes. One to two months prior to use, the material was dissolved in ethylene glycol dimethyl ether (DME) and stored at 4° C. The concentrations were determined from the dry weight of the material. For PKC assays, crude sap (PEP001) and the PEP004 fraction was ether extracted twice to produce an ether-soluble fraction enriched in diterpenes, namely, ingenanes, jatrophanes and pepluanes. The remaining water soluble fraction was also used. An ingenane fraction was prepared from the ether-soluble extract by TLC as described in International Patent Application No. PCT/AU98/00656.
  • PKC Assay [0247]
  • The conventional and novel protein kinase C (PKC) isoforms, in their unstimulated state, are inactive as kinases. The C1 domain of these PKCs contains an autoinhibitory, pseudosubstrate site that binds to the substrate site (C4 domain) and inactivates the kinase functionality of the protein. Activation of PKC results from binding of diacylglycerol (DAG) to the C1 domain, which, via multiple phosphorylation events and conformational changes to the protein, ultimately leads to the release of PKC autoinhibition. TPA and other related compounds have been shown to bind to the C1 domain of various PKC isoforms and presumably by similar means as DAG, lead to their activation. [0248]
  • The kinase activity of rat brain PKC (Promega) was determined using the Peptag™ Non-Radioactive Protein Kinase Kit (Promega). Using agarose gel electrophoresis the technique visualises the opposing electrostatic charge of a fluorescently labeled peptide (PLSRTLSVAAK) compared to the phosphorylated version of the same peptide. [0249]
  • The results of an assay of PKC with the fluorescent substrate (PepTag) are shown in FIG. 1. The reaction mixture was separated by gel electrophoresis, showing migration of the unreacted substrate (a) to the anode (top), and the product (b), which is more negatively charged because of phosphorylation by PKC, moving towards the cathode (bottom). The positive control activator (phosphatidyl serine) supplied by the manufacturer (lane 2) showed strong activation compared with PKC and substrate alone (lane 1). Various dilutions of TPA also showed activation of PKC ([0250] lanes 3, 4 and 5).
  • An ether extract of [0251] E. peplus sap, reconstituted in dimethoxyethane (DME) and incubated with PKC at a final dilution of 1 in 5 relative to the sap, gave a significant level of action (lane 7), as did the crude sap itself (lane 9). In the latter case, however, both the substrate and product (band c, lane 9) were found further towards the cathode. This result was interpreted as being due to a carboxypeptidase activity in the crude sap, cleaving the C-terminal, positively-charged lysine from the substrate peptide. This was confirmed by the finding that the aqueous layer from ether extraction had minimal PKC-activating ability, but altered migration of the substrate in the same way as the crude sap (lane 8). DME itself had no activity (lane 10).
  • FIG. 2 shows the results of testing fractionated materials simultaneously with negative (lane 1) and positive controls (lane 2). Fraction H (mixture of jatrophanes and pepluanes) showed a low activity (lane 3), seen as a halo of product (arrow) moving away from the unreacted substrate. A similar result was found for the ingenane fraction (lane 4). [0252]
  • All of the [0253] E. peplus fractions are tested for activation of all the available protein kinase enzymes using the peptide-based fluorescent tag test described above. The isoenzymes available for this experiment (Panevera) were α, β1 , β11, γ, δ, ε, η and ζ.
  • Essentially, the kinase activity of the PKC sample was assessed before stimulation (Negative Control) and after stimulation with PEP001, phosphatidyl serine (an acid-lipid, known to activate PKC, provided by Promega; Positive Control) and TPA (20 μg/mL). The results presented in FIG. 3 indicate that PEP001, at dilutions of 1:125 and 1:500, activates PKC to a similar level as phosphatidyl serine (200 μg/mL) and to a greater level than TPA (20 μg/mL). From this experiment, it is clear that the PEP001 activates PKC. [0254]
  • EXAMPLE 2 PKC Activation: Translocation of PKC
  • Activation of PKC can also be demonstrated by a simple fluorescence microscopy-based assay. Upon activation, PKC is known to translocate from the cytoplasm to the plasma membrane of the cell. By fusing PKC enzymes to the green fluorescent protein (GFP) or enhanced GFP (EGFP), activation of the PKC can be detected by the movement of diffuse cytoplasmic GFP to a ring of fluorescence associated with the plasma membrane. Using this assay, crude [0255] E. peplus extract has been shown to activate PKCβ and PKCγ.
  • MM96L cells were first transfected using a commercially-available kit (Qiagen Effectine Transfection Kit) with a PKC-GFP expression vector (Clontech; http://www.clontech.com/gfp/) and allowed to produce the PKC-GFP protein for 24 hr. The cells were then treated with crude [0256] E. peplus extract and TPA and observed under a fluorescent microscope (488 nm excitation). Two controls were used—no DNA, which allows for the identification of non-transfected cells, and no drugs, which allows for the calculation of transfection efficiency and the identification of transfected cells without PKC activation. pPKCβ-EGFP and pPKCγ-EGFP were tested, and crude E. peplus extract was shown to induce movement of the fluorescence from the cytosol to the plasma membrane, indicating that crude E. peplus extract activated these PKC enzymes. The results are illustrated in FIGS. 4A and 4B, which respectively show expression of PKCP in the absence of any drug and after exposure to crude E. peplus extract for 2 hr.
  • In another experiment, translocation of individual PKC isoforrns was observed using fluorescence microscopy and used as an indication of activation by PEP003 and PEP005. [0257]
  • Five EGFP-PKC isoforms (Clontech) were available for this experiment, enabling the screening of the three predominant PKC families (i.e. classical, novel and atypical PKCs). The members of the various PKC families are α, β, and γ (classical), θ (novel) and ζ (atypical). [0258]
  • HeLa cells were plated out in a 24-well plate containing coverslips and transfected with PKC isoforms fused to EGFP, using a commercially available effectine-transfection kit (QIAGEN, Pty. Ltd.). Cells were exposed to the transfection reagents for 16-24 hr. Subsequently, transfected cells were treated for one hour with TPA (100 ng/mL), bryostatin-1 (5 pg/mL), PEP003 (2.25 μg/mL; 5 μM) or PEP005 (670 μg/mL) 1.5 μM). Following treatment, cells were fixed on coverslips and mounted on glass slides. The slides were subsequently examined visually by fluorescence microscopy, photographed, and over 150 cells were counted/treatment/PKC isoform. Counted cells were classified according to the localisation of the PKC-EGFP fluorescence as either cytoplasmic or plasma membrane using ImagePro™ 4.1 (FIG. 5). Several cells also showed localisation to the Golgi, or similarly located cellular structure (FIG. 5). The number of these cells was also counted. Results are presented as an average and standard deviation of percentages of cells (Table 1). [0259]
  • The results presented in FIG. 6 show that PKC α, β and γ are translocated from the cytoplasm to the plasma membrane in response to treatment with PEP003, PEP005 and TPA but not with bryostatin-1. As expected, the diacylglycerol-independent PKCζ is not translocated in response to any treatment. PKCθ is translocated in response to PEP003, TPA and bryostatin-1, however, PEP005 does not induce any change in the isoenzymes localization. The results also show that treatment of PKCα and γ transfected cells with TPA, PEP003 and PEP005 leads to an increase in the number of cells displaying Golgi-like fluorescence. PKCβ transfected HeLa cells treated with TPA also show an increase in Golgi-Tike fluorescence. In contrast, treatment with PEP005 and bryostatin-1 decreases the number of cells with PKCβ concentrated in the Golgi. The number of PKCθ transfected HeLa cells with Golgi-like localization is increased in response to all treatments. [0260]
  • The above results indicate that PEP003 and PEP005 induce translocation of the classical and novel PKC isoforms tested, suggesting that these compounds activate members of the classical and novel PKC families. TPA, Bryostatin-1, PEP003 and PEP005 fail to induce translocation of PKCζ, suggesting that PEP003 and PEP005 do not activate members of the atypical PKC family. Furthermore, TPA, Bryostatin-1, PEP003 and PEP005 display differences in their ability to induce translocation of the specific PKC isoforms to the plasma membrane and/or Golgi. These differences may play a role in determining the different biological actions of these compounds. [0261]
  • EXAMPLE 3 Binding of Compounds to PKC
  • A competition assay was performed to determine whether the diterpene esters of the instant invention bind to the phorbol ester binding site of PKC. This competition assay showed that 23 μg/mL PEP003 displaced >90% [3H]-phorbol dibutyrate from binding to rat brain homogenate, used as a source of PKC (Gonzalez et al., 1999). This binding was not blocked by co-incubation with bisindolylmaleimide. These results show that PEP003 binds to the phorbol ester binding site of PKC, and bisindolylmaleimide does not. [0262]
  • EXAMPLE 4 Activation of Latent HIV Infection
  • The use of highly active anti-retroviral therapy such as combinations of reverse transcriptase inhibitors and protease inhibitors (HAART) has significantly prolonged the life of individuals infected with HIV. However, the regimen is very burdensome, requiring strict adherence to prevent a recurrence of viraemia. Long-lived cells capable of actively transcribing virus, such as CD4[0263] + cells, act as a major latent reservoir and enable the virus to avoid anti-retroviral chemotherapy or immune system surveillance. There is, therefore, an urgent need to find an agent which activates latent virus from the infected cells. Activated virus could then be killed by aggressive anti-retroviral chemotherapy and it has been hypothesized that immune system surveillance could also be improved under these conditions. Such an agent could have utility in other disease states in which virus is sequestered in infected cells, e.g. herpes infections. Anti-cancer agents have been widely investigated as potential anti-HIV agents. Several PKC activators have been shown to activate latent retroviruses. For example, PMA has been shown to activate latent HIV in monocytes (Tobiume et al., 1998). However, PMA is a known tumor promoter. A latently HIV-1 infected cell line (U1), derived from the promonocytic cell line U937 after infection with HIV-1 LAI strain, was used in these experiments. In the absence of activation, no or little virus (measured as p24 production) is produced by the U1 cell line. Phorbol esters are known to activate virus production from these cells (Tobiume et al., 1998) and so TPA/PMA was used as a positive control in these experiments.
  • U1 cells were cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum, 10[0264] 5 cells/mL were cultured for 20 hr in the presence and absence of various concentrations of either the phorbol ester TPA or crude E. peplus sap (PEP001) or PEP004 (HI) derived therefrom. Supernatants were collected and viral replication monitored by determination of the amounts of HIV p24 gag protein in the culture supernatants by ELISA, using a NEN Life Science HIV-1 p24 ELISA kit. p24 values were calculated from OD values using a standard curve.
  • TPA, the crude sap (PEP001) from [0265] E. peplus and the PEP004 fraction all activated HIV from U1 cells, as illustrated in FIG. 7. The crude sap (PEP001) was 50 times less active than TPA. The PEP004 fraction was 1000 times less active than TPA.
  • EXAMPLE 5 Lytic HIV Activity Inhibited by PEP003 and PEP004
  • The human immunodeficiency virus (HIV), a retrovirus, is the cause of the fourth greatest killing disease in the world, infecting more than 36 million people. A number of anti-retroviral compounds have been approved for clinical use, but many HIV strains have developed resistance to these drugs. There is clear and immediate need for new anti-retroviral compounds. [0266]
  • Experiments were conducted to assess the effect of the compounds of the instant invention on HIV-1 replication in acutely infected T cells. Peripheral blood mononuclear cells (PBMC) were obtained from non-HIV-1, non-HIV-2, non-Hepatitis B/C infected donors, stimulated with phytohemagglutinin-M and grown in culture media supplemented with 10 U/mL interleukin-2. The activated PBMC were infected with 10 g (Low Titre) and 100 ng (High Titre) of CA-p24 equivalents of the HIV-strain pNL4-3. Cells were infected for two hr after which, the virus was removed and the cells were washed with culture media. Equivalent numbers of cells were seeded into 24 well plates and compounds were added to the cultured cells that included: TPA at 8 riM and 80 nM, Ingenol at 280 nM, PEP003 at 500 nM, 50 nM and 5 nM, or PEP004 at dilutions of 1×10[0267] 4 and 1×105 from the stock (final concentrations). In addition, uninfected activated PBMC were grown in the presence of TPA (80 nM), Ingenol (280 nM), PEP003 (500 nM) and PEP004 (1×104 dilution). Other cultures were neither infected nor treated with any compound, or infected but not treated with any compound. Supernatant was removed from each culture at day 0, 3, 7, and 10. The amount of HIV-1 CA-p24 was determined using a commercially available ELISA assay. Three independent experiments were performed.
  • The data presented in FIGS. [0268] 8A-8D show that PEP003 reduced virus replication kinetics in a dose-dependent manner. PEP003 at concentrations of 500 nM, 50 nM and 5 nM reduced the replication rate by approximately 99.9%, 95% and 47%, respectively, relative to the untreated, infected cells. PEP004 at dilutions of 1×104 and 1×105 reduced the replication rate by approximately 66% and 15%, respectively. Viral load seemed to alter these results slightly, as higher initial inoculums of virus reduced the total inhibition of PEP003 at 500 nM or 50 nM to approximately 97% (t-test; p<0.001) or 88% (t-test; p<0.074), respectively. The control compounds Ingenol (2.8 μM) and TPA (80 nM or 8 nM) reduced HIV-1 replication rates by approximately 35%, 98% and 38%, respectively.
  • EXAMPLE 6 Enhancement of the Cytomegalovirus Promoter Activity as a Method for Improving Gene Therapy
  • Viruses and viral promoters especially adenovirus and CMV are used to deliver gene therapy in a range of human disease conditions. Gene expression and, hence, therapeutic effect will be enhanced if the promoters driving their transcription can be activated further by an agent. [0269]
  • Human melanoma cells were infected with ten-fold dilutions of [0270] adenovirus 5 in culture, treated with dilutions of PEP005, PEP006, PEP008 and PEP010 and adenovirus replication determined 2 days later by immunhistochemical detection of virus-replicating cells. Virus replication (enumerated as the number of stained cells following successive incubations with adenovirus antibody, peroxidase-conjugated protein A and peroxidase substrate) was increased by 344% with 67 ng/mL PEP005, 256% with 295 ng/mL PEP006, 248% with 226 ng/mL PEP008 and 147% with 67.5 ng/mL PEP010.
  • The CMV promoter is commonly used to activate the transcription of genes in constructs transfected into a variety of cells, due to its strong transcriptional activity in a variety of human cell types. The ability of TPA to increase this activity has been demonstrated in cells undergoing non-productive infection with an adenovirus construct (Christenson et al., 1999), thus raising the possibility of increasing the production of a therapeutic protein encoded by a similar construct. [0271]
  • Human melanoma cells (MM96L; 50,000 per microtiter well) were treated with TPA or dilutions of crude [0272] E. peplus sap, infected with a 1/20 dilution of a pool of adenovirus-5 expressing β-galactosidase driven by the CMV promoter. After incubation for 20 hr, the wells were washed with 3× with PBS, 50 μL of chlorophenol red galactoside (GPRG) substrate solution added and the absorbance at 540 nm read after 90 min. The inventors found TPA (100 ng/mL) and crude E. peplus sap (diluted 1 in 10,000) both induced the CMV promoter activity by >3-fold.
  • EXAMPLE 7 Activation of Innate Immune Responses: Induction of Neutrophil Invasion in Skin
  • Neutrophils represent about 70% of peripheral white blood cells in humans and play a pivotal role in inflammation and the innate defense against disease (Mollinedo, 1999). Upon activation, neutrophils release superoxide radicals and granules containing a variety of enzymes and other compounds. These secretions are able to destroy invading pathogens, but also result in inflammation and associated tissue damage. [0273]
  • The inventors found that [0274] E. peplus sap causes accumulation of neutrophils at the site of application, showing that E. peplus sap is capable of recruiting neutrophils. A mixture of active diterpenes obtained as an ether extract from E. peplus sap was applied (2 μL of 100 mg/mL in ethanol) on the skin of a nu/nu mouse. After 24 hr, the animal was sacrificed and the skin fixed in 10% formalin for sectioning and hematoxylin/eosin staining. As shown in FIGS. 9A and 9B, control skin showed normal skin structure with few infiltrating monocytes. The treated skin showed large numbers of infiltrating neutrophils, characterized by their polymorphic nuclei. There was no evidence of gross damage to the skin.
  • EXAMPLE 8 Neutrophil Infiltration Activity
  • Basal cell carcinoma (BCC) is the most common cancer in the Caucasian population, with the highest annual incidence globally having been recorded in Australia (Miller et al., 1994, Marks et al., 1993). New developments have begun looking at treating non-melanoma skin cancer (NMSC) using topical therapies. The essence of this therapy may rely upon the induction of an inflammatory response with infiltration of leucocytes, in particular neutrophils. [0275]
  • To assess whether the compounds of the invention induce neutrophil infiltration, an experiment was designed on C57BL/6J mice. Twenty-four mice were divided into six groups of four mice per group. In three of these groups the mice had a B16 melanoma injected s.c. (2 sites per mouse, 5×10[0276] 5 cells/site), that was left to grow for 8 days to approximate tumor sizes of 5-8 mm in diameter. A single application of one of all three compounds was then applied to the tumor or to normal skin. Each compound was applied on two groups of mice, one with tumor and 1 without tumor. The three compounds were PEP010 (2 μL; 150 mM) in 10 μL of isopropanol gel (isopropyl alcohol 25% (w/w), propyl alcohol 25% (w/w)) (vehicle), PEP009 (2 μL of stock) in 10 μL of vehicle or vehicle alone as a control. One mouse from each group was then sacrificed at either 4 hr, 24 hr, 48 hr or 144 hr post single application of compound and then tissue excised and sections prepared for histology.
  • The results at 4 hr show only minimal response with 1+ patchy neutrophils for both PEP010 on B16 tumor and PEP009 on normal skin and 2+ neutrophils present for PEP009 on B16 tumor (Table 2). At 24 hr, there are no neutrophils present in the control groups with vehicle alone but a 4+ neutrophil infiltration with PEP010 and PEP009 application, on both tumor and normal skin (FIGS. 10A and 10B). In addition, 60-85% of the superficial tumor cells were apoptotic or necrotic in the B16 groups. At 48 hr, there was a similar pattern with a 4+ neutrophil presence with PEP010 and PEP009 application while the control groups showed an absence of neutrophils (FIGS. 10A and 10B). Along with the tumor cell necrosis and apoptosis, there is also evidence of some neutrophil breakdown at the 48 hour interval. The 144 hour group showed a lack of neutrophils in the control group and a presence of 2-4+ neutrophils, which were mostly now degenerate in the PEP010 and PEP009 groups. There was extensive necrosis of tumor and skin, and clear signs of granulation tissue and early repair. [0277]
  • This study shows that the PEP010 and PEP009 induce a marked inflammatory infiltrate of neutrophils as compared to vehicle alone and this influx of polymorphonuclear cells may be significant in altering the growth of certain skin cancers. [0278]
  • EXAMPLE 9 Activation of Innate Immune Responses: Induction of a Respiratory Burst in Peripheral Blood Mononuclear Cells
  • Monocytes/macrophages are blood-borne and tissue cells which are usually activated by T lymphocytes and antibodies. Upon activation, they are able to phagocytose pathogens, release superoxide radicals and are an important source of cytokines. Crude [0279] E. peplus extract was shown to be able to induce the release of superoxide radicals by use of a fluorescence-activated cell sorting (FACS)-based method, in which superoxide radicals are detected by the dye dihydroethidium. In addition, phagocytic activity was activated by E. peplus, as shown by increased uptake of nitroblue tetrazolium and adherence to plastic was increased by E. peplus; this is believed to indicate activation and differentiation of macrophages.
  • Human peripheral blood mononuclear cells (PBMC) prepared by standard Ficoll separation comprise approximately 5% monocytes. PBMC were incubated with dihydroethidium, a reduced form of the dye which becomes fluorescent when oxidized by a respiratory burst, then treated in 10% FCS-RPMI 1640 at 37° C. for 15 min with crude [0280] E. peplus extract diluted 1/1000 or 100 ng/mL TPA and analyzed by flow cytometry using conventional methods (Handbook of Flow Cytometry Methods, p. 151) The mean channel numbers for fluorescence were 618 (controls) and 818 (E. peplus extract diluted 1/1000). These results, illustrated in FIGS. 11A and 11B, show that the E. peplus extract induced intracellular oxidation of the dye, typical of a respiratory burst. Phagocytic activity was determined by a conventional method (Hudson and Hay, Practical Immunology, 3rd edition, p. 74). Cells were treated in 10% FCS-RPMI 1640 at 37° C. for 20 min with introblue tetrazolium (NBT) and crude E. peplus extract (PEP001) diluted 1/1000 or 100 ng/mL TPA, followd by counting the number of blue-stained cells in a haemocytometer. The average of three fields gave figures of <2% (controls), 10% (TPA) and 8.7% (E. peplus sap) cells stained blue. This demonstrates induction of phagocytic activity, part of the normal response to infectious agents, by E. peplus sap, as shown by uptake by cells of the blue NBT precipitate.
  • Experiments were also carried out using 2′,7′-dichlorofluorescein diacetate (DCFH-DA) to measure the production of H[0281] 2O2.(J P Robinson, Oxidative burst methods, in Handbook of Flow Cytometry Methods, Wiley-Liss Inc, pp147-149, 1993). H2O2 oxidizes the non-fluorescent probe (DCFH-DA) to a fluorescent probe that can then be detected by a flow cytometer. Peripheral blood mononuclear cells (PBMC) were extracted from a donor blood sample by lysis of heparinized blood and used in a suspension of 1×106/mL of phosphate buffer, pH 7.3. The cells were then incubated with DCFH-DA (1 μL/mL of 20 mM stock) for 15 minutes to allow it to be taken up and trapped by hydrolysis with cellular esterases. The cells were then stimulated by test compounds for 15 min at 37° C. Controls included in the experiment were unloaded control (cells with no DCFH-DA) and loaded control (cells with DCFH-DA, but no stimulation). These were used to monitor the non-specific oxidation of unstimulated cells. The cells were then analyzed on the flow cytometer (excitation at 488 nm, emission at 525±20 nm), gating each sample for individual cell populations-granulocytes, monocytes and lymphocytes (Table 3).
  • All compounds except Bryostatin induced a respiratory burst, the effect being strongest in granulocytes and monocytes compared with lymphocytes. Similar results were obtained by measuring the reduction, under the same conditions, of nitroblue tetrazolium, measured as the proportion of purple-stained cells counted under the microscope. [0282]
  • Evidence for the requirement of PKC activation was obtained by addition of bisindolylnaleimide (10 μg/mL or 1 μg/mL) at the same time as PEP005, PEP006, PEP008 and PEP010. This PKC inhibitor blocked the respiratory burst seen with TPA and PEP003. [0283]
  • Phagocytosis with Fluorescent Beads [0284]
  • Phagocytosis by peripheral blood mononuclear cells (PBMCs) was assayed (Steinkamp et al., 1982) using 1 μm Fluoresbrite™ yellow-green fluorescent latex spheres (Polysciences, Inc., Warrington, Pa.). A sample of whole, heparinized blood was treated with drug and 5×10E7 fluorescent latex beads in 10 μL of PBS added per mL of suspension. Cells were incubated and maintained in suspension for 30 min by means of a shaker platform at 37° C. The stirnulated and non-stimulated samples were then lysed to isolate PBMCs. The PBMCs were run on the flow cytometer measuring FITC (excitation at 488 nm, emission at 525±20 nm), gated for fluorescence (phagocytosed spheres) and light scatter (cell size). [0285]
  • The data presented in Table 4 indicate that TPA, PEP006, PEP008, PEP003 and PEP005 all stimulate phagocytosis in PBMCs. [0286]
  • EXAMPLE 10 Activation of Innate Antiviral Activity
  • Many viruses, including alphaviruses, are sensitive to innate antiviral activities, which are often mediated by the activation of interferon α/β responses (Antalis et al., 1998). Such antiviral activities inhibit the ability of cells to support viral replication. For many viral infections, including those caused by Ross River virus, viral replication results in virus-induced cytophathic effect (CPE) or cell death. Treatment of human fibroblast cells with [0287] E. peplus ingenanes was shown to activate antiviral activity and prevented CPE induced by an alphavirus infection.
  • Human skin fibroblasts (10e4/well) were seeded in 96 well plate and left overnight to adhere. An extract of [0288] E. peplus ingenanes was added at 5 μg/mL for 48 hr. An alphavirus (Ross River virus, T48) was then added at a dose of 1, 10 and 100 cell culture ID50 for 6 days (La Linn et al., 1996). The cytopathic effect of the viral infection was assayed using crystal violet staining. Protected cells stain violet, whereas cells which have suffered CPE detach from the plate, leaving the well unstained. Alphavirus-induced CPE was observed in treated cells only at a 100-fold greater dose of virus than was required to induce CPE in untreated cells, indicating that a significant degree of protection was conferred by the E. peplus extract.
  • EXAMPLE 11 Protection Against Intra-Peritoneal Streptococcal Infection: Effect of PEP003 and PEP004 on Systemic Group A Streptococcal Infection in Mice
  • Infection of humans with group A streptococcus ([0289] Streptococcus pyogenes) (GAS) can cause a variety of clinical manifestations including the relatively minor pharyngitis (“trep throat” and impetigo (superficial skin infection) to more severe invasive infections such as toxic shock syndrome and necrotizing fasciitis, both of which, may lead to multisystem organ failure. Lastly, the GAS post-infectious sequelae of rheumatic fever (RF), rheumatic heart disease (RHD) and acute glomerulonephritis (AGN) are a major problem in developing countries and indigenous populations, particularly in Australian Aboriginals. Current treatment for controlling GAS infection is with antibiotic therapy, however, since continual high dose administration of antibiotic is required in cases of repeated episodes of acute RF and the development of RHD, poor compliance is often associated with the persistence of these GAS-associated diseases. The development of a vaccine against GAS infection would prevent GAS-associated diseases including RF and RHD. In the absence of a vaccine, however, the development of new drugs with improved anti-bacterial activity may provide promising therapeutic agents.
  • The inventors' aim was to test the ability of the PEP003 and PEP004 to systemically protect against GAS infection, in vivo. Mice (n=10) were treated with 50 μL of PEP003 (500 nM), PEP004 (1:100 dilution from stock) or control (PBS/10% acetone), 24 hr prior to and thereafter i.p. challenge with live GAS. Two different strains of mice (Quackenbush and B10.BR) and four different GAS strains (NS-1, PL-1, 88/30 and M1) were used. Mice were monitored for two weeks post-challenge and the percentage survival of mice determined. Percentage survival in Quackenbush mice challenged with PL-1 GAS was 70% (PEP003), 60% (PEP004) and 40% (control) (Table 5). Control mice that had been given the same successive treatment of PEP003 and PEP004 (but not challenged) to rule out any potential adverse side effects of the compounds were then also challenged with PL-1; survival was 40%, 80%, and 20% for PEP003, PEP004 and controls, respectively (Table 6). In the latter experiment, the protective effect of PEP004 approached significance (p=0.06), however, small numbers of mice were used (n=5). In Quackenbush mice challenged with NS-1 GAS, survival was 50% for PEP003 and controls, and 80% for PEP004 (Table 5). In B10.BR mice challenged with M1 GAS, survival was 10% for controls, 30% for PEP003 and 0% for PEP004 (Table 5). In B10.BR mice challenged with 88/30 GAS, survival was 20% for controls, 30% for PEP004 and 0% for PEP003 (Table 5). The data indicate a possible protective effect of PEP004 against systemic GAS challenge in Quackenbush mice. In addition, these data indicate that a weekly treatment regimen of PEP003 and PEP004 prior to GAS challenge may be more effective. [0290]
  • EXAMPLE 12 Anti-Escherichia coli Activity of PEP003: Activation of Leucocytes
  • Blood was collected into a Sodium Heparin tube (Becton Dickinson VACUTAINER) and leucocytes prepared by lysis of red blood cells (Handbook of Flow Cytometry Methods. Robinson J P. Wiley-Liss Inc 1993. Oxidative Burst Methods H[0291] 2O2 DCF Assay by Flow cytometry p 147-149). Leucocytes were resuspended and divided equally into two tubes such that each tube contained 7×106 peripheral blood cells (PBCs). Both tubes were then centrifuged (Beckman, GS-6) at 1000 rpm for 10 minutes. The supernatant was removed and the volume was then adjusted to 1 mL with RPMI 1640 (Gibco BRL, antibiotic free supplemented with 10% v/v fetal bovine serum. 100 μL of PEP003 (to give a final concentration of 23 μg/mL containing 10% acetone was then added to one tube and to the other, 100 μL of PBS/10% Acetone. To each tube, 10 μL E. coli (competent cells, XL10-Blue, Stratagene) was also added (to give a ˜1/100 dilution of a static culture). Both tubes were vortexed then centrifuged (Beckman, GS-6) at 2500 rpm for 10 minutes. Lids were loosened and the tubes were incubated at 37° C./5% CO2.
  • Following 16 hr incubation, the tubes were vortexed. To estimate the number of [0292] E. coli, 50 μL was taken from both tubes as well as the static starter culture (stored at 4° C.), transferred to Eppendorf tubes and centrifuged (Beckman, GS-15R) at 10,000 rpm for 10 minutes. Supernatant (˜45 μL) was removed and the pellet resuspended in the remaining ˜5 μL. A smear was made on a glass slide using the 5 μL bacterial suspension and stained using Quick Dip (Histo.Labs, Riverstone, Australia), a modified method of the Wright-Giemsa stain, which stains bacteria blue. E. coli were counted using a conventional light microscope (× 400) with an eyepiece micrometer (100 μm×100 μm). This count was then adjusted to give a total count in the smear (area=12.5×105 μm2) and expressed as the number of E. coli per mL. Another method of measuring growth of E. coli was to read the absorbance (595 nm) of the supernatant.
  • The results presented in FIGS. 12 and 13 show that treatment of leucocytes with PEP003 results in a significant reduction in bacterial numbers. [0293]
  • EXAMPLE 13 Treatment of Ringworm
  • Ringworm is a subcutaneous mycosis or dermatophytosis caused by fungi of the species Trichophyton, Microsporum and Epidermophyton, in which the infection is confined to the keratinous structures of the body. A two week old ringworm lesion, determined to be [0294] Trichophyton mentagrophytes var. mentagrophytes by culture, on the volar surface of the forearm of an adult male human was treated with a single topical application of crude E. peplus extract and was shown to resolve after seven days. Resolution of such lesions in the absence of treatment does occur, but is considered extremely rare.
  • EXAMPLE 14 Treatment for Bites of Blood-Sucking Insects
  • The bites of blood sucking insects such as mosquitos and sand flies often cause an itchy inflammatory reaction at the site of the bite. Although the extract mechanism of this reaction is poorly understood, mast cells and histamine release are likely components of this reaction (Greaves and Wall, 1996; Horsmanheimo et al., 1996). [0295]
  • In preliminary experiments, the inventors treated human sand fly bites with [0296] E. peplus extract and found a rapid reduction in the itchy sensation compared to untreated bits at a distant site. Without wishing to be bound by any proposed mechanism, the inventors believe that the E. peplus extract may strongly stimulate mast cell exocytosis and histamine release and thereby prevent the slow release over time of these compounds, a feature associated with itchiness.
  • EXAMPLE 15 Promoter Activation as a Means of Therapy: Effect of PEP003 and PEP004 on Activation of EBV Infected Cell Lines and EBV Positive Burkitt's Lymphoma Cell Lines
  • Initially the effect of PEP003 and PEP004 was tested on the B95-8 cell line (an EBV positive marmoset cell line that is used worldwide as one of the best EBV producers). This cell line was treated with each of these compounds (at different concentrations) for 3 days and 7 days, respectively, and activation of EBV virus production was measured by the appearance of a viral capsid antigen (VCA) on western blots. Also, as a comparison, EBV was activated in this cell line with TPA. [0297]
  • To ensure that equal amounts of each sample were analyzed, the gels were stained with Coomassie blue and the loadings were adjusted to make them equal. Analyses of VCA in each of the samples showed that both PEP003 and PEP004 were capable of activating EBV (at all of the concentrations used) to similar levels as using 65 nM TPA (FIG. 14). Next the PEP003 and PEP004 were assayed on two Burkitt's lymphoma cell lines and an LCL. This time only concentrations of 10[0298] −5 and 10−7 were used. Neither PEP003 and PEP004 had much effect on the LCL (this LCL produces some VCA without and chemical induction and this was not increased by these compounds). PEP004 had no effect on VCA production in any of the cell lines used. However, PEP003 did induce high levels of VCA in both Burkitt's lymphoma cell lines (MutuI and BL74), but only at 105 concentration (FIG. 15). Similar results were obtained when the cell lines were assayed for induction of BZLF1, the initial transactivator of EBV replication (FIG. 16). The results show that PEP003 was capable of activating EBV in Burkitt's lymphoma cell lines, but appeared to have little effect on LCLs.
  • In conclusion, (1) both TPA and PEP003 can modulate gene expression in EBV transformed tumor cells at the doses used; (2) while PEP003 induced VCA in MutuI cells TPA did not, indicating different modes of action; (3) surprisingly, there was no apparent effect of PEP003 on lymphoblastoid cells, indicating potential for activating latent herpesvirus in tumors without affecting the normal infection. [0299]
  • EXAMPLE 16 Investigation Into the Effect of PEP003 on the Ability of Melanoma Cells to Stimulate NK Activity
  • Melanomas and other cancers can be killed by both specific (T cell-mediated) and non-specific (natural killer cell and other mechanisms) arms of the immune response. These killer cells can be generated in vitro by stimulating peripheral blood T cells from selected melanoma patients with melanoma cells derived from the same patient (“autologous”). Natural killer cells can be recognized by their lysis of the natural killer-sensitive cell line K562. It has been theorized that some anti-tumor agents alter the susceptibility of melanomas to immune responses. [0300]
  • Peripheral blood lymphocytes from patient A02, who has a strong specific T cell response to her own melanoma cells (A02-M), were thawed and stimulated by irradiated A02-M pre-treated overnight at 37° C. with (a) PEP003 (2.25 μg/mL; 50 μM); (b) TPA (100 ng/mL); or (c) control solvent/buffer, and washed ×2 before addition to responding lymphocytes (washing × 2 achieves a residual agent dilution of × 100,000). After 10 days of culture, the stimulated cells were harvested and used as effectors against an NK-sensitive cell line (K562) to test for the level of NK activity generated in culture. All determinations were performed in triplicate, at E:T ratios of 45, 15, 5 and 1.7:1. A standard 5 hour[0301] −51 Cr release assay was performed. Stimulations were performed in 10% fetal bovine serurn/RPMI-1640.
  • The results presented in Table 7 and FIG. 17 indicate that pre-treatment of melanoma cells with PEP003 significantly increases the lysis of K562 compared to both TPA and the control treatment at the E:T ratio of 45:1 (P<0.01 in both cases), suggesting that PEP003 increases NK activity in A02 cultures. [0302]
  • EXAMPLE 17 Methods for Obtaining a Low-Chlorophyll, Hydrophobic Fraction From E. peplus and Other Plant Species
  • Standard methods for the isolation of hydrophobic compounds from plants involve alcoholic extraction of the whole plant. This produces an extract containing chlorophyll and other hydrophobic substances from the leaves that interfere with subsequent purification of compounds by solvent extractions and chromatography. This is a particular problem in isolating highly bioactive diterpenes from members of the Euphorbiaceae family, due to co-migration with chlorophyll on silica gel chromatography. Two methods, both of which can be scaled up for economical, commercial production, have been developed to overcome this problem, as described in the present Example and in Example 18. [0303]
  • Fresh [0304] E. peplus plants (17 kg) were chopped and soaked in 150 litres of water at 4° C. for 20 hr. The water was pumped through 50 and 100 mesh sieves, filtered through 5 and 2 micron filters and then recirculated through a 100 mm diameter column of Amberlite XAD-16 (1.5 kg, conditioned successively with ethyl acetate, methanol and water) at 4° C. (approximately 1.2 L/min) for 72 hr. Adsorption of bioactivity to the resin was found to be virtually complete within 20 hr.
  • The resin was then washed successively with water and 50% methanol, then eluted with 1 L of methanol, followed by 2×1 L acetone. The eluates were evaporated and combined to give approximately 7 g of a thick oil. This was shown by HPTLC to be substantially free of chlorophyll and to contain the desired ingenane esters which were then purified as described below. [0305]
  • The ability to extract diterpene esters from chopped plants in water was surprising given their relative hydrophobicity and water insolubility. A variety of manual (cutting with scissors) and mechanical (rotary cutters, motor-driven mulcher) plant maceration methods were successful, as was extraction at room temperature. Adsorption to the XAD-16 could be achieved by stirring the resin with the filtered or unfiltered water extract and then pouring off the latter. Filtration could also be carried out with minimal loss of bioactivity using diatomaceous earth, or membrane filters (220-650 microns). XAD-7 and XAD-4 were as effective as XAD-16. [0306]
  • The hydrophobic adsorbent polyamide (ICN Biomedical Research Products) was also used to trap the diterpenes from water; it had the advantage of allowing the diterpene esters to be selectively eluted with 50-80% methanol, thus separating them from inactive, hydrophobic compounds, which remained on the column. [0307]
  • EXAMPLE 18 Method for Separation of Ingenane Esters From Other Diterpenes
  • The following method is based upon the surprising discovery that the stems of [0308] E. peplus contain approximately 90% of the bioactive diterpenes and significantly less chlorophyll compared with the leaves.
  • The plants are dried in air, shaken to remove the leaves and the stems compressed and covered with an equal weight of methanol for 24 hr. The solvent is then poured off, evaporated to dryness under reduced pressure and the residue dissolved in methanol for chromatography on Sephadex HL20 as described below. This method is also suitable for isolation of low-chlorophyll fractions from other plant species. [0309]
  • A solution of crude methanol extract from [0310] E. peplus in 4 mL 90% ethanol was loaded onto a 25 mm×1000 mm column and eluted with 90% methanol. Fractions (4 mL) were analysed by HPTLC (silica gel, developed with 4:1 toluene: acetone and heated with phosphoric acid at 110 degrees for 15 min). Typically, fractions 54-63 contained jatrophane and pepluane esters and fractions 64-77 the ingenane esters, thus achieving satisfactory separation. Bioactivity, as judged by induction of bipolar morphology in the human melanoma cell line MM96L, was retained, as for example disclosed in PCT/AU98/00656.
  • This separation was surprising because the polarity of the ingenane esters as judged by HPTLC on silica completely overlapped the range shown by the jatrophane and pepluane esters. [0311]
  • EXAMPLE 19 Process for the Purification of Diterpene Esters From E. peplus
  • Crude extracts obtained by the methods according to Examples 17 or 18 above, or by ether extraction of latex, were fractionated by Sephadex HL-20 chromatography (as above). Appropriate fractions from the latter were combined, the methanol evaporated under reduced pressure and the remaining water removed by freeze-drying or by ether extraction. This sample (200 μL of 100 mg/mL in methanol per injection) was fractionated by HPLC on a Phenomenex Luna 250×10 mm C18 column with a Phenomenex guard column in 70-100% methanol at 2 mL/min, with detection at 230 nm. Jatrophane and pepluane esters appeared at 25-42 min, PEP005 at 42-44 min, PEP008 at 46-50 min, and PEP006 at 50-54 min. Similar types of separation have been obtained by HPLC on C3 and C8 columns. [0312]
  • Fractions pooled from repeated runs were evaporated to dryness (rotary evaporater or freeze dryer), and stored in acetone at −20° C. under argon or nitrogen. [0313]
  • EXAMPLE 20 Activation of Leukocytes by Diterpene Esters, for Selective Killing of Human Tumor Cells in Culture
  • Leukocytes obtained by lysis of human peripheral blood were added to 5000 MM96L human melanoma cells or 7000 neonatal foreskin fibroblasts per microtitre well at effector: target ratios of 1000, 100 and 10:1. Ing9 (60 ng/mL) was added and after 48 hr incubation the cultures were washed and labelled with [3H]-thymidine for 2 hr. At 100:1 ratio of effector:target cells, the melanoma cells showed 9% survival with PEP008 whereas the normal fibroblasts had 100% survival. Untreated leukocytes had no effect on cell survival. [0314]
  • These results indicate that the diterpene esters of the invention activate human peripheral blood leukocytes to produce, in a PKC-dependent manner, phagocytosis and a respiratory burst which are potentially lethal to micro-organisms and other cells. [0315]
  • This example shows that drug-activated, PKC-dependent processes can direct tumor-specific killing by cells of the innate immune system. [0316]
  • EXAMPLE 21 Pretreatment of Human Tumor Cells in Culture With Diterpene Esters Potentiates Selective Killing by Untreated Leukocytes
  • The question of whether drug treatment of the target tumor cells causes them to become susceptible to effector cells of the immune system was addressed as follows. [0317]
  • Leukocytes obtained by lysis of human peripheral blood were added to 5000 MM96L human melanoma cells or 7000 neonatal foreskin fibroblasts per microtitre well at effector: target ratios of 1000, 100 and 10:1. The target cells had been treated with 60 ng/mL PEP008 for 20 hr beforehand, and washed and the medium replaced before the leukocytes were added. After 48 hr incubation with the leukocytes the cultures were washed and labelled with [3H]-thymidine for 2 hr. At 100:1 ratio of effector:target cells, the melanoma cells showed 12% survival with PEP008 whereas the normal fibroblasts had 100% survival. Untreated leukocytes had no effect on cell survival. [0318]
  • This result showed that the drugs also act by making tumor cells specifically sensitive to lysis by the immune system. [0319]
  • EXAMPLE 22 Topical Composition A for the Treatment of Conditions Affecting Skin (e.g. Infections, Skin Cancers)
  • Tinctures: Compounds of the invention were diluted into acetone, ethanol or isopropanol to the same final bioactivity as the [0320] E. peplus latex as measured by bipolar activity in MM96L human melanoma cells (10 million bp units per mL). Samples (2-5 μL) were applied daily for 3 days to the surface of mouse melanoma B16 tumor 3-5 days after implanting s.c. 1 million cells on the flanks of nude mice. Efficacy, defined as 67% or more sites cured, was obtained for E. peplus sap, PEP005, PEP008 and a mixture of PEP005, PEP006 and PEP008.
  • EXAMPLE 23 Topical Composition B for the Treatment of Conditions Affecting Skin (e.g. Infections, Skin Cancers)
  • Creams and gels: A variety of hydrophobic cream bases was found to be ineffective when used to deliver compounds to the skin as described above for the tinctures. [0321]
  • Efficacy was obtained with the use of an isopropanol gel, formulated as described for the tinctures. [0322]
  • The results show that [0323] E. peplus sap and its terpenoid components activate PKC, with consequent potential to induce a wide range of cellular responses without the high tumor promoting activity of TPA. The carboxypeptidase activity may have application in enhancement of tissue penetration and in antigen processing for optimal immune responses.
  • Overall, the results indicate that [0324] E. peplus extract induces a set of cellular responses with affects PKC, cell cycle genes and inflammatory mediators, some but by no means all of which are similar to the action of TPA. In particular, the results indicate that E. peplus sap and its terpenoid components are useful in the treatment of a variety of infections and as adjuvants for stimulating immune responses.
  • EXAMPLE 24 Effect of Saps Derived From Other Members of the Euphorbiaceae Family on MM96L Cells
  • Sap was collected from [0325] Synadenium grantii, Synadenium compactum, Mondenium lugardae, Mondenium guentheri, Endadenium gossweileni, and E. peplus and serially diluted ten-fold up to 10−7into sterile 1.5 mL Eppendorf™ tubes using growth medium. Ten-microlitre aliquots of each dilution, in the presence or absence of the PKC inhibitor bisindolylmaleimide (1 μg/mL or 10 μg/mL), were added to 5000 MM96L cells per well of a microtitre plate. After 3 days, cells were examined for cytotoxicity or differentiation to a bipolar dendritic phenotype.
  • The results presented in Table 8 show that the saps of [0326] S. grantii, S. compactum, M. lugardae, M. guentheri, and E. gossweileni, like that of E. peplus, induce the differentiation of MM96L cells to a bipolar phenotype and that this differentiation is inhibited by the bisindolylmaleimide. This inhibition strongly suggests that the active components of the saps induce cell differentiation by inhibition of PKC activity. The results also show that at higher concentrations (10−4 and above), the saps are effective in killing MM96L cells.
  • EXAMPLE 25 Effect of Saps Derived From Other Members of the Euphorbiaceae Family on JAM Cells
  • The saps of Example 24 were also examined for their cytotoxic effect on the ovarian carcinoma cell line JAM. Ten-microlitre aliquots of each dilution of sap, prepared according to Example 24 in the presence or absence of the PKC inhibitor bisindolylmaleimide (10 μg/mL), or in the presence or absence of the PKC phorbol ester binding site ligand phorbol dibutyrate, were added to 5000 JAM cells per well of a microtitre plate. After three days, the cells were fixed with ethanol and the number of cells compared with untreated controls stained with sulfurhodamine B. [0327]
  • The results presented in FIGS. 18A and 18B indicate that, like the sap of [0328] E. peplus, the saps of S. grantii, S. compactum, M. lugardae, M. guentheri, and E. gossweileni, at concentrations of 10−4 and above, are effective in killing JAM cells. These results also show that cytotoxicity is inhibited by bisindolylmaleimide, suggesting that this effect is mediated by modulation of PKC.
  • Inspection of FIG. 18C reveals that the cytotoxic effects of saps derived from [0329] M. guentheri and E. gossweileni were blocked in the presence of phorbol dibutyrate, suggesting that the active components of these saps mediate their cytotoxicity by binding to the phorbol ester binding site of PKC.
  • Those skilled in the art will appreciate that the invention described herein is susceptible to variations and modifications other than those specifically described. It is to be understood that the invention includes all such variations and modifications. The invention also includes all of the steps, features, compositions and compounds referred to or indicated in this specification, individually or collectively, and any and all combinations of any two or more of said steps or features. [0330]
  • BIBLIOGRAPHY
  • Antalis et al., [0331] J. Exp. Med. 187: 1799-1811, 1998.
  • Christenson et al., [0332] Endothelium 7: 75-82, 1999.
  • Elliott et al., [0333] Vaccine 17: 2009-2019, 1999.
  • Evans & Osman, [0334] Nature 250: 348, 1974.
  • Fatope et al., [0335] J. Med. Chem. 39: 1005-1008, 1996.
  • Gonzalez et al., [0336] Melanoma Res. 9: 599-606, 1999.
  • Greaves and Wall, [0337] Lancet 348(9032): 938-940, 1996.
  • Gundidza and Kufa, [0338] Centr. Afr. J. Med. 38: 444-447, 1992.
  • Hecker “Cocarcinogens from Euphorbiaceae and Thymeleaceae” in “[0339] Symposium on Pharmacognosy and Phytochemistry”, 147-165, (Wagner et al., eds., Springer Verlag, 1970).
  • Horsmanheimo et al., [0340] J. Allergy Clin. Immunol. 98: 408-411, 1996.
  • Imai et al., [0341] Anticancer Res. 14: 933-936, 1994.
  • La Linn et al., [0342] J. Gen. Virol. 77: 407-412, 1996.
  • Marks et al., [0343] Int. J. Cancer 53(4). 585-590, 1993.
  • Matsushita et al., [0344] Int. J. Hematol. 72(1): 20-7, 2000.
  • Miller et al., [0345] J. Am. Acad. Dermatol. 30(5): 774-778, 1974.
  • Mollinedo, [0346] Immunol. Today 20(12): 535-7, 1999.
  • Murali-Krishna et al., [0347] Immunity 8: 177-187, 1998
  • Oksuz et al., [0348] Phytochemistry 42: 473-478, 1996.
  • Platzbecker et al., [0349] Transplantation 71(7): 880-885, 2001.
  • Starvic and Stolz, [0350] Food Cosmet. Toxicol. 14: 141, 1976.
  • Steinkamp et al., [0351] Science 215: 64-66, 1982.
  • Tobiume et al., [0352] J. Gen. Virol. 79: 1363-1371, 1998.
  • Westphal et al., [0353] Cancer Res 60(20): 5781-5788, 2000.

Claims (144)

1. A method for the treatment or prophylaxis of a PKC-related condition or disorder in a subject, said method comprising the administration to said subject of a symptom-ameliorating effective amount of a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or chemical analogue thereof which chemical agent is a macrocyclic diterpene selected from compounds of the ingenane, pepluane and jatrophane families and which chemical agent or derivative or chemical analogue is represented by any one of the general formulae (I)-(V):
Figure US20030195168A1-20031016-C00007
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A-T are independently selected from hydrogen, R1, R2, R3, F, Cl, Br, I, CN, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2, (C═X)R3 or X(C═X)R3 where X is selected from sulfur, oxygen and nitrogen;
R1 and R2 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2]alkyl;
R3 is selected from R1, R2, CN, COR1, CO2R1, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2;
A connected to B (or C), D (or E, R (or Q), P (or O) or S (or T) is a selection of C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic rings further substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
J connected to I (or H), G (or F), K (or L), M (or N) or S (or T) is a selection of C1-C8 disubstituted (fused) saturated and unsaturated carbocyclic or heterocyclic rings further substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
D (or E) connected to B (or C) or G (or F); I (or H) connected to G (or F); P (or O) connected to R (or Q) or M (or N); K (or L) connected to N (or M) is a selection of C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic rings substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
B and C, D and E, R and Q, P and O, I and H, G and F, K and L, M and N or S and T are ═X where X is selected from sulfur, oxygen, nitrogen, NR1R2, and ═CR1R2
Figure US20030195168A1-20031016-C00008
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A′-T′ are independently selected from hydrogen, R4, R5, R6, F, Cl, Br, I, CN, COR4, CO2R4, OR4, SR4, NR4R5, CONR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, S02NR4R5, S03NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(R4)2, (C═X)R6 or X(C═X)R6 where X is selected from sulfur, oxygen and nitrogen;
R4 and R5 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR4, SR4, NR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, SO2NR4R5, SO3NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(P4)2]alkyl;
R6 is selected from R4, R5, CN, COR4, CO2R4, OR4, SR4, NR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, SO2NR4R5, SO3NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(R4)2;
E′ and R′ or H′ and O′ is a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R6, including epoxides and thioepoxides;
O′ connected to M′ (or N′) or Q′ (or P′); R′ connected to Q′ (or P′) or S′ (or T′); S′ (or T′) connected to A′ (or B′); A′ (or B′) connected to C′ (or D′); E′ connected to C′ (or D′) or F′ (or G′); H′ connected to I′; I′ connected to J′; J′ connected to K′; K′ connected to L′; L′ connected to M′ (or N′) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R6, (C═X)R6 and X(C═X)R6, including epoxides and thioepoxides;
A′, B′ and C′, D′ and F′, G′ and M′, N′ and P′, Q′ and S′, T′ are ═X where X is selected from sulfur, oxygen, nitrogen, NR4R5, (C═X)R6, X(C═X)R6, and ═CR7R8;
R7 and R8 are each independently selected from R6, (C═X)R6 and X(C═X)R6
Figure US20030195168A1-20031016-C00009
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A1-T1 are independently selected from hydrogen, R9, R10, R11, F, Cl, Br, I, CN, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2, (C═X)R11 or X(C═X)R11 where X is selected from sulfur, oxygen and nitrogen;
R9 and R10 are each independently selected from C1-C20 alkyl (branched and straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and straight chained), C2-C10 alkynyl (branched and straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2]alkyl;
R11 is selected from R9, R10, CN, COR9, CO2R9, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2;
B1 and R1, E1 and Ö1 and Ë1 and M1 are selected from a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R11, including epoxides and thioepoxides;
A1 (or Ä1 connected to Á1 (or Ã1) or T1 (or S1) B1 connected to Á1 (or Ã1) or C1 (or D1) E1 connected to Ë1 or C1 (or D1); Ë1 connected to É1 (or F1); G1 (or H1) connected to É1 (or F1) or I1 (or J1); K1 (or L1) connected to I1 (or J1) or M1; M1 connected to O1 (or N1); Ö1 connected O1 (or N1) or P1 (or Q1); R1 connected P1 (or Q1) or S1 (or T1) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R11, (C═X)R11 and X(C═X)R11, including epoxides and thioepoxides;
A1, Ä and Á, Ã and C1, D1 and F1, É and G1, H1 and I1, J1 and K1, L1 and N1, O1 and P1, Q1 and S1, T1 are ═X where X is selected from sulfur, oxygen, nitrogen, NR9R10, including (C═X)R11 and X(C═X)R11, and ═CR12R13;
R12 and R13 are independently selected from R11, (C═X)R11 and X(C═X)R11
Figure US20030195168A1-20031016-C00010
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A2-X2 are independently selected from hydrogen, R14, R15, R16, F, Cl, Br, I, CN, OR14, SR14, NR14R15, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14), (C═Y)R16 or Y(C═Y)R16 where Y is selected from sulfur, oxygen and nitrogen;
R14 and R15 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR14, SR14, NR14R10, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14)2]alkyl;
R16 is selected from R14, R15, CN, COR14, CO2R15, OR14, SR14, NR14R15, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14)2;
E2 and V2, H2 and S2, and I2 and P2 are C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R16, including epoxides and thioepoxides;
A2 (or B2) connected to C2 (or D2) or W2 (or X2); E2 connected to C2 (or D2) or F2 (or G2); H2 connected to F2 (or G2) or I2; I2 connected to J2 (or K2); L2 (or M2) connected to J2 (or K2) or N2 (or O2); R2 (or Q2) connected to P2 or S2; V2 connected to U2 (or T2) or W2 (or X2) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R16, (C═Y)R16 and Y(C═Y)R16, including epoxides and thioepoxides;
A2, B2; C2, D2; F2, G2; J2, K2; L2, M2; N2, O2; Q2, R2; U2, T2 and X2, W2 are ═Y where Y is selected from sulfur, oxygen, nitrogen, NR14R15 and ═CR17R18;
R17 and R18 are independently selected from R16, (C═Y)R16 and Y(C═Y)R16
Figure US20030195168A1-20031016-C00011
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A3-Z3 are independently selected from hydrogen, R19, R20, R21, F, Cl, Br, I, CN, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R,9)3, P(O)(R19)3, Si(R19)3, B(R19)2, (C═Ø)R21 or Ø(C═Ø)R21 where Ø is sullur, oxygen and nitrogen;
R19 and R20 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R19)3, P(═O)(R19)3, Si(R19)3, B(R19)2]alkyl;
R21 is selected from R19, R20, CN, COR19, CO2R19, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R19)3, P(═O)(R19)3, Si(R19)3, B(R19)2;
D3 connected to X3 is a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R21, including epoxides and thioepoxides;
A3 (or Ä3) connected to B3 (or C3) or E3 (or X); D3 connected to B3 (or C3) or E3 (or F3); G3 (or H3) connected to E3 (or F3) or I3 (or J3); L3 (or K3) connected to I3 (or J3) or M3 (or N3); O3 (or Ö3) connected to N3 (or M3) or P3 (or Q3). S3 (or R3) connected to Q3 (or P3) or U3 (or T3). W3 (or V3) connected to U3 (or T3) or X3; X3 connected to Y3 (or Z3) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R21, (C═Ø)R21 and Ø (C═Ø)R21, including epoxides and thioepoxides;
A3, Ä3; B3, C3; E3, F3; G3, H3; I3, J3; K3, L3; M3, N3; O3, Ö3; Q3, P3, S3, R3, U3, T3, W3, V3, and Z3, Y3 are ═Ø where Ø is selected from sulfur, oxygen, nitrogen, NR19R20, and ═CR22R23; and
R22 and R23 are selected from R21, (C═Ø)R21 and Ø(C═Ø)R21;
and which chemical agent or derivative or chemical analogue thereof is capable of modulating PKC activity, PKC-gene expression or PKC enzyme turnover and wherein said chemical agent or its derivatives or chemical analogues is administered for a time and under conditions sufficient to ameliorate one or more symptoms associated with a PKC-related condition or disorder.
2. A method according to claim 1 wherein the chemical agent is represented by the general formula (VI):
Figure US20030195168A1-20031016-C00012
wherein:
R24, R25 and R26 are independently selected from hydrogen, R27, R28, F, Cl, Br, I, CN, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, SO2R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2, (C═X)R29 or X(C═X)R29 where X is selected from sulfur, oxygen and nitrogen;
R27 and R28 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, SO2R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2]alkyl;
R29 is selected from R27, R28, CN, COR27, CO2R27, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, S02R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2.
3. A method according to claim 2 wherein R24 is H.
4. A method according to claim 2 wherein R24 is OAcetyl.
5. A method according to claim 2 wherein R24 is OH.
6. A method according to claim 2 wherein R25 and R26 are OH.
7. A method according to claim 1 wherein the PKC-related condition is alcoholism.
8. A method according to claim 1 wherein the PKC-related condition is Alzheimer's disease.
9. A method according to claim 1 wherein the PKC-related condition is asthma.
10. A method according to claim 1 wherein the PKC-related condition is atherosclosis.
11. A method according to claim 1 wherein the PKC-related condition is atopic dermatitis.
12. A method according to claim 1 wherein the PKC-related condition is autoimmune disease.
13. A method according to claim 1 wherein the PKC-related condition is bipolar disorder.
14. A method according to claim I wherein the PKC-related condition is blood disorder.
15. A method according to claim 1 wherein the PKC-related condition is cardiac hypertrophy.
16. A method according to claim 1 wherein the PKC-related condition is depression.
17. A method according to claim 1 wherein the PKC-related condition is diabetes.
18. A method according to claim 1 wherein the PKC-related condition is hypertension.
19. A method according to claim 1 wherein the PKC-related condition is hyperplastic dermatosis.
20. A method according to claim 1 wherein the PKC-related condition is multiple sclerosis.
21. A method according to claim 1 wherein the PKC-related condition is mycardial ischemia.
22. A method according to claim 1 wherein the PKC-related condition is osteoarthritis.
23. A method according to claim 1 wherein the PKC-related condition is psoriasis.
24. A method according to claim 1 wherein the PKC-related condition is rheumatoid arthritis.
25. A method according to claim 1 wherein the PKC-related condition is transplantation.
26. A method according to claim 1 wherein the PKC-related condition is a latent virus.
27. A method according to claim 1 wherein the chemical agent is a jatrophane or a derivative thereof or a pharmaceutically acceptable salt of these.
28. A method according to claim 27 wherein said derivative is an ester derivative.
29. A method according to claim 27 wherein said derivative is an acetylated derivative.
30. A method according to claim 1 wherein said chemical agent is a pepluane or a derivative thereof or a pharmaceutically acceptable salt of these.
31. A method according to claim 30 wherein said derivative is an ester derivative.
32. A method according to claim 30 wherein said derivative is an acetylated derivative.
33. A method according to claim 1 wherein said chemical agent is a paraliane or a derivative thereof or a pharmaceutically acceptable salt of these.
34. A method according to claim 33 wherein said derivative is an ester derivative.
35. A method according to claim 33 wherein said derivative is an acetylated derivative.
36. A method according to claim 1 wherein said compound is an angeloyl-substituted ingenane or a derivative thereof or a pharmaceutically acceptable salt of these.
37. A method according to claim 36 wherein said derivative is an acetylated derivative.
38. A method according to claim 36 wherein said jatrophane is of conformation 2.
39. A method according to claim 27 or 30 or 33 or 36 wherein the derivative comprises a substitution as represented in any one of general formulae (I)-(VI).
40. A method according to claim 1 wherein said compound is 5,8,9,10,14-pentaacetoxy-3-benzoyloxy-15-hydroxypepluane (pepluane) or a derivative thereof or a pharmaceutically acceptable salt of these.
41. A method according to claim 40 wherein said derivative is an ester derivative.
42. A method according to claim 1 wherein said compound is 2,3,5,7,15-pentaacetoxy-9-nicotinoyloxy-14-oxojatropha-6(17),11E-diene (jatrophane 1) or a derivative thereof or a pharmaceutically acceptable salt of these.
43. A method according to claim 42 wherein said derivative is an ester derivative.
44. A method according to claim 1 wherein said compound is 2,5,7,8,9,14-hexaacetoxy-3-benzoyloxy-15-hydroxy-jatropha-6(17),11E-diene (jatrophane 2) or a derivative thereof or a pharmaceutically acceptable salt of these.
45. A method according to claim 44 wherein said derivative is an ester derivative.
46. A method according to claim 1 wherein said compound is 2,5,14-triacetoxy-3-benzoyloxy-8,15-dihydroxy-7-isobutyroyloxy-9-nicotinoyloxyjatropha-6(17), 11E-diene (jatrophane 3) or a derivative thereof or a pharmaceutically acceptable salt of these.
47. A method according to claim 46 wherein said derivative is an ester derivative.
48. A method according to claim 1 wherein said compound is 2,5,9,14-tetraacetoxy-3-benzoyloxy-8,15-dihydroxy-7-isobutyroyloxyjatropha-6(17), 11E-diene) (jatrophane 4) or a derivative thereof or a pharmaceutically acceptable salt of these.
49. A method according to claim 48 wherein said derivative is an ester derivative.
50. A method according to claim 1 wherein said compound is 2,5,7,14-tetraacetoxy-3-benzoyloxy-8,15-dihydroxy-9-nicotinoyloxyjatropha-6(17),11E-diene (jatrophane 5) or a derivative thereof or a pharmaceutically acceptable salt of these.
51. A method according to claim 50 wherein said derivative is an ester derivative.
52. A method according to claim 1 wherein said compound is 2,5,7,9,14-pentaacetoxy-3-benzoyloxy-8,15-dihydroxyjatropha-6(17), 11E-diene (jatrophane 6) or a derivative thereof or a pharmaceutically acceptable salt of these.
53. A method according to claim 52 wherein said derivative is an ester derivative.
54. A method according to claim 1 wherein said compound is 20-O-acetyl-ingenol-3-angelate or a derivative thereof or a pharmaceutically acceptable salt of these.
55. A method according to claim 54 wherein said derivative is an ester derivative.
56. A method according to claim 27 or 30 or 33 or 36 or 40 or 42 or 44 or 46 or 48 or 50 or 52 or 54 wherein said compound is provided in the form of a composition comprising a pharmaceutically- or cosmetically-acceptable carrier.
57. A method according to claim 56 wherein said carrier is selected from β-alanine betaine hydrochloride and t-4-hydroxy-N,N-dimethylproline.
58. A method of modulating the expression of a genetic sequence for the treatment or prophylaxis of a condition or disorder in a subject, said genetic sequence being under the control of a promoter whose activity is modulated by a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or chemical analogue thereof which chemical agent is a macrocyclic diterpene selected from compounds of the ingenane, pepluane and jatrophane families and which chemical agent or derivative or chemical analogue is represented by any one of the general formulae (I)-(V):
Figure US20030195168A1-20031016-C00013
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A-T are independently selected from hydrogen, R1, R2, R3, F, Cl, Br, I, CN, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2, (C═X)R3 or X(C═X)R3 where X is selected from sulfur, oxygen and nitrogen;
R1 and R2 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2]alkyl;
R3 is selected from R1, R2, CN, COR1, CO2R1, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2;
A connected to B (or C), D (or E, R (or Q), P (or O) or S (or T) is a selection of C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic rings further substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
J connected to I (or H), G (or F), K (or L), M (or N) or S (or T) is a selection of C1-C8 disubstituted (fused) saturated and unsaturated carbocyclic or heterocyclic rings further substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
D (or E) connected to B (or C) or G (or F); I (or H) connected to G (or F); P (or O) connected to R (or Q) or M (or N); K (or L) connected to N (or M) is a selection of C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic rings substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
B and C, D and E, R and Q, P and O, I and H, G and F, K and L, M and N or S and T are ═X where X is selected from sulfur, oxygen, nitrogen, NR1R2, and ═CR1R2
Figure US20030195168A1-20031016-C00014
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A′-T′ are independently selected from hydrogen, R4, R5, R6, F, Cl, Br, I, CN, COR4, CO2R4, OR4, SR4, NR4R5, CONR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, S02NR4R5, S03NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(R4)2, (C═X)R6 or X(C═X)R6 where X is selected from sulfur, oxygen and nitrogen;
R4 and R5 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR4, SR4, NR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, SO2NR4R5, SO3NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(P4)2]alkyl;
R6 is selected from R4, R5, CN, COR4, CO2R4, OR4, SR4, NR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, SO2NR4R5, SO3NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(R4)2;
E′ and R′ or H′ and O′ is a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R6, including epoxides and thioepoxides;
O′ connected to M′ (or N′) or Q′ (or P′); R′ connected to Q′ (or P′) or S′ (or T′); S′ (or T′) connected to A′ (or B′); A′ (or B′) connected to C′ (or D′); E′ connected to C′ (or D′) or F′ (or G′); H′ connected to I′; I′ connected to J′; J′ connected to K′; K′ connected to L′; L′ connected to M′ (or N′) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R6, (C═X)R6 and X(C═X)R6, including epoxides and thioepoxides;
A′, B′ and C′, D′ and F′, G′ and M′, N′ and P′, Q′ and S′, T′ are ═X where X is selected from sulfur, oxygen, nitrogen, NR4R5, (C═X)R6, X(C═X)R6, and ═CR7R8;
R7 and R8 are each independently selected from R6, (C═X)R6 and X(C═X)R6
Figure US20030195168A1-20031016-C00015
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A1-T1 are independently selected from hydrogen, R9, R10, R11, F, Cl, Br, I, CN, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2, (C═X)R11 or X(C═X)R11 where X is selected from sulfur, oxygen and nitrogen;
R9 and R10 are each independently selected from C1-C20 alkyl (branched and straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and straight chained), C2-C10 alkynyl (branched and straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2]alkyl;
R11 is selected from R9, R10, CN, COR9, CO2R9, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2;
B1 and R1, E1 and Ö1 and Ë1 and M1 are selected from a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R11, including epoxides and thioepoxides;
A1 (or Ä1 connected to Á1 (or Ã1) or T1 (or S1) B1 connected to Á1 (or Ã1) or C1 (or D1) E1 connected to Ë1 or C1 (or D1); Ë1 connected to É1 (or F1); G1 (or H1) connected to É1 (or F1) or I1 (or J1); K1 (or L1) connected to I1 (or J1) or M1; M1 connected to O1 (or N1); Ö1 connected O1 (or N1) or P1 (or Q1); R1 connected P1 (or Q1) or S1 (or T1) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R11, (C═X)R11 and X(C═X)R11, including epoxides and thioepoxides;
A1, Ä and Á, Ã and C1, D1 and F1, É and G1, H1 and I1, J1 and K1, L1 and N1, O1 and P1, Q1 and S1, T1 are ═X where X is selected from sulfur, oxygen, nitrogen, NR9R10, including (C═X)R11 and X(C═X)R11, and ═CR12R13;
R12 and R13 are independently selected from R11, (C═X)R11 and X(C═X)R11
Figure US20030195168A1-20031016-C00016
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A2-X2 are independently selected from hydrogen, R14, R15, R16, F, Cl, Br, I, CN, OR14, SR14, NR14R15, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14), (C═Y)R16 or Y(C═Y)R16 where Y is selected from sulfur, oxygen and nitrogen;
R14 and R15 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR14, SR14, NR14R10, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14)2]alkyl;
R16 is selected from R14, R15, CN, COR14, CO2R15, OR14, SR14, NR14R15, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14)2;
E2 and V2, H2 and S2, and I2 and P2 are C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R16, including epoxides and thioepoxides;
A2 (or B2) connected to C2 (or D2) or W2 (or X2); E2 connected to C2 (or D2) or F2 (or G2); H2 connected to F2 (or G2) or I2; I2 connected to J2 (or K2); L2 (or M2) connected to J2 (or K2) or N2 (or O2); R2 (or Q2) connected to P2 or S2; V2 connected to U2 (or T2) or W2 (or X2) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R16, (C═Y)R16 and Y(C═Y)R16, including epoxides and thioepoxides;
A2, B2; C2, D2; F2, G2; J2, K2; L2, M2; N2, O2; Q2, R2; U2, T2 and X2, W2 are ═Y where Y is selected from sulfur, oxygen, nitrogen, NR14R15 and ═CR17R18;
R17 and R18 are independently selected from R16, (C═Y)R16 and Y(C═Y)R16
Figure US20030195168A1-20031016-C00017
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A3-Z3 are independently selected from hydrogen, R19, R20, R21, F, Cl, Br, I, CN, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R,9)3, P(O)(R19)3, Si(R19)3, B(R19)2, (C═Ø)R21 or Ø(C═Ø)R21 where Ø is sullur, oxygen and nitrogen;
R19 and R20 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R19)3, P(═O)(R19)3, Si(R19)3, B(R19)2]alkyl;
R21 is selected from R19, R20, CN, COR19, CO2R19, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R19)3, P(═O)(R19)3, Si(R19)3, B(R19)2;
D3 connected to X3 is a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R21, including epoxides and thioepoxides;
A3 (or Ä3) connected to B3 (or C3) or E3 (or X); D3 connected to B3 (or C3) or E3 (or F3); G3 (or H3) connected to E3 (or F3) or I3 (or J3); L3 (or K3) connected to I3 (or J3) or M3 (or N3); O3 (or Ö3) connected to N3 (or M3) or P3 (or Q3). S3 (or R3) connected to Q3 (or P3) or U3 (or T3). W3 (or V3) connected to U3 (or T3) or X3; X3 connected to Y3 (or Z3) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R21, (C═Ø)R21 and Ø (C═Ø)R21, including epoxides and thioepoxides;
A3, Ä3; B3, C3; E3, F3; G3, H3; I3, J3; K3, L3; M3, N3; O3, Ö3; Q3, P3, S3, R3, U3, T3, W3, V3, and Z3, Y3 are ═Ø where Ø is selected from sulfur, oxygen, nitrogen, NR19R20, and ═CR22R23; and
R22 and R23 are selected from R21, (C═Ø)R21 and Ø(C═Ø)R21;
and which chemical agent or derivative or chemical analogue thereof is capable of modulating PKC activity, PKC-gene expression or PKC enzyme turnover, said method comprising administering to said subject an expression facilitating amount of said chemical agent or derivative or chemical analogue thereof for a time and under conditions sufficient to facilitate the expression of said genetic sequence to thereby ameliorate one or more symptoms associated with said condition or disorder.
59. A method according to claim 58 wherein the chemical agent is represented by the general formula (VI):
Figure US20030195168A1-20031016-C00018
wherein:
R24, R25 and R26 are independently selected from hydrogen, R27, R28, F, Cl, Br, I, CN, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, SO2R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2, (C═X)R29 or X(C═X)R29 where X is selected from sulfur, oxygen and nitrogen;
R27 and R28 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, SO2R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2]alkyl;
R29 is selected from R27, R28, CN, COR27, CO2R27, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, S02R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2.
60. A method of modulating the expression of a genetic sequence by the administration of a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or chemical analogue thereof which chemical agent is a macrocyclic diterpene selected from compounds of the ingenane, pepluane and jatrophane familes and which chemical agent or derivative or chemical analogue is represented by any one of the general formulae (I)-(V):
Figure US20030195168A1-20031016-C00019
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A-T are independently selected from hydrogen, R1, R2, R3, F, Cl, Br, I, CN, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2, (C═X)R3 or X(C═X)R3 where X is selected from sulfur, oxygen and nitrogen;
R1 and R2 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2]alkyl;
R3 is selected from R1, R2, CN, COR1, CO2R1, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2;
A connected to B (or C), D (or E, R (or Q), P (or O) or S (or T) is a selection of C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic rings further substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
J connected to I (or H), G (or F), K (or L), M (or N) or S (or T) is a selection of C1-C8 disubstituted (fused) saturated and unsaturated carbocyclic or heterocyclic rings further substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
D (or E) connected to B (or C) or G (or F); I (or H) connected to G (or F); P (or O) connected to R (or Q) or M (or N); K (or L) connected to N (or M) is a selection of C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic rings substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
B and C, D and E, R and Q, P and O, I and H, G and F, K and L, M and N or S and T are ═X where X is selected from sulfur, oxygen, nitrogen, NR1R2, and ═CR1R2
Figure US20030195168A1-20031016-C00020
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A′-T′ are independently selected from hydrogen, R4, R5, R6, F, Cl, Br, I, CN, COR4, CO2R4, OR4, SR4, NR4R5, CONR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, S02NR4R5, S03NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(R4)2, (C═X)R6 or X(C═X)R6 where X is selected from sulfur, oxygen and nitrogen;
R4 and R5 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR4, SR4, NR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, SO2NR4R5, SO3NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(P4)2]alkyl;
R6 is selected from R4, R5, CN, COR4, CO2R4, OR4, SR4, NR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, SO2NR4R5, SO3NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(R4)2;
E′ and R′ or H′ and O′ is a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R6, including epoxides and thioepoxides;
O′ connected to M′ (or N′) or Q′ (or P′); R′ connected to Q′ (or P′) or S′ (or T′); S′ (or T′) connected to A′ (or B′); A′ (or B′) connected to C′ (or D′); E′ connected to C′ (or D′) or F′ (or G′); H′ connected to I′; I′ connected to J′; J′ connected to K′; K′ connected to L′; L′ connected to M′ (or N′) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R6, (C═X)R6 and X(C═X)R6, including epoxides and thioepoxides;
A′, B′ and C′, D′ and F′, G′ and M′, N′ and P′, Q′ and S′, T′ are ═X where X is selected from sulfur, oxygen, nitrogen, NR4R5, (C═X)R6, X(C═X)R6, and ═CR7R8;
R7 and R8 are each independently selected from R6, (C═X)R6 and X(C═X)R6
Figure US20030195168A1-20031016-C00021
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A1-T1 are independently selected from hydrogen, R9, R10, R11, F, Cl, Br, I, CN, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2, (C═X)R11 or X(C═X)R11 where X is selected from sulfur, oxygen and nitrogen;
R9 and R10 are each independently selected from C1-C20 alkyl (branched and straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and straight chained), C2-C10 alkynyl (branched and straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2]alkyl;
R11 is selected from R9, R10, CN, COR9, CO2R9, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2;
B1 and R1, E1 and Ö1 and Ë1 and M1 are selected from a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R11, including epoxides and thioepoxides;
A1 (or Ä1 connected to Á1 (or Ã1) or T1 (or S1) B1 connected to Á1 (or Ã1) or C1 (or D1) E1 connected to Ë1 or C1 (or D1); Ë1 connected to É1 (or F1); G1 (or H1) connected to É1 (or F1) or I1 (or J1); K1 (or L1) connected to I1 (or J1) or M1; M1 connected to O1 (or N1); Ö1 connected O1 (or N1) or P1 (or Q1); R1 connected P1 (or Q1) or S1 (or T1) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R11, (C═X)R11 and X(C═X)R11, including epoxides and thioepoxides;
A1, Ä and Á, Ã and C1, D1 and F1, É and G1, H1 and I1, J1 and K1, L1 and N1, O1 and P1, Q1 and S1, T1 are ═X where X is selected from sulfur, oxygen, nitrogen, NR9R10, including (C═X)R11 and X(C═X)R11, and ═CR12R13;
R12 and R13 are independently selected from R11, (C═X)R11 and X(C═X)R11
Figure US20030195168A1-20031016-C00022
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A2-X2 are independently selected from hydrogen, R14, R15, R16, F, Cl, Br, I, CN, OR14, SR14, NR14R15, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14), (C═Y)R16 or Y(C═Y)R16 where Y is selected from sulfur, oxygen and nitrogen;
R14 and R15 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR14, SR14, NR14R10, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14)2]alkyl;
R16 is selected from R14, R15, CN, COR14, CO2R15, OR14, SR14, NR14R15, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14)2;
E2 and V2, H2 and S2, and I2 and P2 are C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R16, including epoxides and thioepoxides;
A2 (or B2) connected to C2 (or D2) or W2 (or X2); E2 connected to C2 (or D2) or F2 (or G2); H2 connected to F2 (or G2) or I2; I2 connected to J2 (or K2); L2 (or M2) connected to J2 (or K2) or N2 (or O2); R2 (or Q2) connected to P2 or S2; V2 connected to U2 (or T2) or W2 (or X2) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R16, (C═Y)R16 and Y(C═Y)R16, including epoxides and thioepoxides;
A2, B2; C2, D2; F2, G2; J2, K2; L2, M2; N2, O2; Q2, R2; U2, T2 and X2, W2 are ═Y where Y is selected from sulfur, oxygen, nitrogen, NR14R15 and ═CR17R18;
R17 and R18 are independently selected from R16, (C═Y)R16 and Y(C═Y)R16
Figure US20030195168A1-20031016-C00023
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A3-Z3 are independently selected from hydrogen, R19, R20, R21, F, Cl, Br, I, CN, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R,9)3, P(O)(R19)3, Si(R19)3, B(R19)2, (C═Ø)R21 or Ø(C═Ø)R21 where Ø is sullur, oxygen and nitrogen;
R19 and R20 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R19)3, P(═O)(R19)3, Si(R19)3, B(R19)2]alkyl;
R21 is selected from R19, R20, CN, COR19, CO2R19, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R19)3, P(═O)(R19)3, Si(R19)3, B(R19)2;
D3 connected to X3 is a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R21, including epoxides and thioepoxides;
A3 (or Ä3) connected to B3 (or C3) or E3 (or X); D3 connected to B3 (or C3) or E3 (or F3); G3 (or H3) connected to E3 (or F3) or I3 (or J3); L3 (or K3) connected to I3 (or J3) or M3 (or N3); O3 (or Ö3) connected to N3 (or M3) or P3 (or Q3). S3 (or R3) connected to Q3 (or P3) or U3 (or T3). W3 (or V3) connected to U3 (or T3) or X3; X3 connected to Y3 (or Z3) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R21, (C═Ø)R21 and Ø (C═Ø)R21, including epoxides and thioepoxides;
A3, Ä3; B3, C3; E3, F3; G3, H3; I3, J3; K3, L3; M3, N3; O3, Ö3; Q3, P3, S3, R3, U3, T3, W3, V3, and Z3, Y3 are ═Ø where Ø is selected from sulfur, oxygen, nitrogen, NR19R20, and ═CR22R23; and
R22 and R23 are selected from R21, (C═Ø)R21 and Ø(C═Ø)R21;
and which chemical agent or derivative or chemical analogue thereof is capable of modulating PKC activity, PKC-gene expression or PKC enzyme turnover and wherein said chemical agent or its derivatives or chemical analogues is administered for a time and under conditions sufficient to facilitate the expression of said genetic sequence.
61. A method according to claim 60 wherein the chemical agent is represented by the general formula (VI):
Figure US20030195168A1-20031016-C00024
wherein:
R24, R25 and R26 are independently selected from hydrogen, R27, R28, F, Cl, Br, I, CN, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, SO2R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2, (C═X)R29 or X(C═X)R29 where X is selected from sulfur, oxygen and nitrogen;
R27 and R28 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, SO2R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2]alkyl;
R29 is selected from R27, R28, CN, COR27, CO2R27, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, S02R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2.
62. A method of stimulating the activation or function of a promoter by the administration of a chemical agent obtainable from a plant of the Euphorbiaceae family or a derivative or chemical analogue thereof which chemical agent is a macrocyclic diterpene selected from compounds of the ingenane, pepluane and jatrophane families and which chemical agent or derivative or chemical analogue is represented by any one of the general formulae (I)-(V):
Figure US20030195168A1-20031016-C00025
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A-T are independently selected from hydrogen, R1, R2, R3, F, Cl, Br, I, CN, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2, (C═X)R3 or X(C═X)R3 where X is selected from sulfur, oxygen and nitrogen;
R1 and R2 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2]alkyl;
R3 is selected from R1, R2, CN, COR1, CO2R1, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2;
A connected to B (or C), D (or E, R (or Q), P (or O) or S (or T) is a selection of C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic rings further substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
J connected to I (or H), G (or F), K (or L), M (or N) or S (or T) is a selection of C1-C8 disubstituted (fused) saturated and unsaturated carbocyclic or heterocyclic rings further substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
D (or E) connected to B (or C) or G (or F); I (or H) connected to G (or F); P (or O) connected to R (or Q) or M (or N); K (or L) connected to N (or M) is a selection of C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic rings substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
B and C, D and E, R and Q, P and O, I and H, G and F, K and L, M and N or S and T are ═X where X is selected from sulfur, oxygen, nitrogen, NR1R2, and ═CR1R2
Figure US20030195168A1-20031016-C00026
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A′-T′ are independently selected from hydrogen, R4, R5, R6, F, Cl, Br, I, CN, COR4, CO2R4, OR4, SR4, NR4R5, CONR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, S02NR4R5, S03NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(R4)2, (C═X)R6 or X(C═X)R6 where X is selected from sulfur, oxygen and nitrogen;
R4 and R5 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR4, SR4, NR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, SO2NR4R5, SO3NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(P4)2]alkyl;
R6 is selected from R4, R5, CN, COR4, CO2R4, OR4, SR4, NR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, SO2NR4R5, SO3NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(R4)2;
E′ and R′ or H′ and O′ is a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R6, including epoxides and thioepoxides;
O′ connected to M′ (or N′) or Q′ (or P′); R′ connected to Q′ (or P′) or S′ (or T′); S′ (or T′) connected to A′ (or B′); A′ (or B′) connected to C′ (or D′); E′ connected to C′ (or D′) or F′ (or G′); H′ connected to I′; I′ connected to J′; J′ connected to K′; K′ connected to L′; L′ connected to M′ (or N′) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R6, (C═X)R6 and X(C═X)R6, including epoxides and thioepoxides;
A′, B′ and C′, D′ and F′, G′ and M′, N′ and P′, Q′ and S′, T′ are ═X where X is selected from sulfur, oxygen, nitrogen, NR4R5, (C═X)R6, X(C═X)R6, and ═CR7R8;
R7 and R8 are each independently selected from R6, (C═X)R6 and X(C═X)R6
Figure US20030195168A1-20031016-C00027
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A1-T1 are independently selected from hydrogen, R9, R10, R11, F, Cl, Br, I, CN, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2, (C═X)R11 or X(C═X)R11 where X is selected from sulfur, oxygen and nitrogen;
R9 and R10 are each independently selected from C1-C20 alkyl (branched and straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and straight chained), C2-C10 alkynyl (branched and straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2]alkyl;
R11 is selected from R9, R10, CN, COR9, CO2R9, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2;
B1 and R1, E1 and Ö1 and Ë1 and M1 are selected from a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R11, including epoxides and thioepoxides;
A1 (or Ä1 connected to Á1 (or Ã1) or T1 (or S1) B1 connected to Á1 (or Ã1) or C1 (or D1) E1 connected to Ë1 or C1 (or D1); Ë1 connected to É1 (or F1); G1 (or H1) connected to É1 (or F1) or I1 (or J1); K1 (or L1) connected to I1 (or J1) or M1; M1 connected to O1 (or N1); Ö1 connected O1 (or N1) or P1 (or Q1); R1 connected P1 (or Q1) or S1 (or T1) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R11, (C═X)R11 and X(C═X)R11, including epoxides and thioepoxides;
A1, Ä and Á, Ã and C1, D1 and F1, É and G1, H1 and I1, J1 and K1, L1 and N1, O1 and P1, Q1 and S1, T1 are ═X where X is selected from sulfur, oxygen, nitrogen, NR9R10, including (C═X)R11 and X(C═X)R11, and ═CR12R13;
R12 and R13 are independently selected from R11, (C═X)R11 and X(C═X)R11
Figure US20030195168A1-20031016-C00028
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A2-X2 are independently selected from hydrogen, R14, R15, R16, F, Cl, Br, I, CN, OR14, SR14, NR14R15, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14), (C═Y)R16 or Y(C═Y)R16 where Y is selected from sulfur, oxygen and nitrogen;
R14 and R15 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR14, SR14, NR14R10, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14)2]alkyl;
R16 is selected from R14, R15, CN, COR14, CO2R15, OR14, SR14, NR14R15, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14)2;
E2 and V2, H2 and S2, and I2 and P2 are C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R16, including epoxides and thioepoxides;
A2 (or B2) connected to C2 (or D2) or W2 (or X2); E2 connected to C2 (or D2) or F2 (or G2); H2 connected to F2 (or G2) or I2; I2 connected to J2 (or K2); L2 (or M2) connected to J2 (or K2) or N2 (or O2); R2 (or Q2) connected to P2 or S2; V2 connected to U2 (or T2) or W2 (or X2) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R16, (C═Y)R16 and Y(C═Y)R16, including epoxides and thioepoxides;
A2, B2; C2, D2; F2, G2; J2, K2; L2, M2; N2, O2; Q2, R2; U2, T2 and X2, W2 are ═Y where Y is selected from sulfur, oxygen, nitrogen, NR14R15 and ═CR17R18;
R17 and R18 are independently selected from R16, (C═Y)R16 and Y(C═Y)R16
Figure US20030195168A1-20031016-C00029
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A3-Z3 are independently selected from hydrogen, R19, R20, R21, F, Cl, Br, I, CN, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R,9)3, P(O)(R19)3, Si(R19)3, B(R19)2, (C═Ø)R21 or Ø(C═Ø)R21 where Ø is sullur, oxygen and nitrogen;
R19 and R20 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R19)3, P(═O)(R19)3, Si(R19)3, B(R19)2]alkyl;
R21 is selected from R19, R20, CN, COR19, CO2R19, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R19)3, P(═O)(R19)3, Si(R19)3, B(R19)2;
D3 connected to X3 is a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R21, including epoxides and thioepoxides;
A3 (or Ä3) connected to B3 (or C3) or E3 (or X); D3 connected to B3 (or C3) or E3 (or F3); G3 (or H3) connected to E3 (or F3) or I3 (or J3); L3 (or K3) connected to I3 (or J3) or M3 (or N3); O3 (or Ö3) connected to N3 (or M3) or P3 (or Q3). S3 (or R3) connected to Q3 (or P3) or U3 (or T3). W3 (or V3) connected to U3 (or T3) or X3; X3 connected to Y3 (or Z3) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R21, (C═Ø)R21 and Ø (C═Ø)R21, including epoxides and thioepoxides;
A3, Ä3; B3, C3; E3, F3; G3, H3; I3, J3; K3, L3; M3, N3; O3, Ö3; Q3, P3, S3, R3, U3, T3, W3, V3, and Z3, Y3 are ═Ø where Ø is selected from sulfur, oxygen, nitrogen, NR19R20, and ═CR22R23; and
R22 and R23 are selected from R21, (C═Ø)R21 and Ø(C═Ø)R21;
and which chemical agent or derivative or chemical analogue thereof is capable of modulating PKC activity, PKC-gene expression or PKC enzyme turnover and wherein said chemical agent or its derivatives or chemical analogues is administered for a time and under conditions sufficient to stimulate the activation or function of said promoter.
63. A method according to claim 60 wherein the chemical agent is represented by the general formula (VI):
Figure US20030195168A1-20031016-C00030
wherein:
R24, R25 and R26 are independently selected from hydrogen, R27, R28, F, Cl, Br, I, CN, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, SO2R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2, (C═X)R29 or X(C═X)R29 where X is selected from sulfur, oxygen and nitrogen;
R27 and R28 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, SO2R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2]alkyl;
R29 is selected from R27, R28, CN, COR27, CO2R27, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, S02R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2. A method according to claim 2 wherein R24 is H.
64. A method according to claim 59 or 61 or 63 wherein R24 is OAcetyl.
65. A method according to claim 59 or 61 or 63 wherein R24 is OH.
66. A method according to claim 59 or 61 or 63 wherein R25 and R26 are OH.
67. A method according to claim 1 or 59 or 61 or 63 wherein the plant is of the genus selected from Acalypha, Acidoton, Actinostemon, Adelia, Adenocline, Adenocrepis, Adenophaedra, Adisca, Agrostistachys, Alchornea, Alchorneopsis, Alcinaeanthus, Alcoceria, Aleurites, Amanoa, Andrachne, Angostyles, Anisophylluin, Antidesma, Aphora, Aporosa, Aporosella, Argythamnia, Astrococcus, Astrogyne, Baccanrea, Baliospermum, Bernardia, Beyeriopsis, Bischofia, Blachia, Blumeodondron, Bonania, Bradleia, Breynia, Breyniopsis, Briedelia, Buraeavia, Caperonia, Caryodendron, Celianella, Cephalocroton, Chaenotheca, Chaetocarpus, Chamaesyce, Cheilosa, Chiropetalum, Choriophyllum, Cicca, Chaoxylon, Cleidon, Cleistanthus, Cluytia, Cnesmone, Cnidoscolus, Coccoceras, Codiaeum, Coelodiscus, Conami, Conceveiba, Conceveibastrum, Conceveibum, Corythea, Croizatia, Croton, Crotonopsis, Crozophora, Cubanthus, Cunuria, Dactylostemon, Dalechampia, Dendrocousinsia, Diaspersus, Didymocistus, Dimorphocalyx, Discocarpus, Ditaxis, Dodecastingma, Drypetes, Dysopsis, Elateriospermum, Endadenium, Endospermum, Erismanthus, Erythrocarpus, Erythrochilus, Eumecanthus, Euphorbia, Euphorbiodendron, Excoecaria, Flueggea, Calearia, Garcia, Gavarretia, Gelonium, Giara, Givotia, Glochidion, Clochidionopsis, Glycydendron, Gymnanthes, Gymnosparia, Haematospermum, Hendecandra, Hevea, Hieronima, Hieronyma, Hippocrepandra, Homalanthus, Hymenocardia, Janipha, Jatropha, Julocroton, Lasiocroton, Leiocarpus, Leonardia, Lepidanthus, Leucocroton, Mabea, Macaranga, Mallotus, Manihot, Mappa, Maprounea, Melanthesa, Mercurialis, Mettenia, Micrandra, Microdesmis, Microelus, Microstachy, Maocroton, Monadenium, Mozinna, Neoscortechinia, Omalanthus, Omphalea, Ophellantha, Orbicularia, Ostodes, Oxydectes, Palenga, Pantadenia, Paradrypeptes, Pausandra, Pedilanthus, Pera, Peridium, Petalostigma, Phyllanthus, Picrodendro, Pierardia, Pilinophytum, Pimeleodendron, Piranhea, Platygyna, Plukenetia, Podocalyx, Poinsettia, Poraresia, Prosartema, Pseudanthus, Pycnocoma, Quadrasia, Reverchonia, Richeria, Richeriella, Ricinella, Ricinocarpus, Rottlera, Sagotia, Sanwithia, Sapium, Savia, Sclerocroton, Sebastiana, Securinega, Senefeldera, Senefilderopsis, Serophyton, Siphonia, Spathiostemon, Spixia, Stillingia, Strophioblachia, Synadenium, Tetracoccus, Tetraplandra, Tetrorchidium, Thyrsanthera, Tithymalus, Trageia, Trewia, Trigonostemon, Tyria and Xylophylla.
68. A method according to claim 67 wherein the plant is of the genus Euphorbia.
69. A method according to claim 68 wherein the species of Euphorbia is selected from Euphorbia aaron-rossii, Euphorbia abbreviata, Euphorbia acuta, Euphorbia alatocaulis, Euphorbia albicaulis, Euphorbia algomarginata, Euphorbia aliceae, Euphorbia alta, Euphorbia anacampseros, Euphorbia andromedae, Euphorbia angusta, Euphorbia anthonyi, Euphorbia antiguensis, Euphorbia apocynifolia, Euphorbia arabica, Euphorbia ariensis, Euphorbia arizonica, Euphorbia arkansana, Euphorbia arteagae, Euphorbia arundelana, Euphorbia astroites, Euphorbia atrococca, Euphorbia baselicis, Euphorbia batabanensis, Euphorbia bergeri, Euphorbia bermudiana, Euphorbia bicolor, Euphorbia biformis, Euphorbia bifurcata, Euphorbia bilobata, Euphorbia biramensis, Euphorbia biuncialis, Euphorbia blepharostipula, Euphorbia blodgetti, Euphorbia boerhaavioides, Euphorbia boliviana, Euphorbia bracei, Euphorbia brachiata, Euphorbia brachycera, Euphorbia brandegee, Euphorbia brittonii, Euphorbia caesia, Euphorbia calcicola, Euphorbia campestris, Euphorbia candelabrum, Euphorbia capitellata, Euphorbia carmenensis, Euphorbia carunculata, Euphorbia cayensis, Euphorbia celastroides, Euphorbia chalicophila, Euphorbia chamaerrhodos, Euphorbia chamaesula, Euphorbia chiapensis, Euphorbia chiogenoides, Euphorbia cinerascens, Euphorbia clarionensis, Euphorbia colimae, Euphorbia colorata, Euphorbia commutata, Euphorbia consoquitlae, Euphorbia convolvuloides, Euphorbia corallifera, Euphorbia creberrima, Euphorbia crenulata, Euphorbia cubensis, Euphorbia cuspidata, Euphorbia cymbiformis, Euphorbia darlingtonii, Euphorbia defoliata, Euphorbia degeneri, Euphorbia deltoidea, Euphorbia dentata, Euphorbia depressa Euphorbia dictyosperma, Euphorbia dictyosperma, Euphorbia dioeca, Euphorbia discoidalis, Euphorbia dorsiventralis, Euphorbia drumondii, Euphorbia duclouxii, Euphorbia dussii, Euphorbia eanophylla, Euphorbia eggersii, Euphorbia eglandulosa, Euphorbia elata, Euphorbia enalla, Euphorbia eriogonoides, Euphorbia eriophylla, Euphorbia esculaeformis, Euphorbia espirituensis, Euphorbia esula, Euphorbia excisa, Euphorbia exclusa, Euphorbia exstipitata, Euphorbia exstipulata, Euphorbia fendleri, Euphorbia filicaulis, Euphorbia filiformis, Euphorbia florida, Euphorbia fruticulosa, Euphorbia garber, Euphorbia gaumerii, Euphorbia gerardiana, Euphorbia geyeri, Euphorbia glyptosperma, Euphorbia gorgonis, Euphorbia gracilior, Euphorbia gracillima, Euphorbia gradyi, Euphorbia graminea, Euphorbia graminiea Euphorbia grisea, Euphorbia guadalajarana, Euphorbia guanarensis, Euphorbia gymnadenia, Euphorbia haematantha, Euphorbia hedyotoides, Euphorbia heldrichii, Euphorbia helenae, Euphorbia helleri, Euphorbia helwigii, Euphorbia henricksonii, Euphorbia heterophylla, Euphorbia hexagona, Euphorbia hexagonoides, Euphorbia hinkleyorum, Euphorbia hintonii, Euphorbia hirtula, Euphorbia hirta, Euphorbia hooveri, Euphorbia humistrata, Euphorbia hypericifolia, Euphorbia inundata, Euphorbia involuta, Euphorbia jaliscensis, Euphorbia jejuna, Euphorbia johnston, Euphorbia juttae, Euphorbia knuthii, Euphorbia lasiocarpa, Euphorbia lata, Euphorbia latazi, Euphorbia latericolor, Euphorbia laxiflora Euphorbia lecheoides, Euphorbia ledienii, Euphorbia leucophylla, Euphorbia lineata, Euphorbia linguiformis, Euphorbia longecornuta, Euphorbia longepetiolata, Euphorbia longeramosa, Euphorbia longinsulicola, Euphorbia longipila, Euphorbia lupulina, Euphorbia lurida, Euphorbia lycioides, Euphorbia macropodoides, macvaughiana, Euphorbia manca, Euphorbia mandoniana, Euphorbia mangleti, Euphorbia mango, Euphorbia marylandica, Euphorbia mayana, Euphorbia melanadenia, Euphorbia melanocarpa, Euphorbia meridensis, Euphorbia mertonii, Euphorbia mexiae, Euphorbia microcephala, Euphorbia microclada, Euphorbia micromera, Euphorbia misella, Euphorbia missurica, Euphorbia montana, Euphorbia montereyana, Euphorbia multicaulis, Euphorbia multiformis, Euphorbia multinodis, Euphorbia multiseta, Euphorbia muscicola, Euphorbia neomexicana, Euphorbia nephradenia, Euphorbia niqueroana, Euphorbia oaxacana, Euphorbia occidentalis, Euphorbia odontodenia, Euiphorbia olivacea, Euphorbia olowaluana, Euphorbia opthalmica, Euphorbia ovata, Euphorbia pachypoda, Euphorbia pachyrhiza, Euphorbia padifolia, Euphorbia palmeri, Euphorbia paludicola, Euphorbia parciflora, Euphorbia parishii, Euphorbia parryi, Euphorbia paxiana, Euphorbia pediculifera, Euphorbia peplidion, Euphorbia peploides, Euphorbia peplus, Euphorbia pergamena, Euphorbia perlignea, Euphorbia petaloidea, Euphorbia petaloidea, Euphorbia petrina, Euphorbia picachensis, Euphorbia pilosula, Euphorbia pilulifera, Euphorbia pinariona, Euphorbia pinetorum, Euphorbia pionosperma, Euphorbia platysperma, Euphorbia plicata, Euphorbia poeppigii, Euphorbia poliosperma, Euphorbia polycarpa, Euphorbia polycnemoides, Euphorbia polyphylla, Euphorbia portoricensis, Euphorbia portulacoides Euphorbia portulana, Euphorbia preslii, Euphorbia prostrata, Euphorbia pteroneura, Euphorbia pycnanthema, Euphorbia ramosa, Euphorbia rapulum, Euphorbia remyi, Euphorbia retroscabra, Euphorbia revoluta, Euphorbia rivularis, Euphorbia robusta, Euphorbia romosa, Eiphorbia rubida, Euphorbia rubrosperma, Euphorbia rupicola, Euphorbia sanmartensis, Euphorbia saxatilis M. Bieb, Euphorbia schizoloba, Euphorbia sclerocyathium, Euphorbia scopulorum, Euphorbia senilis, Euphorbia serpyllifolia, Euphorbia serrula, Euphorbia setiloba Engelm, Euphorbia sonorae, Euphorbia soobyi, Euphorbia sparsiflora, Euphorbia sphaerosperna, Euphorbia syphilitica, Euphorbia spruceana, Euphorbia subcoerulea, Euphorbia stellata, Euphorbia submammilaris, Euphorbia subpeltata, Euphorbia subpubens, Euphorbia subreniforme, Euphorbia subtrifoliata, Euphorbia succedanea, Euphorbia tamaulipasana, Euphorbia telephioides, Euphorbia tenuissima, Euphorbia tetrapora, Euphorbia tirucalli, Euphorbia tomentella, Euphorbia tomentosa, Euphorbia torralbasii, Euphorbia tovariensis, Euphorbia trachysperma, Euphorbia tricolor, Euphorbia troyana, Euphorbia tuerckheimii, Euphorbia turczaninowii, Euphorbia unibellulata, Euphorbia undulata, Euphorbia vermiformis, Euphorbia versicolor, Euphorbia villifera, Euphorbia violacea, Euphorbia whitei, Euphorbia xanti Engelm, Euphorbia xylopoda Greenm., Euiphorbia yayalesia Urb., Euphorbia yungasensis, Euphorbia zeravschanica and Euphorbia zinniiflora.
70. A method according to claim 69 wherein the species of Euphorbia is Euphorbia peplus.
71. A method according to claim 59 or 61 or 63 wherein the chemical agent is a jatrophane or a derivative thereof or a pharmaceutically acceptable salt of these.
72. A method according to claim 71 wherein said derivative is an ester derivative.
73. A method according to claim 71 wherein said derivative is an acetylated derivative.
74. A method according to claim 59 or 61 or 63 wherein said chemical agent is a peptuane or a derivative thereof or a pharmaceutically acceptable salt of these.
75. A method according to claim 74 wherein said derivative is an ester derivative.
76. A method according to claim 74 wherein said derivative is an acetylated derivative.
77. A method according to claim 59 or 61 or 63 wherein said chemical agent is a paraliane or a derivative thereof or a pharmaceutically acceptable salt of these.
78. A method according to claim 77 wherein said derivative is an ester derivative.
79. A method according to claim 77 wherein said derivative is an acetylated derivative.
80. A method according to claim 59 or 61 or 63 wherein said compound is an angeloyl-substituted ingenane or a derivative thereof or a pharmaceutically acceptable salt of these.
81. A method according to claim 80 wherein said derivative is an acetylated derivative.
82. A method according to claim 80 wherein said jatrophane is of conformation 2.
83. A method according to claim 71 or 74 or 77 or 80 wherein the derivative comprises a substitution as represented in any one of general formulae (I)-(VI).
84. A method according to claim 59 or 61 or 63 wherein said compound is 5,8,9,10,14-pentaacetoxy-3-benzoyloxy-15-hydroxypepluane (pepluane) or a derivative thereof or a pharmaceutically acceptable salt of these.
85. A method according to claim 84 wherein said derivative is an ester derivative.
86. A method according to claim 59 or 61 or 63 wherein said compound is 2,3,5,7,15-pentaacetoxy-9-nicotinoyloxy-14-oxojatropha-6(17), 11E-diene (jatrophane 1) or a derivative thereof or a pharmaceutically acceptable salt of these.
87. A method according to claim 86 wherein said derivative is an ester derivative.
88. A method according to claim 59 or 61 or 63 wherein said compound is 2,5,7,8,9,14-hexaacetoxy-3-benzoyloxy-15-hydroxy jatropha-6(17), 11E-diene (jatrophane 2) or a derivative thereof or a pharmaceutically acceptable salt of these.
89. A method according to claim 88 wherein said derivative is an ester derivative.
90. A method according to claim 59 or 61 or 63 wherein said compound is 2,5,14-triacetoxy-3-benzoyloxy-8,15-dihydroxy-7-isobutyroyloxy-9-nicotinoyloxyjatropha-6(17), 11E-diene (jatrophane 3) or a derivative thereof or a pharmaceutically acceptable salt of these.
91. A method according to claim 90 wherein said derivative is an ester derivative.
92. A method according to claim 59 or 61 or 63 wherein said compound is 2,5,9,14-tetraacetoxy-3-benzoyloxy-8,15-dihydroxy-7-isobutyroyloxyjatropha-6(17),11E-diene) (jatrophane 4) or a derivative thereof or a pharmaceutically acceptable salt of these.
93. A method according to claim 92 wherein said derivative is an ester derivative.
94. A method according to claim 59 or 61 or 63 wherein said compound is 2,5,7,14-tetraacetoxy-3-benzoyloxy-8,15-dihydroxy-9-nicotinoyloxyjatropha-6(17),11E-diene (jatrophane 5) or a derivative thereof or a pharmaceutically acceptable salt of these.
95. A method according to claim 94 wherein said derivative is an ester derivative.
96. A method according to claim 59 or 61 or 63 wherein said compound is 2,5,7,9,14-pentaacetoxy-3-benzoyloxy-8,15-dihydroxyjatropha-6(17), 11E-diene (jatrophane 6) or a derivative thereof or a pharmaceutically acceptable salt of these.
97. A method according to claim 96 wherein said derivative is an ester derivative.
98. A method according to claim 59 or 61 or 63 wherein said compound is 20-O-acetyl-ingenol-3-angelate or a derivative thereof or a pharmaceutically acceptable salt of these.
99. A method according to claim 98 wherein said derivative is an ester derivative.
100. A method according to claim 71 or 74 or 77 or 80 or 84 or 86 or 88 or 90 or 92 or 94 or 96 or 98 wherein said compound is provided in the form of a composition comprising a pharmaceutically- or cosmetically-acceptable carrier.
101. A method according to claim 100 wherein said carrier is selected from β-alanine betaine hydrochloride and t-4-hydroxy-N,N-dimethylproline.
102. A method for the treatment or prophylaxis of a PKC-related condition in a subject, said method comprising the administration to said subject of a symptom-ameliorating or immunopotentiating effective amount of a macrocyclic diterpene obtainable from a Euphorbiaceae plant or its botanical or horticultural relative, said macrocyclic diterpene being selected from an ingenane, pepluane or jatrophane, or a derivative or chemical analogue thereof, having the structure represented by any one of the general formulae (I)-(V)
Figure US20030195168A1-20031016-C00031
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A-T are independently selected from hydrogen, R1, R2, R3, F, Cl, Br, I, CN, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2, (C═X)R3 or X(C═X)R3 where X is selected from sulfur, oxygen and nitrogen;
R1 and R2 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2]alkyl;
R3 is selected from R1, R2, CN, COR1, CO2R1, OR1, SR1, NR1R2, N(═O)2, NR1OR2, ONR1R2, SOR1, SO2R1, SO3R1, SONR1R2, SO2NR1R2, SO3NR1R2, P(R1)3, P(═O)(R1)3, Si(R1)3, B(R1)2;
A connected to B (or C), D (or E, R (or Q), P (or O) or S (or T) is a selection of C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic rings further substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
J connected to I (or H), G (or F), K (or L), M (or N) or S (or T) is a selection of C1-C8 disubstituted (fused) saturated and unsaturated carbocyclic or heterocyclic rings further substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
D (or E) connected to B (or C) or G (or F); I (or H) connected to G (or F); P (or O) connected to R (or Q) or M (or N); K (or L) connected to N (or M) is a selection of C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic rings substituted by R3, (C═X)R3 and X(C═X)R3, including epoxides and thioepoxides;
B and C, D and E, R and Q, P and O, I and H, G and F, K and L, M and N or S and T are ═X where X is selected from sulfur, oxygen, nitrogen, NR1R2, and ═CR1R2
Figure US20030195168A1-20031016-C00032
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A′-T′ are independently selected from hydrogen, R4, R5, R6, F, Cl, Br, I, CN, COR4, CO2R4, OR4, SR4, NR4R5, CONR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, S02NR4R5, S03NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(R4)2, (C═X)R6 or X(C═X)R6 where X is selected from sulfur, oxygen and nitrogen;
R4 and R5 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR4, SR4, NR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, SO2NR4R5, SO3NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(P4)2]alkyl;
R6 is selected from R4, R5, CN, COR4, CO2R4, OR4, SR4, NR4R5, N(═O)2, NR4OR5, ONR4R5, SOR4, SO2R4, SO3R4, SONR4R5, SO2NR4R5, SO3NR4R5, P(R4)3, P(═O)(R4)3, Si(R4)3, B(R4)2;
E′ and R′ or H′ and O′ is a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R6, including epoxides and thioepoxides;
O′ connected to M′ (or N′) or Q′ (or P′); R′ connected to Q′ (or P′) or S′ (or T′); S′ (or T′) connected to A′ (or B′); A′ (or B′) connected to C′ (or D′); E′ connected to C′ (or D′) or F′ (or G′); H′ connected to I′; I′ connected to J′; J′ connected to K′; K′ connected to L′; L′ connected to M′ (or N′) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R6, (C═X)R6 and X(C═X)R6, including epoxides and thioepoxides;
A′, B′ and C′, D′ and F′, G′ and M′, N′ and P′, Q′ and S′, T′ are ═X where X is selected from sulfur, oxygen, nitrogen, NR4R5, (C═X)R6, X(C═X)R6, and ═CR7R8;
R7 and R8 are each independently selected from R6, (C═X)R6 and X(C═X)R6
Figure US20030195168A1-20031016-C00033
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A1-T1 are independently selected from hydrogen, R9, R10, R11, F, Cl, Br, I, CN, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2, (C═X)R11 or X(C═X)R11 where X is selected from sulfur, oxygen and nitrogen;
R9 and R10 are each independently selected from C1-C20 alkyl (branched and straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and straight chained), C2-C10 alkynyl (branched and straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2]alkyl;
R11 is selected from R9, R10, CN, COR9, CO2R9, OR9, SR9, NR9R10, N(═O)2, NR9OR10, ONR9R10, SOR9, SO2R9, SO3R9, SONR9R10, SO2NR9R10, SO3NR9R10, P(R9)3, P(═O)(R9)3, Si(R9)3, B(R9)2;
B1 and R1, E1 and Ö1 and Ë1 and M1 are selected from a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R11, including epoxides and thioepoxides;
A1 (or Ä1 connected to Á1 (or Ã1) or T1 (or S1) B1 connected to Á1 (or Ã1) or C1 (or D1) E1 connected to Ë1 or C1 (or D1); Ë1 connected to É1 (or F1); G1 (or H1) connected to É1 (or F1) or I1 (or J1); K1 (or L1) connected to I1 (or J1) or M1; M1 connected to O1 (or N1); Ö1 connected O1 (or N1) or P1 (or Q1); R1 connected P1 (or Q1) or S1 (or T1) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R11, (C═X)R11 and X(C═X)R11, including epoxides and thioepoxides;
A1, Ä and Á, Ã and C1, D1 and F1, É and G1, H1 and I1, J1 and K1, L1 and N1, O1 and P1, Q1 and S1, T1 are ═X where X is selected from sulfur, oxygen, nitrogen, NR9R10, including (C═X)R11 and X(C═X)R11, and ═CR12R13;
R12 and R13 are independently selected from R11, (C═X)R11 and X(C═X)R11
Figure US20030195168A1-20031016-C00034
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A2-X2 are independently selected from hydrogen, R14, R15, R16, F, Cl, Br, I, CN, OR14, SR14, NR14R15, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14), (C═Y)R16 or Y(C═Y)R16 where Y is selected from sulfur, oxygen and nitrogen;
R14 and R15 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR14, SR14, NR14R10, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14)2]alkyl;
R16 is selected from R14, R15, CN, COR14, CO2R15, OR14, SR14, NR14R15, N(═O)2, NR14OR15, ONR14R15, SOR14, SO2R14, SO3R14, SONR14R15, SO2NR14R15, SO3NR14R15, P(R14)3, P(═O)(R14)3, Si(R14)3, B(R14)2;
E2 and V2, H2 and S2, and I2 and P2 are C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R16, including epoxides and thioepoxides;
A2 (or B2) connected to C2 (or D2) or W2 (or X2); E2 connected to C2 (or D2) or F2 (or G2); H2 connected to F2 (or G2) or I2; I2 connected to J2 (or K2); L2 (or M2) connected to J2 (or K2) or N2 (or O2); R2 (or Q2) connected to P2 or S2; V2 connected to U2 (or T2) or W2 (or X2) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R16, (C═Y)R16 and Y(C═Y)R16, including epoxides and thioepoxides;
A2, B2; C2, D2; F2, G2; J2, K2; L2, M2; N2, O2; Q2, R2; U2, T2 and X2, W2 are ═Y where Y is selected from sulfur, oxygen, nitrogen, NR14R15 and ═CR17R18;
R17 and R18 are independently selected from R16, (C═Y)R16 and Y(C═Y)R16
Figure US20030195168A1-20031016-C00035
wherein:
n is 0-10 atoms selected from carbon, oxygen, nitrogen, sulfur, phosphorus, silicon, boron, arsenic and selenium, wherein the ring defined by said atoms is saturated or unsaturated, including epoxides and thioepoxides;
A3-Z3 are independently selected from hydrogen, R19, R20, R21, F, Cl, Br, I, CN, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R,9)3, P(O)(R19)3, Si(R19)3, B(R19)2, (C═Ø)R21 or Ø(C═Ø)R21 where Ø is sullur, oxygen and nitrogen;
R19 and R20 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R19)3, P(═O)(R19)3, Si(R19)3, B(R19)2]alkyl;
R21 is selected from R19, R20, CN, COR19, CO2R19, OR19, SR19, NR19R20, N(═O)2, NR19OR20, ONR19R20, SOR19, SO2R19, SO3R19, SONR19R20, SO2NR19R20, SO3NR19R20, P(R19)3, P(═O)(R19)3, Si(R19)3, B(R19)2;
D3 connected to X3 is a C2-C8 saturated or unsaturated carbocyclic or heterocyclic ring system further substituted by R21, including epoxides and thioepoxides;
A3 (or Ä3) connected to B3 (or C3) or E3 (or X); D3 connected to B3 (or C3) or E3 (or F3); G3 (or H3) connected to E3 (or F3) or I3 (or J3); L3 (or K3) connected to I3 (or J3) or M3 (or N3); O3 (or Ö3) connected to N3 (or M3) or P3 (or Q3). S3 (or R3) connected to Q3 (or P3) or U3 (or T3). W3 (or V3) connected to U3 (or T3) or X3; X3 connected to Y3 (or Z3) are C1-C8 disubstituted (fused) saturated or unsaturated carbocyclic or heterocyclic ring systems further substituted by R21, (C═Ø)R21 and Ø (C═Ø)R21, including epoxides and thioepoxides;
A3, Ä3; B3, C3; E3, F3; G3, H3; I3, J3; K3, L3; M3, N3; O3, Ö3; Q3, P3, S3, R3, U3, T3, W3, V3, and Z3, Y3 are ═Ø where Ø is selected from sulfur, oxygen, nitrogen, NR19R20, and ═CR22R23; and
R22 and R23 are selected from R21, (C═Ø)R21 and Ø(C═Ø)R21;
and which chemical agent or derivative or chemical analogue thereof is capable of modulating PKC activity, PKC-gene expression or PKC enzyme turnover and wherein said chemical agent or its derivatives or chemical analogues is administered for a time and under conditions sufficient to ameliorate one or more symptoms associated with said biological entity and wherein said chemical agent exhibits a potency of agent (PA) of >10, wherein the PA=ΣIV where IV is a numerical value associated with a particular feature as listed below:
Feature Value An ability to modulate PKC activity or effect +1 An ability to induce bipolar dendritic activity +1 An ability to displace phorbol dibutyrate from +1 binding to PKC An ability to induce respiratory burst in leucocytes +1 An ability to stimulate phagocytosis in peripheral +1 blood mononuclear cells Derived from a member of the Euphorbiaceae family +1 Derived from E. peplus +3 Water extractible from the sap of Euphorbia sp. +2 An ability to activate latent virus +4 A lower tumor promotion activity than TPA/PMA +2
or pharmaceutically acceptable salts of these, said chemical agent being administered for a time and under conditions sufficient to ameliorate at least one symptom caused by or associated with said PKC-related condition.
103. A method according to claim 102 wherein the chemical agent is represented by the general formula (VI):
Figure US20030195168A1-20031016-C00036
wherein:
R24, R25 and R26 are independently selected from hydrogen, R27, R28, F, Cl, Br, I, CN, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, SO2R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2, (C═X)R29 or X(C═X)R29 where X is selected from sulfur, oxygen and nitrogen;
R27 and R28 are each independently selected from C1-C20 alkyl (branched and/or straight chained), C1-C20 arylalkyl, C3-C8 cycloalkyl, C6-C14 aryl, C1-C14 heteroaryl, C1-C14 heterocycle, C2-C10 alkenyl (branched and/or straight chained), C2-C10 alkynyl (branched and/or straight chained), C1-C10 heteroarylalkyl, C1-C10 alkoxyalkyl, C1-C10 haloalkyl, dihaloalkyl, trihaloalkyl, haloalkoxy, C1-C10 [CN, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, SO2R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2]alkyl;
R29 is selected from R27, R28, CN, COR27, CO2R27, OR27, SR27, NR27R28, N(═O)2, NR27OR28, ONR27R28, SOR27, S02R27, SO3R27, SONR27R28, SO2NR27R28, SO3NR27R28, P(R27)3, P(═O)(R27)3, Si(R27)3, B(R27)2.
104. A method according to claim 102 wherein R24 is H.
105. A method according to claim 102 wherein R24 is OAcetyl.
106. A method according to claim 102 wherein R24 is OH.
107. A method according to claim 102 wherein R25 and R26 are OH.
108. A method according to claim 101 wherein the plant is of the genus selected from Acalypha, Acidoton, Actinostemon, Adelia, Adenocline, Adenocrepis, Adenophaedra, Adisca, Agrostistachys, Alchornea, Alchorneopsis, Alcinaeanthus, Alcoceria, Aleurites, Amanoa, Andrachne, Angostyles, Anisophyllum, Antidesma, Aphora, Aporosa, Aporosella, Argythamnia, Astrococcus, Astrogyne, Baccanrea, Baliospermum, Bernardia, Beyeriopsis, Bischofia, Blachia, Blumeodondron, Bonania, Bradleia, Breynia, Breyniopsis, Briedelia, Buraeavia, Caperonia, Caryodendron, Celianella, Cephalocroton, Chaenotheca, Chaetocarpus, Chamaesyce, Cheilosa, Chiropetalum, Choriophyllum, Cicca, Chaoxylon, Cleidon, Cleistanthus, Cluytia, Cnesmone, Cnidoscolus, Coccoceras, Codiaeum, Coelodiscus, Conami, Conceveiba, Conceveibastrum, Coneeveibum, Corythea, Croizatia, Croton, Crotonopsis, Crozophora, Cubanthus, Cunuria, Dactylostemon, Dalechampia, Dendrocousinsia, Diaspersus, Didymocistus, Dimorphocalyx, Discocarpus, Ditaxis, Dodecastingma, Drypetes, Dysopsis, Elateriospermum, Endadenium, Endospermum, Erismanthus, Erythrocarpus, Erythrochilis, Eumecanthus, Euphorbia, Euphorbiodendron, Excoecaria, Flueggea, Calearia, Garcia, Gavarretia, Gelonium, Giara, Givotia, Glochidion, Clochidionopsis, Glycydendron, Gymnanthes, Gymnosparia, Haematospermum, Hendecandra, Hevea, Hieronima, Hieronyma, Hippocrepandra, Homalanthus, Hymenocardia, Janipha, Jatropha, Julocroton, Lasiocroton, Leiocarpus, Leonardia, Lepidanthus, Leucocroton, Mabea, Macaranga, Mallotus, Manihot, Mappa, Maprounea, Melanthesa, Mercurialis, Mettenia, Micrandra, Microdesmis, Microelus, Microstachy, Maocroton, Monadenium, Mozinna, Neoscortechinia, Omalanthus, Omphalea, Ophellantha, Orbicilaria, Ostodes, Oxydectes, Palenga, Pantadenia, Paradrypeptes, Pausandra, Pedilanthus, Pera, Peridium, Petalostigma, Phyllanthus, Picrodendro, Pierardia, Pilinophytum, Pimeleodendron, Piranhea, Platygyna, Plukenetia, Podocalyx, Poinsettia, Poraresia, Prosartema, Pseudanthus, Pycnocoma, Quadrasia, Reverchonia, Richeria, Richeriella, Ricinella, Ricinocarpus, Rottlera, Sagotia, Sanwithia, Sapium, Savia, Sclerocroton, Sebastiana, Securinega, Senefeldera, Senefilderopsis, Serophyton, Siphonia, Spathiostemon, Spixia, Stillingia, Strophioblachia, Synadenium, Tetracoccus, Tetraplandra, Tetrorchidium, Thyrsanthera, Tithymalus, Trageia, Trewia, Trigonostemon, Tyria and Xylophylla.
109. A method according to claim 102 wherein the plant is of the genus Euphorbia.
110. A method according to claim 109 wherein the species of Euphorbia is selected from Euphorbia aaron-rossii, Euphorbia abbreviata, Euphorbia acuta, Euphorbia alatocaulis, Euphorbia albicaulis, Euphorbia algomarginata, Euphorbia aliceae, Euphorbia alta, Euphorbia anacampseros, Euphorbia andromedae, Euphorbia angusta, Euphorbia anthonyi, Euphorbia antiguensis, Euphorbia apocynifolia, Euphorbia arabica, Euphorbia ariensis, Euphorbia arizonica, Euphorbia arkansana, Euphorbia arteagae, Euphorbia arundelana, Euphorbia astroites, Euphorbia atrococca, Euphorbia baselicis, Euphorbia batabanensis, Euphorbia bergeri, Euphorbia bermudiana, Euphorbia bicolor, Euphorbia biformis, Euphorbia bifurcata, Euphorbia bilobata, Euphorbia biramensis, Euphorbia biuncialis, Euphorbia blepharostipula, Euphorbia blodgetti, Etiphorbia boerhaavioides, Euphorbia boliviana, Euphorbia bracei, Euphorbia brachiata, Euphorbia brachycera, Euphorbia brandegee, Euphorbia brittonii, Euphorbia caesia, Euphorbia calcicola, Euphorbia campestris, Euphorbia candelabrum, Euphorbia capitellata, Euphorbia carmenensis, Euphorbia carunculata, Euphorbia cayensis, Euphorbia celastroides, Euphorbia chalicophila, Euphorbia chamaerrhodos, Euphorbia chamaesula, Euphorbia chiapensis, Euphorbia chiogenoides, Euphorbia cinerascens, Euphorbia clarionensis, Euphorbia colimae, Euphorbia colorata, Euphorbia commutata, Euphorbia consoquitlae, Euphorbia convolvuloides, Euphorbia corallifera, Euphorbia creberrima, Euphorbia crenulata, Euphorbia cubensis, Euphorbia cuspidata, Euphorbia cymbiformis, Euphorbia darlingtonii, Euphorbia defoliata, Euphorbia degeneri, Euphorbia deltoidea, Euphorbia dentata, Euphorbia depressa Euphorbia dictyosperma, Euphorbia dictyosperma, Euphorbia dioeca, Euphorbia discoidalis, Euphorbia dorsiventralis, Euphorbia drumondii, Euphorbia duclouxii, Euphorbia dussii, Euphorbia eanophylla, Euphorbia eggersii, Euphorbia eglandulosa, Euphorbia elata, Euphorbia enalla, Euphorbia eriogonoides, Euphorbia eriophylla, Euphorbia esculaeformis, Euphorbia espirituensis, Euphorbia esula, Euphorbia excisa, Euphorbia exclusa, Euphorbia exstipitata, Euphorbia exstipulata, Euphorbia fendleri, Euphorbia filicaulis, Euphorbia filiformis, Euphorbia florida, Euphorbia fruticulosa, Euphorbia garber, Euphorbia gaumerii, Euphorbia gerardiana, Euphorbia geyeri, Euphorbia glyptosperma, Euphorbia gorgonis, Euphorbia gracilior, Euphorbia gracillima, Euphorbia gradyi, Euphorbia graminea, Euphorbia graminiea Euphorbia grisea, Euphorbia guadalajarana, Euphorbia guanarensis, Euphorbia gymnadenia, Euphorbia haematantha, Euphorbia hedyotoides, Euphorbia heldrichii, Euphorbia helenae, Euphorbia helleri, Euphorbia helwigii, Euphorbia henricksonii, Euphorbia heterophylla, Euphorbia hexagona, Euphorbia hexagonoides, Euphorbia hinkleyorum, Euphorbia hintonii, Euphorbia hirtula, Euphorbia hirta, Euphorbia hooveri, Euphorbia humistrata, Euphorbia hypericifolia, Euphorbia inundata, Euphorbia involuta, Euphorbia jaliscensis, Euphorbia jejuna, Euphorbia johnston, Euphorbia juttae, Euphorbia knuthii, Euphorbia lasiocarpa, Euphorbia lata, Euphorbia latazi, Euphorbia latericolor, Euphorbia laxiflora Euphorbia lecheoides, Euphorbia ledienii, Euphorbia leucophylla, Euphorbia lineata, Euphorbia linguiformis, Euphorbia longecornuta, Euphorbia longepetiolata, Euphorbia longeramosa, Euphorbia longinsulicola, Euphorbia longipila, Euphorbia lupulina, Euphorbia lurida, Euphorbia lycioides, Euphorbia macropodoides, macvaughiana, Euphorbia manca, Euphorbia mandoniana, Euphorbia mangleti, Euphorbia mango, Euphorbia marylandica, Euphorbia mayana, Euphorbia melanadenia, Euphorbia melanocarpa, Euphorbia meridensis, Euphorbia mertonii, Euphorbia mexiae, Euphorbia microcephala, Euphorbia microclada, Euphorbia micromera, Euphorbia misella, Euphorbia missurica, Euphorbia montana, Euphorbia montereyana, Euphorbia multicaulis, Euphorbia multiformis, Euphorbia multinodis, Euphorbia multiseta, Euphorbia muscicola, Euphorbia neomexicana, Euphorbia nephradenia, Euphorbia niqueroana, Euphorbia oaxacana, Euphorbia occidentalis, Euphorbia odontodenia, Euphorbia olivacea, Euphorbia olowaluana, Euphorbia opthalmica, Euphorbia ovata, Euphorbia pachypoda, Euphorbia pachyrhiza, Euphorbia padifolia, Euphorbia palmeri, Euphorbia paludicola, Euphorbia parciflora, Euphorbia parishii, Euphorbia parryi, Euphorbia paxiana, Euphorbia pediculifera, Euphorbia peplidion, Euphorbia peploides, Euphorbia peplus, Euphorbia pergamena, Euphorbia perlignea, Euphorbia petaloidea, Euphorbia petaloidea, Euphorbia petrina, Euphorbia picachensis, Euphorbia pilosula, Euphorbia pilulifera, Euphorbia pinariona, Euphorbia pinetorum, Euphorbia pionosperma, Euphorbia platysperma, Euphorbia plicata, Euphorbia poeppigii, Euphorbia poliosperma, Euphorbia polycarpa, Etuphorbia polycnemoides, Euphorbia polyphylla, Euphorbia portoricensis, Euphorbia portulacoides Euphorbia portulana, Euphorbia preslii, Euphorbia prostrata, Euiphorbia pteroneura, Euphorbia pycnanthema, Euphorbia ramosa, Euphorbia rapulum, Euphorbia remyi, Euphorbia retroscabra, Euphorbia revoluta, Euphorbia rivularis, Euphorbia robusta, Euphorbia romosa, Euphorbia rubida, Euphorbia rubrosperma, Euphorbia rupicola, Euphorbia sanmartensis, Euphorbia saxatilis M. Bieb, Euphorbia schizoloba, Euphorbia sclerocyathium, Euphorbia scopulorum, Euphorbia senilis, Euphorbia serpyllifolia, Euphorbia serrula, Euphorbia setiloba Engelm, Euphorbia sonorae, Euphorbia soobyi, Euphorbia sparsiflora, Euphorbia sphaerosperma, Euphorbia syphilitica, Euphorbia spruceana, Euphorbia subcoerulea, Euphorbia stellata, Euphorbia submammilaris, Euphorbia subpeltata, Euphorbia subpubens, Euphorbia subreniforme, Euphorbia subtrifoliata, Euphorbia succedanea, Euphorbia tamaulipasana, Euphorbia telephioicles, Euphorbia tenuissima, Euphorbia tetrapora, Euphorbia tirucalli, Euphorbia tomentella, Euphorbia tomentosa, Euphorbia torralbasii, Euphorbia tovariensis, Euphorbia trachysperma, Euphorbia tricolor, Euphorbia troyana, Euphorbia tuerckheimii, Euphorbia turczaninowii, Euphorbia umbellulata, Eiphorbia undulata, Euphorbia vermiformis, Euphorbia versicolor, Euphorbia villifera, Euphorbia violacea, Euphorbia whitei, Euphorbia xanti Engelm, Euphorbia xylopoda Greenm., Euphorbia yayalesia Urb., Euphorbia yungasensis, Euphorbia zeravschanica and Euphorbia zinniiflora.
111. A method according to claim 110 wherein the species of Euphorbia is Euphorbia peplus.
112. A method according to claim 102 wherein the chemical agent is a jatrophane or a derivative thereof or a pharmaceutically acceptable salt of these.
113. A method according to claim 112 wherein said derivative is an ester derivative.
114. A method according to claim 112 wherein said derivative is an acetylated derivative.
115. A method according to claim 102 wherein said chemical agent is a pepluane or a derivative thereof or a pharmaceutically acceptable salt of these.
116. A method according to claim 115 wherein said derivative is an ester derivative.
117. A method according to claim 115 wherein said derivative is an acetylated derivative.
118. A method according to claim 102 wherein said chemical agent is a paraliane or a derivative thereof or a pharmaceutically acceptable salt of these.
119. A method according to claim 118 wherein said derivative is an ester derivative.
120. A method according to claim 118 wherein said derivative is an acetylated derivative.
121. A method according to claim 102 wherein said compound is an angeloyl-substituted ingenane or a derivative thereof or a pharmaceutically acceptable salt of these.
122. A method according to claim 121 wherein said derivative is an acetylated derivative.
123. A method according to claim 121 wherein said jatrophane is of conformation 2.
124. A method according to claim 112 or 115 or 118 or 121 wherein the derivative comprises a substitution as represented in any one of general formulae (I)-(VI).
125. A method according to claim 102 wherein said compound is 5,8,9,10,14-pentaacetoxy-3-benzoyloxy-15-hydroxypepluane (pepluane) or a derivative thereof or a pharmaceutically acceptable salt of these.
126. A method according to claim 125 wherein said derivative is an ester derivative.
127. A method according to claim 102 wherein said compound is 2,3,5,7,15-pentaacetoxy-9-nicotinoyloxy-14-oxojatropha-6(17),11E-diene (jatrophane 1) or a derivative thereof or a pharmaceutically acceptable salt of these.
128. A method according to claim 127 wherein said derivative is an ester derivative.
129. A method according to claim 102 wherein said compound is 2,5,7,8,9,14-hexaacetoxy-3-benzoyloxy-15-hydroxy-jatropha-6(17),11E-diene (jatrophane 2) or a derivative thereof or a pharmaceutically acceptable salt of these.
130. A method according to claim 129 wherein said derivative is an ester derivative.
131. A method according to claim 102 wherein said compound is 2,5,14-triacetoxy-3-benzoyloxy-8,15-dihydroxy-7-isobutyroyloxy-9-nicotinoyloxyjatropha-6(17), 11E-diene (jatrophane 3) or a derivative thereof or a pharmaceutically acceptable salt of these.
132. A method according to claim 131 wherein said derivative is an ester derivative.
133. A method according to claim 102 wherein said compound is 2,5,9,14-tetraacetoxy-3-benzoyloxy-8,15-dihydroxy-7-isobutyroyloxyjatropha-6(17),11E-diene) (jatrophane 4) or a derivative thereof or a pharmaceutically acceptable salt of these.
134. A method according to claim 133 wherein said derivative is an ester derivative.
135. A method according to claim 102 wherein said compound is 2,5,7,14-tetraacetoxy-3-benzoyloxy-8,15-dihydroxy-9-nicotinoyloxyjatropha-6(17),11E-diene (jatrophane 5) or a derivative thereof or a pharmaceutically acceptable salt of these.
136. A method according to claim 135 wherein said derivative is an ester derivative.
137. A method according to claim 102 wherein said compound is 2,5,7,9,14-pentaacetoxy-3-benzoyloxy-8,15-dihydroxyjatropha-6(17),11E-diene (jatrophane 6) or a derivative thereof or a pharmaceutically acceptable salt of these.
138. A method according to claim 137 wherein said derivative is an ester derivative.
139. A method according to claim 102 wherein said compound is 20-O-acetyl-ingenol-3-angelate or a derivative thereof or a pharmaceutically acceptable salt of these.
140. A method according to claim 139 wherein said derivative is an ester derivative.
141. A method according to claim 112 or 115 or 118 or 121 or 125 or 127 or 129 or 131 or 133 or 135 or 137 or 139 wherein said compound is provided in the form of a composition comprising a pharmaceutically- or cosmetically-acceptable carrier.
142. A method according to claim 141 wherein said carrier is selected from β-alanine betaine hydrochloride and t-4-hydroxy-N,N-dimethylproline.
143. A computer program product for assessing the likely usefulness of a candidate compound or group of compounds for the treatment or prophylaxis of a PKC-related condition or disorder in a subject, said product comprising:
(1) code that receives as input index values for at least two features associated with said compound(s), wherein said features are selected from:
(a) the ability to modulate PKC activity or effect;
(b) the ability to induce bipolar dendritic activity;
(c) the ability to be derived from a member of the Euphorbiaceae family;
(d) the ability to be derived from E. peplus;
(e) the ability to be water extractable from the sap of a Euphorbia species;
(f) the ability to activate latent virus; or
(g) less tumor promoting capacity than TPA or MPA;
(2) code that adds said index values to provide a sum corresponding to a potency value for said compound(s); and
(3) a computer readable medium that stores the codes.
144. A computer for assessing the likely usefulness of a candidate compound or group of compounds for the treatment of a PKC-related conditinon or disorder in a subject, wherein said computer comprises:
(1) a machine-readable data storage medium comprising a data storage material encoded with machine-readable data, wherein said machine-readable data comprise index values for at least two features associated with said compound(s), wherein said features are selected from:
(a) the ability to modulate PKC activity or effect;
(b) the ability to induce bipolar dendritic activity;
(c) the ability to be derived from a member of the Euphorbiaceae family;
(d) the ability to be derived from E. peplus;
(e) the ability to be water extractable from the sap of a Euphorbia species;
(h) the ability to activate latent virus; or
(i) less tumor promoting capacity than TPA or MPA;
(2) a working memory for storing instructions for processing said machine-readable data;
(3) a central-processing unit coupled to said working memory and to said machine-readable data storage medium, for processing said machine readable data to provide a sum of said index values corresponding to a potency value for said compound(s); and
(4) an output hardware coupled to said central processing unit, for receiving said potency value.
US10/315,319 2000-06-07 2002-12-09 Therapeutic agents - III Abandoned US20030195168A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
AUPQ8017A AUPQ801700A0 (en) 2000-06-07 2000-06-07 Enzyme and viral activation
AUPQ8017 2000-06-07
PCT/AU2001/000678 WO2001093883A1 (en) 2000-06-07 2001-06-07 Therapeutic agents - iii

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2001/000678 Continuation-In-Part WO2001093883A1 (en) 2000-06-07 2001-06-07 Therapeutic agents - iii

Publications (1)

Publication Number Publication Date
US20030195168A1 true US20030195168A1 (en) 2003-10-16

Family

ID=3822089

Family Applications (6)

Application Number Title Priority Date Filing Date
US10/315,319 Abandoned US20030195168A1 (en) 2000-06-07 2002-12-09 Therapeutic agents - III
US10/315,288 Expired - Fee Related US7449492B2 (en) 2000-06-07 2002-12-09 Therapeutic agents—I
US10/315,318 Expired - Fee Related US7838555B2 (en) 2000-06-07 2002-12-09 Macrocyclic diterpenes for the treatment and prophylaxis of acne vulgaris
US11/000,712 Abandoned US20050209192A1 (en) 2000-06-07 2004-12-01 Therapeutic agents - II
US12/543,087 Abandoned US20100048698A1 (en) 2000-06-07 2009-08-18 Macrocyclic diterpenes for treating conditions associated with protein kinase c
US13/034,172 Expired - Fee Related US9314458B2 (en) 2000-06-07 2011-02-24 Topical use of ingenol-3-angelate or a salt thereof to treat skin cancer

Family Applications After (5)

Application Number Title Priority Date Filing Date
US10/315,288 Expired - Fee Related US7449492B2 (en) 2000-06-07 2002-12-09 Therapeutic agents—I
US10/315,318 Expired - Fee Related US7838555B2 (en) 2000-06-07 2002-12-09 Macrocyclic diterpenes for the treatment and prophylaxis of acne vulgaris
US11/000,712 Abandoned US20050209192A1 (en) 2000-06-07 2004-12-01 Therapeutic agents - II
US12/543,087 Abandoned US20100048698A1 (en) 2000-06-07 2009-08-18 Macrocyclic diterpenes for treating conditions associated with protein kinase c
US13/034,172 Expired - Fee Related US9314458B2 (en) 2000-06-07 2011-02-24 Topical use of ingenol-3-angelate or a salt thereof to treat skin cancer

Country Status (8)

Country Link
US (6) US20030195168A1 (en)
EP (3) EP1296697A4 (en)
JP (3) JP5822418B2 (en)
AU (1) AUPQ801700A0 (en)
BR (3) BR0111423A (en)
CA (3) CA2411642C (en)
NZ (3) NZ522426A (en)
WO (3) WO2001093885A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030166613A1 (en) * 2000-06-07 2003-09-04 Aylward James Harrison Therapeutic agents - I
US20050070565A1 (en) * 2003-09-26 2005-03-31 Arnsten Amy F.T. Chelerythrine, analogs thereof and their use in the treatment of bipolar disorder and other cognitive disorders
EP1838330A1 (en) * 2004-12-13 2007-10-03 Peplin Research Pty Ltd. Treatment of solid cancers
US7378445B2 (en) 2000-08-07 2008-05-27 Peplin Research Pty. Ltd. Treatment of prostate cancer
US7410656B2 (en) 1997-08-19 2008-08-12 Peplin Research Pty. Ltd. Anti-cancer compounds
US20090076132A1 (en) * 2005-03-11 2009-03-19 Gary Pekoe Antiviral compositions and methods of treatment
US20090215884A1 (en) * 2005-11-25 2009-08-27 Peplin Research Pty Ltd. Method for wound healing
KR101089314B1 (en) 2010-06-07 2011-12-02 한국식품연구원 Composition containing euphobiasteroid for prevention, treatment or improvement of obesity
US9895334B2 (en) 2007-04-30 2018-02-20 Leo Laboratories Limited Treatment of virally induced lesions

Families Citing this family (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2881568C (en) 2000-10-27 2019-09-24 Novartis Vaccines And Diagnostics, Inc. Nucleic acids and proteins from streptococcus groups a & b
US20050065205A1 (en) 2002-03-07 2005-03-24 Daniel Alkon Methods for Alzheimer's disease treatment and cognitive enhance
US6825229B2 (en) 2002-03-07 2004-11-30 Blanchette Rockefeller Neurosciences Institute Methods for Alzheimer's Disease treatment and cognitive enhancement
AU2003260102A1 (en) * 2002-08-26 2004-03-11 Chiron Corporation Conserved and specific streptococcal genomes
DE602004010059T2 (en) * 2003-07-24 2008-09-11 Teva Gyogyszergyar Zartköruen Muködo Reszvenytarsasag METHOD FOR PURIFYING MAKROLIDES
PT1648500E (en) * 2003-07-31 2014-10-10 Novartis Vaccines & Diagnostic Immunogenic compositions for streptococcus pyogenes
US8945589B2 (en) * 2003-09-15 2015-02-03 Novartis Vaccines And Diagnostics, Srl Immunogenic compositions for Streptococcus agalactiae
TW201207390A (en) 2004-05-18 2012-02-16 Brni Neurosciences Inst Method for screening agent for antidepressant activity
JP2008508320A (en) * 2004-07-29 2008-03-21 カイロン コーポレイション Immunogenic composition against gram positive bacteria such as STREPTOCOCCUSAGALACTIAE
DE102004044428A1 (en) * 2004-09-14 2006-03-30 Toximed Gmbh Process and pharmaceutical agent for the control of plasmodia
US7838010B2 (en) * 2004-10-08 2010-11-23 Novartis Vaccines And Diagnostics S.R.L. Immunogenic and therapeutic compositions for Streptococcus pyogenes
EP1807038A1 (en) * 2004-11-03 2007-07-18 Cognis France, S.A.S. An extract of a plant belonging to the genus plukenetia volubilis and its cosmetic use
US20060204600A1 (en) * 2005-03-11 2006-09-14 Paul Konz Dragon's blood anti-viral materials and methods
EP1915145A1 (en) 2005-07-29 2008-04-30 Blanchette Rockefeller Neurosciences Institute Use of a pkc activator, alone or combined with a pkc inhibitor to enhance long term memory
JP2009516651A (en) * 2005-11-14 2009-04-23 ペプリン リサーチ プロプライエタリー リミティッド Use of angeloyl-substituted ingenane in combination with other drugs to treat cancer
GB0525680D0 (en) 2005-12-16 2006-01-25 Peplin Ltd Therapeutic compositions
WO2008020335A2 (en) * 2006-06-09 2008-02-21 Novartis Ag Immunogenic compositions for streptococcus agalactiae
EP2094297A2 (en) * 2006-10-30 2009-09-02 Novartis AG Immunogenic and therapeutic compositions for streptococcus pyogenes
JP5518489B2 (en) 2007-02-09 2014-06-11 ブランシェット・ロックフェラー・ニューロサイエンスィズ・インスティテュート Therapeutic effects of bryostatin, bryolog, and other related substances in head injury-induced memory impairment and brain injury
CA2699513C (en) 2007-09-12 2018-03-13 Novartis Ag Gas57 mutant antigens and gas57 antibodies
KR100926408B1 (en) 2007-10-09 2009-11-12 고려대학교 산학협력단 Euphorbiae Kansui Radix reactivating gammaherpesvirus and pharmaceutical composition containing the same
PL2235046T3 (en) 2007-12-21 2012-12-31 Novartis Ag Mutant forms of streptolysin o
ES2753998T3 (en) * 2010-07-20 2020-04-15 Leo Laboratories Ltd A method for the production of ingenol-3-angelate
WO2012080466A2 (en) 2010-12-17 2012-06-21 Leo Pharma A/S Ingenols for treating seborrheic keratosis
WO2012085189A1 (en) 2010-12-22 2012-06-28 Leo Pharma A/S Ingenol-3-acylates i
CN103402969A (en) 2010-12-22 2013-11-20 利奥实验室有限公司 3-acyl-ingenols II
AU2011348637B2 (en) * 2010-12-22 2015-08-20 Leo Laboratories Limited Ingenol-3-acylates III and ingenol-3-carbamates
CN102977019A (en) * 2011-09-06 2013-03-20 高峰 New C19 diterpenoid alkaloid in Aconitum hemsleyanum var. janyuanum and applications thereof in preparation of anticancer drugs
JP2013063938A (en) * 2011-09-20 2013-04-11 Kao Corp Melanogenesis promoter
KR101406201B1 (en) * 2011-10-19 2014-06-12 한국생명공학연구원 Ingenane type diterpene compound and a pharmaceutical composition for treatment and prevention of virus infection comprising the same
EP2790694A1 (en) 2011-12-12 2014-10-22 Leo Laboratories Limited A topical composition comprising an ingenol derivative and a surfactant-cosolvent mixture
EP2790672A1 (en) 2011-12-12 2014-10-22 Leo Laboratories Limited Gel compositions
US20140348905A1 (en) * 2011-12-12 2014-11-27 Leo Laboratories Limited Gel compositions
US20130251782A1 (en) 2012-03-22 2013-09-26 Leo Laboratories Limited Topical application of ingenol mebutate with occlusion
US20150133543A1 (en) 2012-06-08 2015-05-14 Leo Laboratories Limited Topical gel composition comprising an ingenol derivative and a solvent mixture
ES2691077T3 (en) 2012-07-16 2018-11-23 Leo Laboratories Limited Process for the preparation of ingenol-3-angelato from 20-desoxi-ingenol
CN103086882B (en) * 2013-02-05 2015-04-15 中国科学院新疆理化技术研究所 Macrocyclic diterpene compounds in fruits of Euphorbia sororia, and preparation method and use thereof
US9526714B2 (en) 2014-03-24 2016-12-27 Leo Laboratories Limited Method for treating skin lesions with ingenol mebutate
EP3145903A4 (en) 2014-05-23 2018-01-03 Alphora Research Inc. A continuous flow process for the preparation of ingenol-3-mebutate
WO2016022358A1 (en) 2014-08-08 2016-02-11 The Regents Of The University Of California Compositions and methods for reactivating latent viral infections
CN104649901B (en) * 2015-02-09 2017-03-29 中国科学院昆明植物研究所 A kind of preparation method of the material medicine ingenol methyl butene acid esters for treating actinic keratosiss
KR101735785B1 (en) 2015-06-15 2017-05-15 대한민국(환경부 국립생물자원관장) Composition for Anti-Allergy Using an Extract of Trigonostemon reidioides
KR101735784B1 (en) 2015-06-15 2017-05-15 대한민국(환경부 국립생물자원관장) Composition for Anti-inflammation Using an Extract of Trigonostemon reidioides
CN105348095A (en) * 2015-12-07 2016-02-24 西宁意格知识产权咨询服务有限公司 Novel diterpenoid as well as preparation method and medical application thereof
US10275356B2 (en) * 2015-12-11 2019-04-30 Quanta Computer Inc. Component carrier with converter board
KR101755017B1 (en) 2016-08-10 2017-07-06 호서대학교 산학협력단 Method for preparation of extracts of Dudleya brittonii and composition for anti-cancer or anti-oxidation comprising the extracts of Dudleya brittonii as active ingredient
WO2018034318A1 (en) * 2016-08-18 2018-02-22 国立大学法人 奈良先端科学技術大学院大学 Immunomodulator
US10425485B2 (en) * 2016-11-21 2019-09-24 Cisco Technology, Inc. Integrating information centric networking (ICN) over low power and lossy networks (LLNs)
CN109912419A (en) * 2017-12-13 2019-06-21 复旦大学 Ingane type diterpene and its preparing the purposes in anti-hiv drug
KR101978915B1 (en) * 2018-08-08 2019-05-15 애경산업(주) The composition comprising extract of Euphorbia pulcherrima for improving acne
KR102508255B1 (en) * 2021-02-15 2023-03-09 연세대학교 산학협력단 Compositon for Controlling Sugar Containing Atypical PKC Activator

Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4418064A (en) * 1982-09-29 1983-11-29 The United States Of America As Represented By The Secretary Of Agriculture Chemotherapeutically active maytansinoids: treflorine, trenudine, and N-methyltrenudone
US4560774A (en) * 1982-11-17 1985-12-24 Arizona State University Macrocyclic lactones
US4716179A (en) * 1979-01-23 1987-12-29 Stiftung Deutches Krebsforschungszentrum Use of non-irritating or slightly irritating and/or promoting diterpene alcohol and of derivatives thereof as antineoplastic preparations
US5145842A (en) * 1986-06-11 1992-09-08 Alder Research Center Limited Partnership Protein kinase c. modulators. d.
US5643948A (en) * 1986-06-11 1997-07-01 Procyon Pharmaceuticals, Inc. Protein kinase C modulators. K.
US5716968A (en) * 1986-06-11 1998-02-10 Procyon Pharmaceuticals, Inc. Protein kinase C modulators. H.
US5750568A (en) * 1986-06-11 1998-05-12 Procyon Pharmaceuticals, Inc. Protein kinase C Modulators. L.
US5886019A (en) * 1986-06-11 1999-03-23 Procyon Pharmaceuticals, Inc. Protein kinase C modulators. F.
US5886017A (en) * 1986-06-11 1999-03-23 Procyon Pharmaceuticals, Inc. Protein kinase C modulators. E.
US5891906A (en) * 1986-06-11 1999-04-06 Procyon Pharmaceuticals, Inc. Polyacetate-derived phorboids having anti-inflammatory and other uses
US5891870A (en) * 1986-06-11 1999-04-06 Procyon Pharmaceuticals, Inc. Protein kinase C modulators Q
US5932613A (en) * 1996-07-03 1999-08-03 Millennium Pharmaceuticals, Inc. Anticancer agents
US5962498A (en) * 1986-06-11 1999-10-05 Procyon Pharmaceuticals, Inc. Protein kinase C modulators. C. indolactam structural-types with anti-inflammatory activity
US6268395B1 (en) * 1999-11-11 2001-07-31 Lead Chemical Co., Ltd. Phorbol derivatives having antivirus activity
US6432452B1 (en) * 1997-08-19 2002-08-13 Peplin Biotech Pty. Ltd. Anti-cancer compounds
US6593371B1 (en) * 1993-05-19 2003-07-15 Jeff J. Staggs Treatment for wart and related disorders
US20030166613A1 (en) * 2000-06-07 2003-09-04 Aylward James Harrison Therapeutic agents - I

Family Cites Families (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3809749A (en) 1971-03-05 1974-05-07 Amazon Natural Drug Co Topical pharmaceutical composition and method employing sap from the tree croton lechleri
IL82811A0 (en) 1986-06-11 1987-12-20 Alder Res Center Corp Anti-inflammatory compositions containing methanol derivatives and novel compounds contained therein
DE3805965A1 (en) 1988-02-25 1989-09-07 Tamas Geb Szenasi Eszter USE OF THE EUPHORBIA HIRTA L. PLANT AND ITS EXTRACTS AND ITS ACTIVE SUBSTANCES
DE4102054A1 (en) 1991-01-24 1992-07-30 Geb Szenasi Tamas Euphorbia hirta L. to increase immunity - e.g. against influenza, winter colds and AIDS and as antifungal agent to treat open wounds
US5317009A (en) * 1991-08-26 1994-05-31 New York University Anti-HIV proteins GAP 31, DAP 30 and DAP 32 and therapeutic uses thereof
HU208790B (en) 1991-12-05 1994-01-28 Tamas Process for producing pharmaceutical compositions against arc and aids, comprising euphorbia hirta l. plant or its parts or their extracts
CN1047084C (en) 1993-03-30 1999-12-08 刘振宪 Oral liquid for treatment of esophagus disease and its preparing method
CN1048164C (en) 1993-08-24 2000-01-12 吴雪姣 Oral liquid
JP2913451B2 (en) 1994-06-22 1999-06-28 株式会社イナックス Height adjustment structure of waterproof pan
JPH08245505A (en) 1994-09-09 1996-09-24 Tosoh Corp Diterpene derivative, its production and antitumor agent containing the same as active ingredient
JPH08176004A (en) 1994-12-21 1996-07-09 Lion Corp Living body-aging preventive and composition for skin
JPH08176002A (en) 1994-12-27 1996-07-09 Kao Corp Cell-anchoring inhibitor
US5874464A (en) * 1995-01-13 1999-02-23 The United States Of America As Represented By The Department Of Health And Human Services Conformationally constrained diacylglycerol analogues
CN1070372C (en) 1995-02-15 2001-09-05 赵国强 Powerful anticancer plaster and its preparing method
WO1997015575A1 (en) 1995-10-27 1997-05-01 Procyon Pharmaceuticals, Inc. Protein kinase c modulators. y.
CN1058620C (en) 1995-11-12 2000-11-22 卢颖 Yingfengaidi medicine and its production process
US5749111A (en) * 1996-02-14 1998-05-12 Teksource, Lc Gelatinous cushions with buckling columns
US6329459B1 (en) * 1996-09-23 2001-12-11 Bridgestone Corporation Extended syndiotactic polystyrene-elastomeric block copolymers
US6361501B1 (en) * 1997-08-26 2002-03-26 Seiko Epson Corporation Pulse wave diagnosing device
US6228939B1 (en) * 1999-05-19 2001-05-08 Bridgestone Corporation Thermoreversible gels comprising near gelation polymers
ATE287903T1 (en) * 1999-08-16 2005-02-15 Dow Global Technologies Inc HYDROGENATED BLOCK COPOLYMERS AND OPTICAL MEDIA PLATES MADE THEREFROM
AUPQ923100A0 (en) 2000-08-07 2000-08-31 Peplin Research Pty Ltd Treatment of prostate cancer
JP4880816B2 (en) 2000-12-15 2012-02-22 株式会社ヤクルト本社 Skin anti-aging agent
US6923993B2 (en) * 2001-12-12 2005-08-02 Nicholas J. Donato Process of isolating extract from the Euphorbia obesa plant and methods for using the same
JP3935360B2 (en) 2002-01-22 2007-06-20 丸善製薬株式会社 Hyaluronic acid production promoter, skin cosmetics and cosmetics
EP1676132B1 (en) * 2003-10-21 2014-01-22 Cedars-Sinai Medical Center Combination of chemotherapy and administration of glioma-antigen-pulsed dendritic cells in the treatment of glioma
PL1628673T3 (en) 2004-01-01 2007-07-31 Panacea Biotec Ltd Pharmaceutical compositions comprising an extract of euphorbia prostrata
US8106092B2 (en) 2004-12-13 2012-01-31 Leo Laboratories Limited Treatment of solid cancers
US7378455B2 (en) * 2005-06-30 2008-05-27 General Electric Company Molding composition and method, and molded article
BRPI0504797B1 (en) * 2005-10-27 2020-02-04 Pele Nova Biotecnologia S A topical formulation, cosmetic treatment method for skin rejuvenation, cosmetic treatment method and use of a formulation
JP2009516651A (en) 2005-11-14 2009-04-23 ペプリン リサーチ プロプライエタリー リミティッド Use of angeloyl-substituted ingenane in combination with other drugs to treat cancer
EP1965818A4 (en) 2005-11-25 2010-02-17 Peplin Research Pty Ltd Methods for wound healing
GB0525680D0 (en) 2005-12-16 2006-01-25 Peplin Ltd Therapeutic compositions
CN101742996B (en) 2007-04-30 2013-08-21 Leo实验室有限公司 Use of phorbol in preparing medicine for non-cancer skin lesions

Patent Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4716179A (en) * 1979-01-23 1987-12-29 Stiftung Deutches Krebsforschungszentrum Use of non-irritating or slightly irritating and/or promoting diterpene alcohol and of derivatives thereof as antineoplastic preparations
US4418064A (en) * 1982-09-29 1983-11-29 The United States Of America As Represented By The Secretary Of Agriculture Chemotherapeutically active maytansinoids: treflorine, trenudine, and N-methyltrenudone
US4560774A (en) * 1982-11-17 1985-12-24 Arizona State University Macrocyclic lactones
US5891870A (en) * 1986-06-11 1999-04-06 Procyon Pharmaceuticals, Inc. Protein kinase C modulators Q
US5962498A (en) * 1986-06-11 1999-10-05 Procyon Pharmaceuticals, Inc. Protein kinase C modulators. C. indolactam structural-types with anti-inflammatory activity
US5716968A (en) * 1986-06-11 1998-02-10 Procyon Pharmaceuticals, Inc. Protein kinase C modulators. H.
US5750568A (en) * 1986-06-11 1998-05-12 Procyon Pharmaceuticals, Inc. Protein kinase C Modulators. L.
US5886019A (en) * 1986-06-11 1999-03-23 Procyon Pharmaceuticals, Inc. Protein kinase C modulators. F.
US5886017A (en) * 1986-06-11 1999-03-23 Procyon Pharmaceuticals, Inc. Protein kinase C modulators. E.
US5891906A (en) * 1986-06-11 1999-04-06 Procyon Pharmaceuticals, Inc. Polyacetate-derived phorboids having anti-inflammatory and other uses
US5145842A (en) * 1986-06-11 1992-09-08 Alder Research Center Limited Partnership Protein kinase c. modulators. d.
US5643948A (en) * 1986-06-11 1997-07-01 Procyon Pharmaceuticals, Inc. Protein kinase C modulators. K.
US6593371B1 (en) * 1993-05-19 2003-07-15 Jeff J. Staggs Treatment for wart and related disorders
US5932613A (en) * 1996-07-03 1999-08-03 Millennium Pharmaceuticals, Inc. Anticancer agents
US6432452B1 (en) * 1997-08-19 2002-08-13 Peplin Biotech Pty. Ltd. Anti-cancer compounds
US6787161B2 (en) * 1997-08-19 2004-09-07 Peplin Biotech Pty. Ltd. Anti-cancer compounds
US20050003031A1 (en) * 1997-08-19 2005-01-06 Aylward James Harrison Anti-cancer compounds
US6844013B2 (en) * 1997-08-19 2005-01-18 Peplin Research Pty Ltd Methods of stimulating the immune system
US6268395B1 (en) * 1999-11-11 2001-07-31 Lead Chemical Co., Ltd. Phorbol derivatives having antivirus activity
US20030166613A1 (en) * 2000-06-07 2003-09-04 Aylward James Harrison Therapeutic agents - I
US20030171337A1 (en) * 2000-06-07 2003-09-11 Aylward James Harrison Therapeutic agents - II
US20050209192A1 (en) * 2000-06-07 2005-09-22 Peplin Research Pty. Ltd. Therapeutic agents - II

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7410656B2 (en) 1997-08-19 2008-08-12 Peplin Research Pty. Ltd. Anti-cancer compounds
US20030166613A1 (en) * 2000-06-07 2003-09-04 Aylward James Harrison Therapeutic agents - I
US20030171337A1 (en) * 2000-06-07 2003-09-11 Aylward James Harrison Therapeutic agents - II
US9314458B2 (en) 2000-06-07 2016-04-19 Leo Laboratories Limited Topical use of ingenol-3-angelate or a salt thereof to treat skin cancer
US20050209192A1 (en) * 2000-06-07 2005-09-22 Peplin Research Pty. Ltd. Therapeutic agents - II
US20110213002A1 (en) * 2000-06-07 2011-09-01 Peplin Research Pty Ltd. Macrocyclic diterpenes for treating conditions associated with protein kinase c
US7838555B2 (en) 2000-06-07 2010-11-23 Peplin Research Pty Ltd Macrocyclic diterpenes for the treatment and prophylaxis of acne vulgaris
US7449492B2 (en) 2000-06-07 2008-11-11 Peplin Research Pty, Ltd. Therapeutic agents—I
US7378445B2 (en) 2000-08-07 2008-05-27 Peplin Research Pty. Ltd. Treatment of prostate cancer
US20100222376A1 (en) * 2003-09-26 2010-09-02 Yale University Chelerythrine, analogs thereof and their use in the treatment of bipolar disorder and other cognitive disorders
US20050070565A1 (en) * 2003-09-26 2005-03-31 Arnsten Amy F.T. Chelerythrine, analogs thereof and their use in the treatment of bipolar disorder and other cognitive disorders
EP1838330A4 (en) * 2004-12-13 2010-07-07 Peplin Research Pty Ltd Treatment of solid cancers
US20080187513A1 (en) * 2004-12-13 2008-08-07 Peplin Research Pty Ltd. Treatment of Solid Cancers
EP1838330A1 (en) * 2004-12-13 2007-10-03 Peplin Research Pty Ltd. Treatment of solid cancers
US8106092B2 (en) 2004-12-13 2012-01-31 Leo Laboratories Limited Treatment of solid cancers
US20090076132A1 (en) * 2005-03-11 2009-03-19 Gary Pekoe Antiviral compositions and methods of treatment
US20090215884A1 (en) * 2005-11-25 2009-08-27 Peplin Research Pty Ltd. Method for wound healing
US9895334B2 (en) 2007-04-30 2018-02-20 Leo Laboratories Limited Treatment of virally induced lesions
KR101089314B1 (en) 2010-06-07 2011-12-02 한국식품연구원 Composition containing euphobiasteroid for prevention, treatment or improvement of obesity

Also Published As

Publication number Publication date
US7449492B2 (en) 2008-11-11
CA2411726A1 (en) 2001-12-13
US20030171337A1 (en) 2003-09-11
JP5822418B2 (en) 2015-11-24
US9314458B2 (en) 2016-04-19
US20100048698A1 (en) 2010-02-25
EP1296698A1 (en) 2003-04-02
JP2003535137A (en) 2003-11-25
EP1296697A4 (en) 2005-02-16
EP1296699A4 (en) 2005-02-16
EP1296699A1 (en) 2003-04-02
US20050209192A1 (en) 2005-09-22
AUPQ801700A0 (en) 2000-06-29
EP1296698A4 (en) 2005-02-16
JP2003535138A (en) 2003-11-25
CA2411642C (en) 2011-05-17
JP2003535136A (en) 2003-11-25
EP1296698B1 (en) 2015-01-14
JP5339662B2 (en) 2013-11-13
EP1296699B1 (en) 2015-08-12
JP5822417B2 (en) 2015-11-24
NZ522425A (en) 2004-05-28
CA2411596A1 (en) 2001-12-13
EP1296697A1 (en) 2003-04-02
US20030166613A1 (en) 2003-09-04
BR0111458A (en) 2003-06-24
CA2411642A1 (en) 2001-12-13
BR0111423A (en) 2003-04-29
US20110213002A1 (en) 2011-09-01
CA2411596C (en) 2014-08-19
WO2001093883A1 (en) 2001-12-13
NZ522427A (en) 2004-05-28
BR0111375A (en) 2003-06-10
NZ522426A (en) 2004-05-28
US7838555B2 (en) 2010-11-23
CA2411726C (en) 2011-09-20
WO2001093884A1 (en) 2001-12-13
WO2001093885A1 (en) 2001-12-13

Similar Documents

Publication Publication Date Title
CA2411726C (en) Use of ingenanes in treating a pkc-related condition or disorder
US7378445B2 (en) Treatment of prostate cancer
Thomé et al. Chloroquine: modes of action of an undervalued drug
AU748542B2 (en) Therapeutic agents - III
RU2741801C2 (en) Pharmaceutical composition from galleria mellonella larvae and a method for production thereof
US20020031509A1 (en) Pharmaceutical composition and method for its manufacture
AU752462B2 (en) Therapeutic agents - I
AU752435B2 (en) Therapeutic agents - II
Viana EFFECT OF CASEARIA SYLVESTRIS OIL IN CHRONIC INFLAMMATION INDUCED BY SYNTHETIC IMPLANTS IN MICE
AU754778B2 (en) Treatment of prostate cancer
Ameen et al. Effect of extract of Azadirchta indica and octreotide on induced Acute Pancreatitis in Rats

Legal Events

Date Code Title Description
AS Assignment

Owner name: PEPLIN RESEARCH PTY LTD, AUSTRALIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:AYLWARD, JAMES HARRISON;PARSONS, PETER GORDON;SUHRBIER, ANDREAS;AND OTHERS;REEL/FRAME:014014/0305;SIGNING DATES FROM 20030402 TO 20030409

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION