WO2008154705A1 - Transdermal delivery of active agents - Google Patents

Transdermal delivery of active agents Download PDF

Info

Publication number
WO2008154705A1
WO2008154705A1 PCT/AU2008/000908 AU2008000908W WO2008154705A1 WO 2008154705 A1 WO2008154705 A1 WO 2008154705A1 AU 2008000908 W AU2008000908 W AU 2008000908W WO 2008154705 A1 WO2008154705 A1 WO 2008154705A1
Authority
WO
WIPO (PCT)
Prior art keywords
agents
microemulsion
treatment
skin
topical
Prior art date
Application number
PCT/AU2008/000908
Other languages
French (fr)
Inventor
Michael R Luke
Stewart R Himes
Original Assignee
Apollo Life Sciences Limited
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
Priority claimed from AU2007903316A external-priority patent/AU2007903316A0/en
Application filed by Apollo Life Sciences Limited filed Critical Apollo Life Sciences Limited
Publication of WO2008154705A1 publication Critical patent/WO2008154705A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • 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

Definitions

  • the present invention generally relates to a vehicle useful for delivering a pharmaceutically active compound transdermally to a subject. More particularly, the present invention provides microemulsions for transdermal delivery of active agents to a subject.
  • transdermal delivery is a highly desired method by which therapeutic amounts of peptides, proteins or other pharmaceutical agents could be administered particularly for local and/or systemic therapy.
  • biologically-active macromolecules such as certain peptides or proteins, generally have low oral bioavailability, making oral administration difficult, and often have short biological half-lives, making parenteral delivery impractical outside a hospital setting (Shahrokh et al. American Chemical Society, 1997).
  • there are many medical conditions and diseases of the skin which would greatly benefit from direct local therapy, without exposing the other organs of the body to the deleterious side-effects of delivery via other routes.
  • NSAIDs non-steroidal anti-inflammatory drugs
  • an emulsion is a composition comprising more than one phase where at least one of the phases consists of a finely divided phase domain (such as, for example, particles or droplets) distributed throughout a continuous phase domain.
  • the finely divided domains are generally referred to as dispersed or discontinuous phase domains.
  • a microemulsion generally is a system comprising an oil phase, a water phase and a surfactant. Some microemulsions are quaternary systems and further comprise a co- surfactant or a co-solvent.
  • Microemulsions differ from ordinary emulsions (or macroemulsions) in that they are thermodynamically stable and form spontaneously upon mixing.
  • the average drop size of ordinary emulsions grows continuously with time so that phase separation ultimately occurs under gravitational force, i.e. they are thermodynamically unstable and their formation requires input of work.
  • the drops of the dispersed phase are generally large (> 0.2 ⁇ m) so that ordinary emulsions often take on a milky opaque appearance.
  • microemulsions have an observable transparency, which is due to the fact that the maximum size of the droplets of the dispersed phase is not larger than one-half of the wavelength of visible light.
  • the droplet diameter in stable microemulsions is usually within the range of 5-200 nm.
  • Microemulsions also possess specific physicochemical properties such as optical isotropy, low viscosity and thermodynamic stability.
  • This microemulsion system was loaded with 2.5% lidocaine (a highly lipophilic molecule of low molecular weight) as base or as a hydrochloride salt.
  • Labrasol a mixture consisting of 30% mono-, di and triglycerides of C and C fatty acids, 50% of the ap- mono- and di-esters of poly(ethylene glycol) (PEG 400) and 20% of free PEG 400
  • Plurol Isostearique isostearic acid ester of polyglycerol, containing 30-35% of diglycerol, 20-25% of triglycerol, 15-20% tetraglycerol, and 10% of pentaglycerol and higher oligomers
  • isostearylic isostearate (92% purity)
  • microemulsions were applied to excised rat skin in a Franz-type diffusion cell and were found to increase transdermal flux of lidocaine through the excised rat skin by up to four times compared to a conventional oil-in- water emulsion, and that of prilocaine hydrochloride by almost 10 times compared to a hydrogel.
  • Li et al. (International Journal of Pharmaceutics 237:77-85, 2002) examined the use of a microemulsion for intranasal delivery of the hydrophobic drug diazepam.
  • Diazepam a practically water-insoluble drug, displayed a high solubility of 41 mg/ml in a microemulsion consisting of 15% ethyl laurate, 15% H 2 O, and 70% (w/w) surfactant/co- surfactant (Tween 80:propylene glycolethanol at 1 :1 :1 weight ratio).
  • the study demonstrated an intranasal bioavailability of 50%.
  • Escribano et al. (European Journal of Pharmaceutical Sciences 7P/203-210, 2003) studied the use of microemulsion formed with 19% Transcutanol, 19.5% Plurol oleique, 30.6% water, 10.9% Isostearol isostearate, Labrasol 19% for transdermal delivery of the low molecular weight chemical diclofenac across human skin.
  • the microemulsion was found to be less efficient than a preparation made with the absorption enhancers oleic acid and d- limonene.
  • microemulsions have not been demonstrated to carry macromolecules, such as certain peptides or proteins, water soluble vitamins, NSAIDs and chemical compounds across the dermis.
  • macromolecules such as certain peptides or proteins, water soluble vitamins, NSAIDs and chemical compounds
  • the present invention provides a topical delivery system which assists in the penetration of a desired active agent, such as peptides, polypeptides and proteins, water soluble vitamins, NSAIDs, genetic molecules and chemical compounds into the skin, which does not necessitate the use of chemical enhancers, nor electrical or ultrasonic energy to facilitate penetration into and though the skin. Furthermore, the delivery system is easy to apply to small or large areas of the skin.
  • a desired active agent such as peptides, polypeptides and proteins, water soluble vitamins, NSAIDs, genetic molecules and chemical compounds into the skin, which does not necessitate the use of chemical enhancers, nor electrical or ultrasonic energy to facilitate penetration into and though the skin.
  • the delivery system is easy to apply to small or large areas of the skin.
  • the present invention further enables the use of hydrophilic molecules which are incorporated within the submicron sized water droplets contained within the microemulsion, thereby enhancing transdermal delivery of desired active agents.
  • the topical delivery system of the present invention is in the form of a microemulsion.
  • the subject microemulsions for transdermal delivery of peptides, polypeptides and proteins or other molecules such as water soluble vitamins, NSAIDs, genetic molecules and chemical compounds are able to be formed with little or no shear force applied to the molecules during the preparation of the microemulsion, which aids in the preservation of the structure and bioactivity of these molecules.
  • the microemulsion also provides both a hydrophilic and hydrophobic environment, which can aid in the solubilization of various molecules and which additionally provides the ability to mix hydrophobic molecules and hydrophilic molecules within the same formulation.
  • the microemulsions of the present invention provide an environment which allows for the solubilization of various water- soluble molecules at a wide range of concentrations.
  • the present invention provides a microemulsion comprising a water phase; an oil phase; an active ingredient; one or more zwitterionic surfactants; and one or more co-surfactants.
  • the present invention provides a microemulsion which is capable of transdermal delivery of active agents.
  • the present invention further extends to a pharmaceutical composition comprising the microemulsion and an active agent wherein the pharmaceutical composition is suitable for topical application on a subject.
  • the present invention further provides a method suitable for the transdermal delivery of active agents in a range of therapeutic, cosmetic and research applications.
  • the method comprises (a) preparing a water phase, including dissolving one or more selected active agents in water; (b) preparing an oil phase; (c) emulsifying the water phase and the oil phase using one or more zwitterionic surfactants and one or more co-surfactants, thereby forming a topical pharmaceutical composition; and (d) applying the topical pharmaceutical composition on a subject.
  • the microemulsion of the present invention is useful inter alia for the topical delivery of anti-inflammatory agents, such as small molecules, proteins, antibodies, Fc fusion molecules and soluble receptors; vaccines; imaging agents; anti-cancer agents, particularly anti-melanoma agents; skin lightening including whitening agents; UV blocking agents, agents to reduce or nullify nitric oxide, agents useful for the treatment of erectile dysfunction; agents for increasing local circulation, agents for the amelioration of facial and skin ageing, agents for the treatment of obesity, re-pigmentation agents; genetic molecules such as RNAi, DNA, virosomes and anti-sense RNA; and agents useful in treating muscle dystrophy.
  • the microemulsions have application in human and veterinary medicine as well as for agricultural animals.
  • a list of agents contemplated for use in the microemulsions includes but is not limited to the list provided in Table 1. TABLE 1
  • PDE5 inhibitors phosphodiesterases type 5 inhibitors; sildenafil, tadalafil, vardenafil
  • a pharmaceutically active agent includes a single pharmaceutically active agent as well as two or more pharmaceutically active agents
  • an agent includes a single agent as well as two or more agents
  • the microemulsion includes one or more microemulsions
  • compound used interchangeably herein to refer to a chemical compound and in particular a protein or a chimeric molecule thereof that induces a desired pharmacological and/or physiological effect.
  • the terms also encompass pharmaceutically acceptable and pharmacologically active ingredients of those active agents specifically mentioned herein including but not limited to salts, esters, amides, prodrugs, active metabolites, analogs and the like.
  • the desired pharmacological effect is dependent on the agent used but includes an antiinflammatory effect, an immune response, a change in the level of gene expression, a pigmentation effect including a lightening effect, an imaging effect on a target such as a target lymph node, an anti-muscle dystrophic effect, weight loss, treatment of cancer, reduction in pain or a physiological effect such as correction of erectile dysfunction.
  • Reference to a "compound”, “agent”, “active agent”, “chemical agent”, “pharmaceutical agent”, “pharmacologically active agent”, “medicament”, “active” and “drug” includes combinations of two or more active agents such as two or more proteins.
  • a “combination” also includes multi-part such as a two-part composition where the agents are provided separately and given or dispensed separately or admixed together prior to dispensation.
  • agents include peptides, polypeptides and proteins, large and small chemical molecules, hormones, steroids and nucleic acid molecules as well as viral components such as virosomes.
  • an agent as used herein mean a sufficient amount of the pharmaceutical agent, alone or in combination with other agents to provide the desired therapeutic or physiological effect or outcome. Undesirable effects, e.g. side effects, are sometimes manifested along with the desired therapeutic effect; hence, a practitioner balances the potential benefits against the potential risks in determining what is an appropriate "effective amount”.
  • the exact amount required will vary from subject to subject, depending on the species, age and general condition of the subject, mode of administration and the like. Thus, it may not be possible to specify an exact "effective amount”. However, an appropriate "effective amount” in any individual case may be determined by one of ordinary skill in the art using only routine experimentation.
  • an "effective amount”, for example, would include an amount of a cytokine or anti- cytokine antibody or anti-sense or RNAi molecule directed to a gene or mRNA transcript encoding an inflammatory cytokine to inhibit an inflammatory response by a reduction in aberrant skin proliferation, antibody production, cytokine production and/or immune response effector molecule production.
  • pharmaceutically acceptable carrier excipient or diluent
  • a pharmaceutical vehicle comprised of a material that is not biologically or otherwise undesirable, i.e. the material may be administered to a subject along with the selected active agent without causing any or a substantial adverse reaction.
  • Carriers may include excipients and other additives such as diluents, detergents, coloring agents, wetting or emulsifying agents, pH buffering agents, preservatives, and the like.
  • a "pharmacologically acceptable" salt, ester, amide, prodrug or derivative of a compound as provided herein is a salt, ester, amide, prodrug or derivative that this not biologically or otherwise undesirable.
  • treating and “treatment” as used herein refer to reduction in severity and/or frequency of symptoms of the condition being treated, elimination of symptoms and/or the underlying cause, prevention of the occurrence of symptoms of the condition and/or their underlying cause and improvement or remediation or amelioration of damage following a condition.
  • Treating" a subject may involve prevention of a condition or other adverse physiological event in a susceptible individual as well as treatment of a clinically symptomatic individual by ameliorating the symptoms of the condition.
  • a "subject” as used herein refers to an animal, in a particular embodiment, a mammal and in a further embodiment human who can benefit from the pharmaceutical formulations and methods of the present invention. There is no limitation on the type of animal that could benefit from the presently described pharmaceutical formulations and methods. A subject regardless of whether a human or non-human animal may be referred to as an individual, patient, animal, host or recipient.
  • the compounds and methods of the present invention have applications in human medicine, veterinary medicine as well as in general, domestic or wild animal husbandry including agriculturally-relevant animals (e.g. livestock animals).
  • the animals are humans or other primates such as orangutans, gorillas or marmosets, further contemplated are livestock animals, laboratory test animals, companion animals or captive wild animals, as well as avian species.
  • laboratory test animals include mice, rats, rabbits, guinea pigs and hamsters. Rabbits and rodent animals, such as rats and mice, provide a convenient test system or animal model. Livestock animals include sheep, cows, pigs, goats, horses, yaks, llamas, camels and donkeys. Companion animals include dogs and cats. Non-mammalian animals such as avian species, fish and amphibians.
  • protein is used interchangeably with the term “peptide” or “polypeptide” and is used in its most general sense including ligands and receptors.
  • reference to a particular protein should be understood to refer to the "complete” protein as well as fragments, isoforms, derivatives or homologs or chimeras thereof comprising one or more amino acid additions, deletions or substitutions, but which substantially retain the biological activity of the complete protein.
  • polypeptide refers to a polymer of amino acids and its equivalent but does not imply a limitation as to a specific length of the product, thus, peptides, oligopeptides, polypeptides and proteins are included within the definition of a "polypeptide”. This term also includes all co- or post-translationally modified forms of a polypeptide. Also included within the definition are, for example, polypeptides containing one or more analogs of an amino acid including, unnatural amino acids or polypeptides with substituted linkages.
  • co- or post-translational modifications refer to covalent modifications occurred during or after translation of the peptide chain.
  • exemplary co- or post-translational modifications include, but are not limited, to acylation (including acetylation), amidation or deamidation, biotinylation, carbamylation (or carbamoylation), carboxylation or decarboxylation, disulfide bond formation, fatty acid acylation (including myristoylation, palmitoylation and stearoylation), formylation, glycation, glycosylation, hydroxylation, incorporation of selenocysteine, lipidation, lipoic acid addition, methylation, N- or C-terminal blocking, N- or C-terminal removal, nitration, oxidation of methionine, phosphorylation, proteolytic cleavage, prenylation (including farnesylation, geranyl geranylation), pyridoxal phosphate addition, sialy
  • a “derivative" of a polypeptide of the present invention also encompasses a portion or a part of a full-length parent polypeptide, which retains partial transcriptional activity of the parent polypeptide and includes a variant.
  • Such "biologically-active fragments” include deletion mutants and small peptides, for example, for at least 10, in a particular embodiment, at least 20 and in a further embodiment at least 30 contiguous amino acids, which exhibit the requisite activity.
  • Peptides of this type may be obtained through the application of standard recombinant nucleic acid techniques or synthesized using conventional liquid or solid phase synthesis techniques.
  • peptides can be produced by digestion of an amino acid sequence of the invention with proteinases such as endoLys-C, endoArg-C, endoGlu-C and staphylococcus V8-protease.
  • the digested fragments can be purified by, for example, high performance liquid chromatographic (HPLC) techniques. Any such fragment, irrespective of its means of generation, is to be understood as being encompassed by the term "derivative" as used herein.
  • the active agent of the present invention may be a chimeric molecule comprising an isolated protein or a fragment thereof, such as an extra-cellular domain of a membrane bound protein, linked to the constant (Fc) or framework region of a human immunoglobulin via one or more protein linkers.
  • a chimeric molecule is also referred to herein as protein-Fc.
  • Other chimeric molecules contemplated by the present invention include the protein or protein-Fc or a fragment thereof, linked to a lipid moiety such as a polyunsaturated fatty acid molecule.
  • Such lipid moieties may be linked to an amino acid residue in the backbone of the molecule or to a side chain of such an amino acid residue.
  • the human immunoglobulin may be selected from IgGl, IgG2, IgG3, IgG4, IgAl, IgA2, IgM, IgE 5 IgD.
  • the active agent of the present invention may further include a chimeric molecule comprising an isolated protein or a fragment thereof, such as an extra-cellular domain of a membrane bound protein, linked to the constant (Fc) or framework region of a mammalian immunoglobulin via one or more protein linker.
  • the mammal Fc or framework region of the immunoglobulin is derived from a mammal selected from the group consisting of primates, including humans, marmosets, orangutans and gorillas, livestock animals (e.g.
  • the Fc or framework region is a human immunoglobulin.
  • the mammal is a human.
  • Such a chimeric molecule is also referred to herein as protein-Fc.
  • Other chimeric molecules contemplated by the present invention include the protein or protein-Fc or a fragment thereof linked to a lipid moiety such as a polyunsaturated fatty acid molecule. Such lipid moieties may be linked to an amino acid residue in the background of the molecule or to a side chain of such an amino acid residue.
  • the active agent has one or more of the following activities: anti -infective, antiinflammatory, anti-tumor, skin repair, skin damage prevention, skin anti-aging, skin lightening or whitening, hair growth promotion or hair growth inhibition, muscle repair, weight loss promoting, erectile dysfunction correcting, anti-acne, induction of immune response (antibody response and/or a cytotoxic T-cell and/or a T helper response) and CNS healing activities.
  • the active agent used in the compositions and methods of the present invention may be of any size.
  • the active agents used in the present invention comprise molecules which are greated than 500Da in size.
  • molecules contained in the active ingredients are between 500Da and 15OkDa, such as, for example, 500, 600, 700, 800, 900,
  • the molecules contained within the active ingredients are between 500 and 1000Da, such as, for example 500, 510 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990, 1000Da.
  • microemulsions of the present invention are useful inter alia for delivering small molecules, proteins (including peptides and polypeptides and chimeras), antibodies, other water soluble molecules as well as oil-soluble molecules useful in treating skin-related inflammatory conditions.
  • the microemulsions can be used to deliver nitric oxide and TNF alpha inhibitors.
  • Vitamin B 12 for example, is a useful nitric oxide chelator and is useful in reducing skin-related inflammatory responses.
  • the present invention contemplates a topical composition comprising a vitamin (VB 12) derivative (or analogs thereof) for the treatment of skin-related inflammatory conditions.
  • any inhibitor of i-nitric oxide synthase, e-nitric oxide synthase or n-nitric oxide synthase is contemplated by the present invention. Nicotinamide may also be used to block nitric oxide synthase.
  • Other agents useful for treating psoriasis, dermatitis and itch include curcuminoids, COX-I and COX-2 inhibitors, vitamins such as vitamin E and vitamin C.
  • the present invention contemplates a topical composition comprising a curcuminoid derivative (or an analog thereof) for the treatment of inflammatory conditions of the skin.
  • Proteins useful for treating skin-related inflammatory conditions such as psoriasis, dermatitis, systemic lupus erythematosus (SLE) and itch include antibodies directed against IL-20, C5aR, IFN alpha, TNF (e.g. infliximab, adalimumab), IL-12, IL-I, IL-6, leukotrienes and IL-4.
  • TNF e.g. infliximab, adalimumab
  • IL-12 e.g. infliximab, adalimumab
  • IL-12 e.g. infliximab, adalimumab
  • IL-12 e.g. infliximab, adalimumab
  • IL-12 e.g. infliximab, adalimumab
  • IL-12 e.g. infliximab, a
  • the vitamin B 12 derivative is adenosylcobalamin and the curcuminoid is tetrahydrocurcumin.
  • an anti-TNF molecule is administered within a microemulsion containing both adenosylcobalamin and tetrahydrocurcumin.
  • microemulsions of the present invention are useful inter alia for delivering small molecules, proteins (including peptides and polypeptides and chimeras), antibodies, other water soluble molecules as well as oil-soluble molecules useful in treating bruising.
  • This approach avoids the toxicity common with systemic or oral delivery of, for example, anti-ecchymosis therapies such as extracts of Arnica montana for the treatment of bruising.
  • the microemulsions can be used to deliver nitric oxide and TNF alpha inhibitors to a site of ecchymosis following cosmetic surgery such as rhytidectomy.
  • Vitamin B 12 for example, is a useful nitric oxide chelator and is useful in reducing bruising, also known as ecchymosis.
  • the present invention contemplates a topical composition comprising a vitamin B 12 derivative (or analogs thereof) for the treatment of bruising.
  • the present invention contemplates a topical composition comprising a vitamin B12 derivative (or an analog thereof) and a curcuminoid derivative (or an analog thereof) for the treatment of bruising.
  • the vitamin B 12 derivative is adenosylcobalamin and the curcuminoid is tetrahydrocurcumin.
  • the microemulsions of the present invention are useful inter alia for delivering small molecules, proteins (including peptides and polypeptides and chimeras), antibodies, other water soluble molecules as well as oil-soluble molecules useful in treating musclar or tendon damage. Conditions such as delayed onset muscle soreness (DOMS) can prevent athletes from recovering following a training event or game, resulting in measurable reductions in power, flexibility or endurance.
  • Vitamin B 12 for example, is a useful nitric oxide chelator and is useful in reducing DOMS.
  • the microemulsions of the present invention can be used to deliver nitric oxide and TNF alpha inhibitors to muscular tissue to ameliorate conditions such as DOMS.
  • the present invention contemplates a topical composition comprising a vitamin B 12 derivative (or analogs thereof) for the treatment of muscular damage such as DOMS. Additionally such treatment also be used for tendon injuries such as tenosynovitis,
  • the present invention contemplates a topical composition comprising a vitamin B12 derivative (or an analog thereof) and a curcumioid derivative (or an analog thereof) for the treatment of muscular damage such as DOMS.
  • the vitamin B 12 derivative is adenosylcobalamin and the curcuminoid is tetrahydrocurcumin. 4. Anti-inflammatory Activities for the Treatment of Joint Damage.
  • microemulsions of the present invention are useful inter alia for delivering small molecules, proteins (including peptides and polypeptides and chimeras), antibodies, other water soluble molecules as well as oil-soluble molecules useful in treating joint damage.
  • Joint damage may arise through chronic inflammatory disease states such as rheumatoid arthritis and systemic lupus erythematosus (SLE) or in response to acute damage such as that experienced by individuals during sporting activities (e.g. ankle sprain).
  • Vitamin B 12 for example, is a useful nitric oxide chelator and is useful in reducing joint-related inflammation.
  • microemulsions of the present invention can be used to deliver nitric oxide and TNF alpha inhibitors to joint tissue to ameliorate conditions such as rheumatoid arthritis and joint damage following acute injury.
  • This approach avoids the toxicity common with systemic or oral delivery of other anti-inflammatory therapies such as COX inhibitors.
  • the present invention contemplates a topical composition comprising a vitamin B 12 derivative (or analogs thereof) for the treatment of rheumatoid arthritis, SLE, or acute joint damage.
  • the present invention contemplates a topical composition comprising a vitamin B 12 derivative (or an analog thereof) and a curcuminoid derivative (or an analog thereof) for the treatment of rheumatoid arthritis, SLE, or acute joint damage,
  • the vitamin B 12 derivative is adenosylcobalamin and the curcuminoid is tetrahydrocurcumin.
  • the microemulsions are useful as topical vaccines such as to induce an antibody response and/or a cytotoxic T-cell and/or a T helper response including a TH1/TH2 response.
  • Topical vaccines are particularly useful for mobilizing dendritic cells in the dermal layer and increasing numbers of activated dendritic cells.
  • hyperdermic needles were able to deliver agents to the subcutaneous layer or muscle layer, however, the procedure was invasive and did not necessarily target the desired region.
  • the topical microemulsion may be provided with or without an adjuvant.
  • Adjuvants contemplated herein include microorganisms or viruses or components thereof and chemical substances which enhance the immune response and/or chemical which act as preservatives and tissue fixatives.
  • Particularly useful adjuvants include aluminium hydroxide, aluminium phosphate and calcium phosphate.
  • oil based emulsions and products from bacteria including synthetic derivatives and liposomes of gram-negative bacteria, endotoxins, cholesterol, fatty acids, aliphatic amines, parafmic and vegetable oils, monophosphoryl lipid A, iscoms with Quil-A and Syntex agents formulations containing threonyl derivaties or muramyl type peptides may be employed.
  • adjuvants contemplated by the present invention include Freund's emulsify oil adjuvants (complete and incomplete), ARLACEL A, mineral oil, emulsified peanut oil adjuvant (adjuvant 65), mineral compounds and bacterial products from Bordetella pertussis, Corynebacterium granulosom-de ⁇ vQd P40 component, lipopolysaccaride, Mycobacterium or components thereof, inulin, cholera toxin and/or liposomes may also be employed.
  • adjuvants contemplated by the present invention include Freund's emulsify oil adjuvants (complete and incomplete), ARLACEL A, mineral oil, emulsified peanut oil adjuvant (adjuvant 65), mineral compounds and bacterial products from Bordetella pertussis, Corynebacterium granulosom-de ⁇ vQd P40 component, lipopolysaccaride, Mycobacterium or components thereof, inulin, cholera toxin
  • adjuvants contemplated by the present invention include proteinacous pathogen recognition receptors or related chimeric molecules such as soluble C-type lectins, for example, CD209L, CD209L- Fc, DC-SIGN, DC-SIGN-Fc, CLEC4K, CD 207. Binding of pathogen by C-type lectins induces internalisation and degradation in lysosomes, which facilitates antigen processing and presentation and the initiation of an immune response.
  • proteinacous pathogen recognition receptors or related chimeric molecules such as soluble C-type lectins, for example, CD209L, CD209L- Fc, DC-SIGN, DC-SIGN-Fc, CLEC4K, CD 207. Binding of pathogen by C-type lectins induces internalisation and degradation in lysosomes, which facilitates antigen processing and presentation and the initiation of an immune response.
  • HBV Hepatitis B virus
  • HCV Hepatitis C virus
  • HPV Human paploma virus
  • shigella shigella
  • malaria rabies
  • measles mumps
  • rubella whooping cough
  • herpes zoster a topical composition comprising a antigen and an adjuvant within a microemulsion for the vaccination of a vertebrate host.
  • the microemulsions of the present invention may be used to induce weight loss via lymphodystrophy. It is proposed that topically applied insulin induces insulin- mediated lymphodystrophy. There is minimal if any entry of the insulin to the blood system and hence no increase in glucose levels.
  • Reference to "insulin” includes insulin analogs and precursor forms of insulin which may have the ability to interact with insulin receptors but lack signalling capability.
  • the present invention contemplates a topical composition comprising insulin for the amelioration of sub-cutaneuous fat deposits.
  • a topical composition comprising growth factor administered within a microemulsion for the amelioration of obesity.
  • a topical composition comprising a growth hormone releasing hormone, or growth hormone releasing peptide for the amelioration of obesity.
  • agents which are useful in sub-cutaneuous fat deposits include Coleus Forskohlii Root Extract and Bodyfit.
  • the microemulsions are able to access or introduce imaging reporter molecules to the sentinel lymph node in relation to particular cancers such as melanoma and breast carcinoma.
  • the sentinel lymph node is the first lymph node to which cancer is likely to spread from the primary tumor. Cancer cells may appear first in the sentinel node before spreading to other lymph nodes and other places in the body.
  • imaging prior to the removal of the sentinel lymph node increases the likelihood that the correct lymph node is removed in its entirety.
  • Any imaging agent may be employed, such as a radio-active agents, chelates, fluorescent compounds and metals.
  • the identification of the sentinel lymph node is improved and can be successfully removed thus reducing the risk of spread of a cancer.
  • a radionuclide is bound to the targeting molecule indirectly, through a bifunctional chelating agent (BFCA)
  • radioactive agents which may be admixed with the microemulsions include the following: Antimony-124, Antimony-125, Arsenic-74, Barium-103, Barium- 140, Beryllium-7, Bismuth-206, Bismuth-207, Cadmium- 109, Cadmium- HSm, Calcium-45, Cerium-139, Cerium-141, Cerium-144, Cesium-137, Chromium-51, Cobalt-56, Cobalt-57, Cobalt-58, Cobalt-60, Cobalt-64, Erbium-169,
  • Europium-152 Gadolinium-153, Gold-195, Gold-199, Hafnium-175, HafniumO175- 181, Indium-I l l, Iridium-192, Iron-55, Iron-59, Krypton-85, Lead-210, Manganese- 54, Mercury- 197, Mercury-203, Molybdenum-99, Neodymium-147, Neptunium- 237, Nickel-63, Niobium-95, Osmium-185+191, Palladium- 103, Platinum- 195m, Preseodymium-143, Promethium-147, Protactinium-233, RadiumO226, Rhenium-
  • Yttrium-90 Yttrium-91, Zinc-65 or Zirconium-95.
  • chelating groups which may be admixed with the microemulsions include the following: 1, 3, 5 triaminocyclohexane; 1, 3, 5 triaminocyclohexane N-pyridine; 1,1- cyclobutanedicarboxylic acid; l,2-Dimethyl-3-hydroxypyridin-4-one; 1 ,2-dimethyl- 3-hydroxypyridin-4-one (Deferiprone); 1, 4,7,10-tetraaza-4,7, 10- tris(carboxymethyl)- 1 -cyclododecylacetyl-i?-(+)-R-methylbenzylamine (DOTA- MBA); l,4,7,10-tetraazacyclododecane-N, ⁇ ' ' ,N 5 ⁇ ' ' -tetraacetic acid (DOTA); 1,6- dimethyl-2-(l-hydroxyethyl)-3-hydroxypyridin-4-one; l-ethyl-2- (1-
  • PCAH 2-pyridylcarboxaldehyde p-hydroxybenzoyl hydrazone
  • PCHH 2-pyridylcarboxaldehyde thiophenecarboxyl hydrazone
  • PCTH 3-11 2-hydroxy-l- napthylaldehyde isonicotinoyl hydrazone
  • 3-aminopyridine-2-carboxaldehyde thiosemicarbazone Triapine®
  • 3-aminopyridine-2-carboxyaldehyde thiosemicarbazone 3-hydroxypyridin-2-one; 3-hydroxypyridin-4-one; 4-[3,5-bis-
  • D-penicillamine N-ethyl N, N 5 N-tris(pyridylmethyl)-cis,cis, 1,3,5,- triaminocyclohexane; NOTP ( 1 ,4,7-triazacyclononane- 1 ,4,7- tris(methylenephosphonate)); NOTPME (1 ,4,7-triazacyclononane-l ,4,7- tris(methylenephosphonatemonoethylester)); N-pyridine; NTA nitrilotriacetic acid; oxalic acid; pyridoxal hydrochloride; pyridoxal isonicotinoyl hydrazone; Pyridoxal isoonicotinoyl hydrazone (PIH); pyridoxal metachlorobenzoyl hydrazone; pyridoxal metafluorobenzoyl hydrazone; pyridoxal paramethoxybenzoyl hydrazone; rh
  • Chelates for use during imaging are readily formed by mixing the appropriate radionuclide (formulated within the microemulsion) with an equimolar amount of the particular chelating moiety (also formulated within the microemulsion).
  • the resultant radionuclide-chelate forms spontaneously within the microemulsion and can be used almost immediately for topical application.
  • Microemulsions can also be used to topically introduce chemotherapeutic agents and cytotoxic compounds to a site close to the tumor. This approach avoids the toxicity common with the systemic or oral delivery of most chemotherapies.
  • Particular cancers that may be treated in this way include skin cancers such as melanoma, squamous cell carcinoma, basal cell carcinoma, solar keratosis as well as breast carcinoma.
  • Examples of chemotherapeutic agents which may be admixed with the microemulsions include the following:
  • Antimetabolites substances that interfere with the body's chemical processes, such as creating proteins, DNA, and other chemicals needed for cell growth and reproduction; in cancer treatment, antimetabolite drugs disrupt DNA production, which in turn prevents cell division. Examples include Azaserine, D-Cycloserine, Mycophenolic acid, Trimethoprim, 5-fluorouracil, capecitabine, methotrexate, gemcitabine, cytarabine (ara-C) and fludarabine.
  • Antitumor antibiotics which interfere with DNA by stopping enzymes and mitosis or altering the membranes that surround cells. These agents work in all phases of the cell cycle. Thus, they are widely used for a variety of cancers. Examples of antitumor antibiotics include dactinomycin, daunorubicin, doxorubicin
  • Mitotic inhibitors which are plant alkaloids and other compounds derived from natural products. They can inhibit, or stop, mitosis or inhibit enzymes for making proteins needed for reproduction of the cell. These work during the M phase of the cell cycle. Examples of mitotic inhibitors include paclitaxel, docetaxel, etoposide (VP- 16), vinblastine, vincristine, and vinorelbine.
  • Steroids which are natural hormones and hormone-like drugs that are useful in treating some types of cancer (lymphoma, leukemias, and multiple myeloma) as well as other illnesses.
  • chemotherapy drugs are often combined with other types of chemotherapy drugs to increase their effectiveness. Examples include prednisone and dexamethasone.
  • Sex hormones, or hormone-like drugs which alter the action or production of female or male hormones. They are used to slow the growth of breast, prostate, and endometrial (lining of the uterus) cancers, which normally grow in response to hormone levels in the body. These hormones do not work in the same ways as standard chemotherapy drugs.
  • anti-estrogens tamoxifen, fulvestrant
  • aromatase inhibitors anastrozole, letrozole
  • progestins megestrol acetate
  • anti-androgens bicalutamide, flutamide
  • LHRH agonists leuprolide, goserelin, histerillin, nafarelin, buserelin.
  • Alkylating agents which work directly on DNA to prevent the cancer cell from reproducing. As a class of drugs, these agents are not phase-specific (in other words, they work in all phases of the cell cycle). These drugs are active against chronic leukemias, non-Hodgkin's lymphoma, Hodgkin's disease, multiple myeloma, and certain cancers of the lung, breast, and ovary.
  • alkylating agents include busulfan, chlorambucil, cyclophosphamide, ifosfamide, dacarbazine (DTIC), mechlorethamine (nitrogen mustard), and melphalan.
  • Platinum and other heavy metals may also be used in the treatment of cancers.
  • platinum compounds and other compounds that may be used in the treatment of cancers include but are not limited to Cisplatin, oxaliplatin, carboplatin, diaminocyclohexylplatinin (DACH-platinum), nedaplatin, cycloplatan, SKI2053R, Loboplatin, enloplatin, zeniplatin, Miboplatin, L-NDDP, TRK-710, MBA (bis monobromo acetato trans 1 ,2-diaminocyclohexane platinum II), PYP (Bis-pyruvato 1,2-diaminocyclohexaneplatinum II), Sulphato 1,2 diaminocyclohexane platinum II, Malonato 1,2 diaminocyclohexane platinum II, Isocitrato 1,2-diaminocyclohexane platinum II, 4-Carboxyp
  • Lymphocide epratuzumab
  • MDX-210 Mylotarg
  • Panorex Edorcolomab
  • Rituxan Rituximab
  • Theragyn pemtumomab
  • Zamyl Zevalin (Ibritumomab)
  • Gefitinib Iressa
  • Erlotinib ABX-EGF
  • GSK 572016 lapatinibditosylate
  • CI-1033 canertinib diHCl
  • Bortezomib Arsenic trioxide
  • CC-394 Actimid
  • CDC-501 Revlimid
  • Thalidomide Thalomid
  • Oblimersen (Genasense), Abarelix (Plenaxis), Zevalin, Gemtuzumab, 2-MPPA, CPI-0004Na.
  • Nitrogen mustard in the form of its crystalline hydrochloride which is used as a drug in the treatment of Hodgkin's disease, non-Hodgkin's lymphomas, and brain tumors.
  • Cells vary in their susceptibility to nitrogen mustards, with rapidly proliferating tumor and cancer cells most sensitive; bone marrow, which produces red blood cells, is also sensitive, and depression of red blood cell production is a frequent side effect of nitrogen mustard therapy.
  • the nitrogen mustards also suppress the immune response (see immunity).
  • Other types include the aromatic mustards melphalan and chlorambucil, cyclophosphamide,
  • Nitrosoureas act in a similar way to alkylating agents. They interfere with enzymes that help repair DNA. These agents are able to travel to the brain so they are used to treat brain tumors as well as non-Hodgkin's lymphomas, multiple myeloma, and malignant melanoma. Examples of nitrosoureas include carmustine (BCNU) and lomustine (CCNU).
  • Hormone agonists such as Leuprolide (Lupron, Viadur, Eligard) for prostate cancer, Goserelin (Zoladex) for breast and prostate cancers and Triptorelin (Trelstar) for ovarian and prostate cancers and nafarelin acetate (Synarel).
  • Microtubule inhibitors include "Vinca” alkaloids, taxoids and benzimidazoles.
  • Curcuminoid derivatives such as tetrahydrocurcumin (THC), sodium circuminate, bisdemethoxycurcumin, demethoxycurcumin, 5'- methoxycurcumin and dihydrocurcumin, tetrahydrodemethoxycurcumin (INCI: tetrahydrodemethoxy- diferuloymethane), and tetrahydrobisdemethoxycurcumin (INCI: tetrahydro- bisdemethoxydif eruloylmethane .
  • TSA Trichostatin A
  • the present invention contemplates a topical composition comprising 99m Tc chelated to DTPA for the identification of a sentinel lymph node in relation to melanoma or breast carcinoma.
  • the present invention contemplates a topical composition comprising one or more chemotherapeutic agents for the treatment of skin or breast cancer.
  • the present invention contemplates a topical composition comprising a curcuminoid derivative for the treatment of melanoma or breast carcinoma.
  • the present invention contemplates a topical composition comprising TSA for the treatment of melanoma or breast carcinoma.
  • the present invention contemplates a topical composition comprising an anthracyclin for the treatment of breast carcinoma.
  • This composition may optimally also contain 5-fluorouracil and chlorambucil.
  • the present invention contemplates a topical composition comprising a chelate between an imaging agent and a chelating agent.
  • a chelate between an imaging agent and a chelating agent.
  • examples include, but are not limited to 99m -Tc-HYNIC, 99m -Tc-DTPA, "" ⁇ -ciprofloxacin, 99m Tc-methylene diphosphonate (MDP), indium or technetium- 99 " 1 hexamethylpropylene amine oxime (HMPAO), 11 1 In-DTP A-Folate, 99m Tc diadenosine tetraphosphate (Ap4A; AppppA, -Pl,.P4-di(adenosme-5*)- tetraphosphate) and its analog 99m Tc AppCHClppA, 99m Tc-HYNIC-IL-8., 99m Tc-
  • the present invention contemplates a topical composition comprising a chemotherapeutic agent linked to a targeting agent.
  • a chemotherapeutic agent selected from Amphiregulin, Antibodies including Avastin
  • GM-CSF GM-CSF, hGH, hGHR, Growth Hormone Releasing Hormone and analogs, Growth Hormone Releasing Peptides, IFN, IgG, IGF-I, IGFBP-3, Interleukins, Langerin, LHRH analogs, LIF, L-Selectin, Lymphotoxin-a, MC-148, MCP-I, MIP-Ia, MIP- Ib, MLIF, NGF, NGFR, OSM, OX-40, PDGF, RANTES, Tf, TGF, TGF-bl, TNF, TNF-a, TPO, TrkA, TrkB, VEGF, Vitamin derivatives, including derivatives of vitamin B 12, biotin, folate and/or riboflavin.
  • microemulsions of the present invention can be used to treat melasma and other areas where the skin contains no pigment or to lighten the skin where there areas of colored blotches.
  • the microemulsions are used to introduce nucleic acid molecules to induce gene expression in order to produce pigment. Melanin and other pigmentation molecules may also be introduced.
  • Examples of skin lightening, skin whitening and re-pigmentation molecules include estrogen-like compounds such as chromenes, isoflavones and coumestran groups of phyloestrogens (coumestan glycosides).
  • Examples of chromenes include microestrol and deoxymicroestrol.
  • Examples of isoflavones include diadzen, genisten, mirificin, puerarin and puerarin-6"-monoacetate.
  • Examples of coumestran glycosides include moumestrol, mirificoumestan, mirificoumestan glycol and mirificoumestran hydrate.
  • a useful source of estrogen-like compounds is from the plant genus Pueraria such as Pueraria lobata and Pueraria muriflca and hence the present invention extends to the estrogen-like compound contained within an extract of a Pueraria plant such as an extract of P. muriflca and/or P lobata.
  • Collagen fragment mimetics include dermaxyl (a palmitoyl oligopeptide), ceramide 2 (a natural skin lipid) and matrixyl 3000 which is a mixture of palmitoyl oligopeptides (e.g. palmitoyl-GHK and palmitoyl GQPR in a ratio of 2:1) may also be administered with the microemulsion alone or with an anti-oxidant.
  • dermaxyl a palmitoyl oligopeptide
  • ceramide 2 a natural skin lipid
  • matrixyl 3000 which is a mixture of palmitoyl oligopeptides (e.g. palmitoyl-GHK and palmitoyl GQPR in a ratio of 2:1) may also be administered with the microemulsion alone or with an anti-oxidant.
  • Suitable anti-oxidants include retinyl palmitate (Vitamin A) 5 allantoin (also called 5-ureidohydantoin, glyoxyldiureide and 5-ureidohydantoin; it is a diureide of glycoxylic acid) and D- ⁇ tocopherol (Vitamin E).
  • microemulsions of the present invention are a useful vehicle to introduce nucleic acid molecules such as DNA, siRNA, anti-sense RNA, effector RNA molecules and microRNAs to inhibit gene expression or to facilitate gene expression.
  • nucleic acid molecules such as DNA, siRNA, anti-sense RNA, effector RNA molecules and microRNAs
  • Reference to gene therapy in this regard includes the introduction of virosomes or constructs to facilitate gene expression.
  • Gene therapy is useful inter alia to the treatment of cancer, diabetes and inflammatory conditions.
  • the present invention contemplates a topical composition comprising a siRNA for the treatment of cancer.
  • a microemulsion of the present invention may be used in the formation of DNA/Ca++ nanoparticles.
  • the DNA/Ca++ nanoparticles of the present invention can be used to increase the frequency of in vitro cellular transfection.
  • the microemulsions of the present invention are useful inter alia for delivering small molecules, proteins (including peptides and polypeptides and chimeras), antibodies, other water soluble molecules as well as oil-soluble molecules useful in the treatment of muscular dystrophy.
  • small molecules with therapeutic potential in the treatment of muscular dystrophy is trichostatin A (TSA), an inhibitor of the enzyme deacetylase.
  • TSA trichostatin A
  • the present invention contemplates a topical composition comprising TSA for the treatment of muscular dystrophy.
  • microemulsions of the present invention may be used to introduce analgesic agents such as to include but not limited to fentanyl, oxycodone, codeine, dihydrocodeine, dihydrocodeinone enol acetate, morphine, desomorphine, apomorphine, diamorphine, pethidine, methadone, dextropropoxyphene, pentazocine, dextromoramide, oxymorphone, hydromorphone, dihydromorphine, noscapine, papverine, papveretum, alfentanil, buprenorphine and tramadol and pharmaceutically acceptable salts, derivatives, homologs or analogs thereof as well as opioid agonists.
  • analgesic agents such as to include but not limited to fentanyl, oxycodone, codeine, dihydrocodeine, dihydrocodeinone enol acetate, morphine, desomorphine, apomorphine, diamorphine, peth
  • neuropathic and nociceptic pain may be treated and agents useful for either neuropathic or nociceptic pain may be introduced via the microemulsion.
  • Pain includes the treatment of sport injuries, such as but not limited to, inflammatory conditions, planta fasciatis, bursitis of the knee, elbow, shoulder, subcutaneous prepatella bursa, olecranon bursa, tenosynovitis, Carpal Tunnel syndrome, bruising, Repetitive Strain Injury (RSI), patellotendonitis, paratendonitis, proliferative tendonitis, Degenerative tendonitis, enthesis and other conditions such as delayed onset muscle soreness (DOMS) and stings.
  • RSI Repetitive Strain Injury
  • patellotendonitis paratendonitis
  • proliferative tendonitis proliferative tendonitis
  • Degenerative tendonitis enthesis and other conditions such as delayed onset muscle soreness (DOMS) and stings.
  • the present invention contemplates a topical composition comprising one or more analgesic agents for the treatment of neuropathic or nociceptic pain.
  • the treatment of pain involves the use of a microemulsion containing a nitric oxide scavenger or an inhibitor of NOS such as Niacinamide, vitamin B 12 and derivatives, curcumin and derivatives L-NAME 5 JV 0 - nitro-L-arginine methyl ester; L-NMA, JV ⁇ -methyl-L-arginine, L-NMMA, JV 0 - 8 000908
  • the microemulsion of the present invention can be used to topically apply agents which are useful in treating erectile dysfunction including inhibitors of cGMP specific phosphodiesterases type 5 (PDE5). These molecules act by inhibiting the breakdown of nitric oxide (NO). Examples of PDE5 inhibitors include sildenafil, tadalafil and vardenafil. It is proposed that the microemulsion be topically applied to overcome erectile dysfunction. This approach avoids the side effects often associated with oral delivery of PDE5 inhibitors, including headache, lightheadedness, dizziness, flushing and change in vision. In a related embodiment, the present invention contemplates a topical composition comprising a PDE5 inhibitor for the treatment of erectile dysfunction.
  • PDE5 inhibitors include sildenafil, tadalafil and vardenafil.
  • Persistent, painful erection of the penis is relatively common in young males with sickle cell anaemia but can also result following oral delivery of PDE5 inhibitors. It is therefore contemplated that the microemulsion of the present invention can be used to topically apply agents which enhance the break-down of
  • NO for the treatment of persistent erection examples include NO chelators such as vitamin B12 derivatives as well as inhibitors of NO synthase (e.g. Niacinamide, curcumin and derivatives, L-NAME ⁇ -nitro-L-arginine methyl ester; L-NMA, ⁇ -methyl-L-arginine, L-NMMA, JV°-monomethyl-L-arginine, S- Methylisothiourea sulphate, Aminoguanidine. 7-nitro indazole, N-nitro-L-arginine).
  • NO synthase e.g. Niacinamide, curcumin and derivatives, L-NAME ⁇ -nitro-L-arginine methyl ester; L-NMA, ⁇ -methyl-L-arginine, L-NMMA, JV°-monomethyl-L-arginine, S- Methylisothiourea sulphate, Aminoguanidine. 7-nitro indazole
  • the present invention contemplates a topical composition comprising an NO chelator for the treatment of persistent erection. In another embodiment, the present invention contemplates a topical composition comprising an NO synthase inhibitor for the treatment of persistent erection. 13. Acne Treatment.
  • the microemulsion of the present invention can be used to topically treat acne.
  • agents which are useful in treating acne include curcumin, tetrahydrocurcumin, sodium circuminate, bisdemethoxycurcumin, demethoxycurcumin, 5'- methoxycurcumin and dihydrocurcumin, vitamin E, selenium, zinc, Vitamin C, azaleic acid, pantothenic acid, benzyl peroxide and extract of tea tree oil.
  • the present invention contemplates a topical composition
  • a topical composition comprising one or more compounds selected from the list of: Curcumin, tetrahydrocurcumin, sodium circuminate, bisdemethoxycurcumin, demethoxycurcumin, 5'- methoxycurcumin and dihydrocurcumin, vitamin E, selenium, zinc, vitamin C, azaleic acid, pantothenic acid, benzyl peroxide and extract of tea tree oil for the treatment of acne.
  • the microemulsion of the present invention can be used to topically treat oxidative damage associated with neurological disease.
  • diseases include neurodegenerative conditions like Parkinson's disease, Alzheimer's disease and other dementias as well as amyotrophic lateral sclerosis and multiple sclerosis.
  • Other neurological diseases include those following acute injury such as stroke and spinal cord damage.
  • agents which are useful in treating oxidative damage associated with neurological disease include NO chelators such as vitamin B 12 derivatives as well as inhibitors of NO synthase (e.g. Niacinamide, vitamin B12 and derivatives, curcumin and derivatives L-NAME 5 N 0 -nitro-L-arginine methyl ester; L-
  • the present invention contemplates a topical composition comprising an NO synthase inhibitor for the treatment of acute CNS injuries such as stroke and spinal cord damage. 15. Treatment of Abrasions.
  • the microemulsion of the present invention can be used to topically treat skin damage associated with cuts and abrasions. Such skin damage is susceptible to microbial infection.
  • Povidone-iodine type compounds such as Betadine are commonly used broad-spectrum topical microbicides as treatments for abrasions but can display poor capacity to penetrate within the wound.
  • the present invention contemplates a topical composition comprising a povidone-iodine type compound for the treatment of skin damage associated with cuts and abrasions.
  • the microemulsion of the present invention can be used topically to ameliorate the symptoms of skin aging, especially in relation to the face.
  • Symptoms of facial (and other non facial) skin ageing include wrinkles, a lowering of the collagen content of the dermis, facial "puffiness", the appearance of hyperpigmentation adjacent to the eyes, photo-aging of skin such as by free radical scavenging and an increase in trans- epidermal water loss from the skin.
  • agents which are useful in counteracting the effects of skin ageing include collagen stimulating agents such as Dermican, Pueraria Mirifica Extract; anti-wrinkle agents such as Vialox, SynAke,
  • Gatuline Expression melanin inhibitors and/or tyrosinase inhibitors including Haloxyl, phytoestrogens, Melaclear2, Lumiskin, Sepiwhite and stilbene compounds such as resveratrol; compounds that affect skin "puffiness including Haloxyl, Eyeliss, Esculoside and other mixtures containg esculin, anti-bacterial compounds suitable for application to skin including Triclosan; exfoliation compounds suitable for application to skin including bromelain and papain.
  • microemulsion refers to a system comprising an oil phase, a water phase, one or more zwitterionic surfactants; one or more co-surfactants and an active agent wherein the microemulsion is a single optically isotropic and thermodynamically stable liquid solution.
  • transdermal delivery is not to be construed as passage of an agent solely through the dermal layer although such passage is also contemplated by the present invention. The term provides passage of agents through all layers of the skin.
  • the microemulsion of the present invention comprises an oil phase, a water phase and one or more zwitterionic surfactantsand an active agent, wherein the microemulsion is capable of transdermal delivery of the active agents.
  • the microemulsions further comprises a co-surfactant, a co-solvent, or a combination thereof.
  • microemulsion of the present invention may be produced by methods known in the art.
  • microemulsions are produced by emulsifying components under conditions including typically sufficient force or the required temperature to generate the required dispersion level, conductivity, viscosity, percolativity or other dispersion characteristics.
  • Microemulsion formation can be assessed using scattering and spectroscopic techniques such as neutron scattering, time-average scattering, quasi-electric light scattering i.e., high- resolution ultrasonic spectroscopy or photon correlation spectroscopy.
  • the partition coefficients of microemulsions may also be measured chromatographically.
  • formulations are based on a number of different paradigms depending upon the desired application.
  • Illustrative paradigms include the hydrophilic- lipophilic balance, the phase-inversion temperature, or the cohesive-energy ratio.
  • Microemulsions may be formulated using a wide range of immiscible liquids and other additional agents.
  • the microemulsion of the present invention may comprise an oil phase of between 50 and
  • microemulsion may further comprise 0.1 to 90% by weight co-surfactant or cosolvent; preferably 1 to 90% by weight co-surfactant or cosolvent.
  • the oil phase may comprise natural oils derived from plants or animals, such as vegetable oils, sunflower oils, coconut oils, almond oils; purified synthetic or natural di or triglycerides (such as Crodamol GTCCTM and Capmul MCMTM); phospholipids and their derivatives (such as lecithin or lysolecithin); fatty acid esters (such as isopropyl myristate, isopropyl palmitate, ethyl oleate, oleic acid ethyl ester); hydrocarbons (such as hexane, the n-decane through n-octadecane series); and/or glycerolysed fats and oils (such as glyceryl monooleate, glyceryl monocaprylate, glycerol monocaprate, propylene glycol monocaprylate, propyleme glycol monolaurate).
  • natural oils derived from plants or animals such as vegetable oils, sunflower oils, coconut oils, almond oils; pur
  • oil phase ingredients include, but are not limited to, Labrafil M 1944 CSTM, benzene, tetrahydrofuran, and n-methyl pyrrolidone, or halogenated hydrocarbons, such as methylene chloride, or chloroform.
  • the oil phase comprises Crodamol GTCCTM and Capmul MCMTM, at 3: 1 ratio.
  • the oil component is either used alone or in combination with other oil component.
  • Each oil or unique mixture of oils may require a different surfactant or mixture of surfactants or surfactants and co-surfactants to form a microemulsion with the water phase, as can routinely be determined by those of skill in the art.
  • Water phase ingredients may comprise water and any water-soluble components in water, including one or more active agents.
  • Zwitterionic surfactants are used in the compositions and methods of the present invention. These surfactants are very mild and can be anionic (negatively charged), cationic (positively charged) or non-ionic (no charge) in solution, depending on the acidity or pH of the water.
  • Zwitterionic surfactants may contain two charged groups of different sign. Whereas the positive charge is almost always ammonium, the source of the negative charge may vary (carboxylate, sulphate, sulphonate).
  • Zwitterionic surfactants include the betaines, sulfobetaines, i.e. sultaines and fatty acid ethanolamides.
  • betaines include dodecyldimethylammonium acetate, tetradecyldimethylammonium acetate, hexadecyldimethylammonium acetate, alkyldimethylammonium acetate wherein the alkyl group averages about 14.8 carbon atoms in length, dodecyldimethylammonium butanoate, tetradecyldimethylammonium butanoate, hexadecyldimethylammonium butanoate, dodecyldimethylammonium hexanoate, hexadecyldimethylammonium hexanoate, tetradecyldiethylammonium pentanoate and tetradecyldipropyl ammoni
  • fatty acid betaines include coco-betaine.
  • fatty acid ethanolamides include cocamide monoethanolamide.
  • Still other examples of zwitterionic surfactants include 3-(N,N- dimethyl-N-hexadecylammonio)-propane-l -sulfonate; 3-(N,N-dimethyl-N- hexadecylammonio)-2-hydroxypropane- 1 -sulfonate; N,N-dimethyl-N-dodecylammonio acetate; 3-(N,N-dimethyl-N-dodecylammonio)propionate; 2-(N,N-dimethyl-N- octadecylammonio)ethyl sulfate; 3-(P,P-dimethyl-P-dodecylphosphonio)propane-l- sulfonate; 2-(S-methyl-S-tert-hexa
  • Co-surfactants used according to the subject invention are known surface-active agents in the art that act, in addition to surfactants, to further lower the interfacial energy of a microemulsion.
  • Co-surfactants include, but are not limited to non-toxic amphiphilic molecules; alcohols (including aliphatic alcohols, shorter chain alcohols, such as ethanol); fatty acid alcohols (such as n-alkane-l,2-diols); acids (such as acetic acid); esters (such as butyl lactate); anionic co-surfactants, cationic co-surfactants and non-ioninc co-surfactants; any surfactants as herein listed or mixtures thereof.
  • Alcohol content may range, for example, from 0 to about 30% by volume in the microemulsion.
  • Anionic co-surfactants include fatty acid soaps (including sodium oleate, sodium palmitate, sodium myristate, sodium sterate, potassium oleate and triethanolamine oleate); alkyl sulfates (including sodium dodecyl sulfate, ammonium lauryl sulfate, triethanolamine lauryl sulfate and sodium alkyl sulfate); alkyl lactylates (including calcium stearoxyl-2-lactylate), alkyl lactates (including sodium-0-stearyllactate and sodium stearoyllactylate) alkyl benzenesulfonates (including calcium dodecyl benzene sulfonate); alkyl sulfonates (including alkyl aryl sulfonate); alkyl phosphates; alkyl oleates; alkyl stearates (including self-emulsifying glycerol monostearate);
  • Cationic co-surfactants include alkyl primary, secondary, tertiary, or quaternary amines; high-molecular-weight amine and fatty amine blends; polyoxyethylene fatty amines (including tallow amine); alkyl sulfates (including N-cetyl-N-ethyl morpholinium ethyl sulfate (35%)); alkyl pyridinium and quaternary ammonium salts.
  • Non-ionic co-surfactants include alcohol ethoxylate, alkylphenol ethoxylate, fatty acids (such as oleic acid), lanolin alcohols (such as polyoxyethylene (5) lanolin alcohol (ether and ester), polyoxyethylene (50) lanolin (ether and ester), acetylated polyoxyethylene (10) lanolin, polyoxyethylene (16) lanolin alcohol, acetylated polyoxyethylene (9) lanolin), alkyl polyglycosides, mono-, di- or glyceride esters (such as diglycerine sesquioleate), acetylated monoglycerides, polyglycerols, polyglycerol esters (such as decaglycerol decaoleate, decaglycerol octaoleate, decaglycerol tetraoleate), phospholipids (such as lecithin), mono- or diglyceride esters of citric acid, tartaric acid and
  • Zwitterionic surfactants in the microemulsion of the present invention may be used alone or in combination with each other.
  • microemulsion of the present invention may further comprise solvents or other agents to enhance emulsion formation or stability.
  • Other agents may be introduced to provide functions such as pH, ionic content, polymerisation, taste, smell, sterility, colour, viscosity etc.
  • microemulsions of the present invention may also be generated using any suitable synthetic plastic or polymeric, monomeric or hybrid colloidal material.
  • the present invention further extends to a pharmaceutical composition
  • a pharmaceutical composition comprising the microemulsion and one of more active or pharmaceutical agents as described herein, wherein such pharmaceutical composition is suitable for topical application on a subject.
  • Suitable pharmaceutical agents for use in the present invention include any water soluble solute, including, but not limited to peptides, proteins, polysaccharides, oligonucleotides, salts, sugars, nutrients, vitamins, nucleic acids minerals, acids, anti-oxidants, or any biological active compounds for administration to a subject, such as a human, animal or other mammal.
  • the pharmaceutical agent is selected based upon the intended application or therapy, wherein the effect of the pharmaceutical agent is suitable to treat a particular condition.
  • Suitable peptides and proteins that can be delivered via the microemulsion of the present invention include molecules, protein chimeric molecules, other chimeric molecules or fragments selected from the TNF superfamily (including TNF- ⁇ , TNFRl, TNFR2, BAFF, BAFFR, OX-40, Lymphotoxin- ⁇ , Fas-ligand); chemokines (including MCP-I, MIP-Ia, MIP-Ib 3 RANTES, IL-8 and viral like chemokine antagonist MC148); interleukins, interleukin receptors and antagonist (including IL-Ia IL-Ib 5 IL-2, IL-3, IL-4, IL-5, IL-6, IL- 7, IL-8, IL-10, IL-I l 5 IL-12, IL-13, IL-15, IL-16, IL-18, IL-20, IL-21, IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28,
  • Suitable chimeric proteins that can be delivered via the microemulsion of the present invention include TNFRl-Fc, TNFR2-Fc, OX-40-Fc, MC148-Fc, IL-IRa-Fc, IL-2Ra-Fc,
  • IL-2Rb-Fc IL-2Rb-Fc
  • IL-2Rg-Fc IL-3Ra-Fc
  • IL-4Ra-Fc IL-5Ra-Fc
  • IL-6Ra-Fc IL-7Ra-Fc
  • IL-IORa IL-2Rb-Fc, IL-2Rg-Fc, IL-3Ra-Fc, IL-4Ra-Fc, IL-5Ra-Fc, IL-6Ra-Fc, IL-7Ra-Fc, IL-IORa-
  • the active agent of the present invention comprises MLIF or analogs thereof, such as iodacetylated MLIF.
  • suitable proteins include monoclonal and polyclonal antibodies, single-chain antibodies, other antibody fragments, analogs and derivatives thereof.
  • Polynucleotides, including antisense oligonucleotides, aptamers and therapeutic genes can also be delivered using the methods and compositions of the present invention.
  • suitable antibodies that can be delivered via the microeniulsion of the present invention include antibodies directed against TNF alpha, IL-I, IL-8, IL-6 or other cytokines involved in the inflammatory response and, most preferably, anti-TNF alpha antibodies with the technical names of infliximab and adalimumab.
  • Anticoagulants such as Heparin, Warfarin, Herbal extracts such as those derived from Danshen Devil's Claw, Eleuthero, Garlic, Ginger, Ginkgo, Horse Chestnut, Panax Ginseng, Papain, Red Clover, Saw Palmetto, capsaicin, also can be delivered using the methods and compositions of the invention.
  • bioactive molecules such as anticancer drugs, e.g., Curcumin derivatives, Glycyrrhizin, Glycyrrhetinic acid, antracycline, platinum drugs, methotrexate derivatives, heary metals, genistein, chlorambucil, cyclophosphamide, melphalan, cyclopropane, doxorubicin, daunomycin, adriamycin, mitomycin C, [2-(hydroxymethyl)anthraquinone], methotrexate, dichloromethatrexate, cisplatin, carboplatin, metallopeptides containing platinum, copper, vanadium, iron, cobalt, gold, cadmium, zinc and nickel, DON, thymidine, pentamethylmelamin, dianhydrogalactitol, 5-Methyl-THF, anguidine, maytansine, ne
  • anticancer drugs e.g.,
  • NSAIDs include salicylates (including aspirin, choline magnesium trisalicylate, dilfunisal, salasalate, benorylate); phenylalkanoic acids (including carpofen, fenoprofen, fluribiprofen, ibuprofen, ketoprofen, naproxen, naproxen Na, suprofen, oxaprozin,); acetic acids or indoles (including alclofenac, diclofenac, fenclofenac, indomethacin, sulindac, tolemetin); enolic acids (including isoxicam, meloxicam, piroxicam, tenoxicam); fenamic acids (including flufenamic acid, meclofenamate, mefenamic acid); napthylalkanone
  • the active agent suitable to be delivered via the microemulsion of the present invention comprises copper complexes of NSAIDs. Formation of copper complexes of NSAIDs reduces toxicity and may enhance the anti-inflammatory properties of the complex due to the anti-inflammatory property of copper.
  • Such pharmaceutical agents include copper complexes of salicylates (including aspirin, choline magnesium trisalicylate, dilfunisal, salasalate, benorylate); copper complexes of phenylalkanoic acids (including carpofen, fenoprofen, fluribiprofen, ibuprofen, ketoprofen, naproxen, naproxen Na, suprofen, oxaprozin); copper complexes of acetic acids or indoles (including alclofenac, diclofenac, fenclofenac, indomthacin, sulindac, tolemetin); copper complexes of enolic acids (including isoxicam, meloxicam, piroxicam, tenoxicam); copper complexes of fenamic acids (including flufenamic acid, meclofenamate, mefenamic acid); copper complexes of napthylalkanones (including nabu
  • the active agent may be selected from copper complexes of tyrosine, lysine, valproic acid, 2, 2,-bypyridine, 1, 10-phenanthroline, 2, 9-dimethyl-l, 10- phenanthroline, glycylglycine, cimetidine, sulfathiazole, tetraanhydroaminmobenzaldehyde, metronidazole and imidazole.
  • Other potential complexes suitable for delivery as active agents include copper complexes with amino acids, aromatic carboxylic acid, corticoids, tetrazoles, histamines, penicillamines, anti-ulcer histamine antagonists, rantidine and cimetidine.
  • the aforementioned copper complexes may further comprise one of more molecules of
  • DMF DMF, DMSO, Pyridine, Caffeine, Methylimidazole, Imidazole, 2-methylbenzimidazole, metronidazole, 2-methylimidazole, 3-picoline, 4-picoline, imidazole, 1 -methylimidazole, diethylamine, nicotinamide, papaverine, N, N-dimethylacetamide, N-methyl-2- pyrrolidone, ethanol and methanol.
  • the pharmaceutical composition of the present invention comprising one or more copper- NSAID complexes may be formed by mixing two microemulsions of the present invention containing NSAID and copper respectively.
  • Suitable vitamins include but are not limited to water soluble vitamins such as vitamin C, analogs or derivatives thereof; folic acid and analogs or derivatives thereof (including but not limited to methotrexate, aminopterin, 10-deazaminopterin, 10-ethyl-lO- deazaaminopterin, 5,10-dideazatetrahydrofolate, folinic acid, 7-hydroxyaminopterin); niacin (nicotinic acid, vitamin B3) and analogs or derivatives thereof (including but not limited to beta-hydroxybutyrate, acipimox, niceritrol, nicotinamide (niacin)); thiamine (vitamin B-I), analogs and derivatives thereof; riboflavin (vitamin B2) and its analogs or derivatives thereof (including but not limited to 7-nor-7-chlororiboflavin, 8-nor-8- chlororiboflavin, 7-nor-7-bromoriboflavin, 8-nor-8-bromori
  • Suitable vitamin B 12 (VB 12) analogs include descobaltocorrinoids such as alpha (5,-6-dimethylbenzymidazolyl)-hydrogenobamide, hydroxocobalamin (OH-CbI), methylcobalamin, adenosylcobalamin (AdeCbl), aquocobalamin, methylcobalamin, cyanocobalamin, carbanalide, 5 methoxybenzylcyanocobalamin [(5-MeO)CN-CbI], as well as the desdimethyl, monoethylamide and the methylamide analogs of all of the above VB 12 molecules.
  • descobaltocorrinoids such as alpha (5,-6-dimethylbenzymidazolyl)-hydrogenobamide, hydroxocobalamin (OH-CbI), methylcobalamin, adenosylcobalamin (AdeCbl), aquocobalamin, methylcobalamin, cyanocobalamin
  • Suitable VB 12 analogs include all alkyl cobalamins in which the alkyl chain is linked to the corrin nucleus by a direct CoC covalent bond. Still other suitable VB 12 analogs include chlorocobalamin, sulfitocobalamin, nitrocobalamin, thiocyanatocobalamin, benzimidazolecyanocobalamin derivatives such as 5,6 dichlorobenzimidazole, 5- hydroxybenzimidazole, trimethylbenzimidazole, as well as adenosylcyanocobalamin [(Ade)CN-Cbl], cobalamin lactone, cobalamin lactam and the anilide, ethylamide, monocarboxylic and dicarboxylic acid derivatives of VB 12 or its analogs.
  • VB 12 derivatives include adeninylalkylcobalamin, adeninylethylcobalamin (AdeEtCbl), adeninylpropylcobalamin (AdePrCbl) and adeninylpentylcobalamin (AdePeCbl)).
  • Alternative derivatives of VB 12 include the mono, di- and tricarboxylic acid derivatives or the propionamide derivatives of VB 12.
  • suitable analogs of VB 12 include those in which the cobalt ion is replaced by either a zinc or nickel ion.
  • the active agent of the present invention comprises aquocobalamin, adenosylcobalamin, methylcobalamin, hydroxycobalamin, cyanocobalamin, carbanalide, or 5 methoxybenzylcyanocobalamin [(5-MeO)CN-CbI].
  • VB 12 analogs or derivatives of the present invention are involved in the sequestration and/or neutralization of nitric oxide and other reactive nitrogen species.
  • nitric oxide and nitric superoxide rapidly combine to form a toxic reaction product, peroxynitrite anion (ONOO).
  • the ratio of superoxide to NO is important in determining the reactivity of peroxynitrite: excess NO or excess superoxide reduces the oxidation elicited by peroxynitrite.
  • the oxidant reactivity of peroxynitrite is mediated by an intermediate with the biological activity of the hydroxyl radical.
  • Activated macrophages have been shown to secrete high levels of NO due to inducible NO synthase (iNOS).
  • Molecules such as vitamin B12 have been shown to reduce the activity of NO by complexation to the CN group. Additionally, the skin contains many dendritic cells, which have also been shown to produce iNOS when stimulated. Such stimulation may occur in clinical conditions such as psoriasis.
  • Suitable curcumin analogs include tetrahydrocurcumin, bisdemethoxycircumin, extracts from Curcuma xanthorrhiza or C. domestica such as 25 P54FP and extracts from C. phaeocaulis valeton.
  • Suitable curcumin derivatives include 5'- methoxycurcumin, dihydrocurcumin, cyclocurcumin and demethoxy curcumin, as well as curcumins and curcuminoids such as those described in the Chinese Pharmacopoeia; Yujin (from the tubers of C. wenyujin, C. longa, C. kwangsiensis or C. phaeocaulis); Jianghuang (the rhizome of C.
  • Pian-Jianghuang the rhizome of C. wenyujin
  • Ezhu the rhizomes of C. phaeocaulis, C. kwangsiensis or C. wenyujin
  • the rhizomes of C. zedoaria and C. aromatica as well as in the rhizomes of C. zedoaria and C. aromatica.
  • Additional molecules with anti-inflammatory properties include those that are structurally related to curcumin, such as diarylheptanoids obtained from Alpinia oxyphylla, several sesquiterpenes, germacrone, turmerone, ar-(+)-, a-, ⁇ - turmerones; ⁇ - bisabolene; a-curcumene; zingiberene; ⁇ - sesquiphellandene, bisacurone; curcumenone; dehydrocurdione; procurcumadiol; bis-acumol; curcumenol; isoprocurcumenol epiprocurcumenol; procurcumenol; zedoaronediol; curlone; and turmeronol A and turmeronol B, as well as four polysaccharides-ukonans - having activity on the Reticuloendothelial system, along with stigmasterol, ⁇ -sitosterol, cholesterol and 2- hydroxymethyl an
  • the active agent of the present invention comprises a curcumin analog and, in particular, tetrahydrocurcumin.
  • the pharmaceutical composition of the present invention comprises a microemulsion, an emulsion or a cream and one of more active agent as described herein, wherein such pharmaceutical composition is suitable for topical application on a subject.
  • the present invention further provides a method suitable for the transdermal delivery of active agents in a range of therapeutic, cosmetic and research applications.
  • the method comprises first preparing a water phase, wherein any water-soluble components, including any active agents, are dissolved in water. Oil soluble components are dissolved in the oil phase as appropriate. Any undissolved material is separated by centrifugation or filtration.
  • the oil and water phases and one or more suitable zwitterionic surfactants and co- surfactants are then mixed in a suitable vessel, and given time to equilibrate. Standard emulsification techniques, like stirring, use of membranes, applying shear, ultrasound and the like, can be used to facilitate the mixing of the two phases.
  • the pharmaceutical composition is then applied on a subject.
  • Examples of therapeutic, cosmetic and research applications of the present invention include any skin conditions, or any dermatologic disorders, including dermatitis, bacterial, fungal, parasitic, viral infections of the skin, disorders of hair follicles and sebaceous glands, scaling papular diseases, psoriasis, inflammatory reactions, reaction to sunlight, bullous diseases, disorders of cornification, pressure sores, pigmentation disorders, disorders of sweating, benign and malignant tumours.
  • Examples of dermatitis include contact dermatitis, atopic dermatitis, seborrheic dermatitis, nummular dermatitis, chronic dermatitis of hands and feet, generalized exfoliative dermatitis, stasis dermatitis, lichen simplex chronicus.
  • Examples of bacterial infections of the skin include cellulitis, acute lymphangitis, lymphadenitis, erysipelas, cutaneous abscesses, necrotizing subcutaneous infections, staphylococcal scalded skin syndrome, folliculitis, furuncles, hidrandenitis suppurativa, carbuncles, paronychial infections, erythrasma.
  • fungal skin infections include dermatophyte infections including tinea corporis, tinea pedis, tinea unguium, tinea capitis, tinea cruris, tinea barbae, dermatiphytids or Id Eruptions; yeast infections including candidiasis, tinea versicolor.
  • Examples of parasitic skin infections include scabies, pediculosis, creeping eruption.
  • Examples of viral skin infections include warts and molluscum contagiosum.
  • hair follicles and sebaceous glands disorders include acne, rosacea, perioral dermatitis, hypertrichosis, alopecia, pseudofolliculitis barbae, keratinous cyst.
  • scaling papular diseases include psoriasis, pityriasis rosea, lichen planus, pityriasis rubra pilaris.
  • Examples of inflammatory reactions of the skin include drug eruptions, toxic epidermal necrolysis, erythema multiforme, erythema nodosum, granuloma annulare.
  • Examples of skin reaction to sunlight include sunburn, chronic effects of sunlight, such as actinic keratoses, squamous and basal cell carcinoma and malignant melanomas, and photosensitivity.
  • Examples of bullous diseases include pemphigus, bullous pemphigoid, dermatitis herpetiformis, linear immunoglobulin A disease.
  • cornification disorders include ichthyosis, keratosus pilaris, calluses and corns.
  • Pressure sores results from ischemic necrosis and ulceration of tissues overlying a bony prominence that has been subjected to prolonged pressure against an external object, such as bed, wheelchair, cast or splint.
  • Examples of pressure sores include bedsores, decubitus ulcers, trophic ulcers.
  • pigmentation disorders include hypopigmentation and hyperpigmentation.
  • sweating disorders include miliaria and hyperhidrosis.
  • benign tumours include moles, dysplastic nevi, skin tags, lipomas, angiomas, pyogenic granuloma, seborrheic keratoses, dermatofibroma, keratoacanthoma, keloid.
  • malignant tumours include basal cell carcinoma, squamous cell carcinoma, Bowen's disease, malignant melanoma, breast cancer, Paget' s disease of the nipples, and Kaposi's sarcoma.
  • cosmetic applications of the present invention include the amelioration of skin wrinkles, the lightening of skin colour, the amelioration of facial "puffiness”, the cleansing of the skin of bacteria and skin exfoliation.
  • therapeutic, cosmetic and research applications of the present invention include any musculoskeletal and connective tissue disorders, including rheumatoid arthritis, Sjogren's syndrome, Behcet's syndrome, relapsing polychondritis, systemic lupus erythematosus, discoid lupus erythematousis, systemic sclerosis, eosinophilic fasciitis, polymyositis, dermatomyositis, polymyalgia rheumatica, vasculitis, termporal arteritis, polyarteritis nodosa, Wegener's granulomatosis, mixed connective tissue disease, ankylosing spondylitis, Reiter's syndrome, psoriatic arthritis, osteoarthritis, neurogenic arthropathy, avascular necrosis, infectious arthritis, ostemyelitis, gout, idiopathic hyperuricemia, calcium pyrophosphate dihydrate crystal de
  • therapeutic, cosmetic and research applications of the present invention include inflammatory diseases not related to skin or musculoskeletal and connective tissue such as multiple sclerosis, Crohn's disease and ulcerative colitis.
  • An oil phase of a 3:1 ratio of Crodamol GTCC to Capmul MCM C8 is prepared.
  • a surfactant/co-surfactant phase of a 3:2:0.75 ratio of Crillet 4 (Tween 80) to Crill 4 (Span 80) is prepared.
  • An oil phase of a 3:1 ratio of Crodamol GTCC to Capmul MCM C8 was prepared.
  • a surfactant/co-surfactant phase of an 8:5:2 ratio of Crillet 4 (Tween 80) to Dehyton AB30 (coco-betaine) to Crill 4 (Span 80) was also prepared.
  • An oil-surfactant phase was then prepared by mixing the oil and surfactant phases at a ratio of 6.8:2.2. Microemulsions were formed by mixing the oil-surfactant phase with one ninth - one tenth volume of water.
  • microemulsion formation was often assisted by gentle shaking and/or heating the mixture to 50 degrees Celsius.
  • An oil phase of a 3:1:0.9 ratio of Crodamol GTCC to Capmul MCM C8 to phytosqualene (Sophim) was prepared.
  • An oil-surfactant phase was then prepared by mixing the oil and surfactant phases at a ratio of 6.8:2.2. The following preparations were added to the oils/surfactant: 15-20 mg/ml salicylic acid, 5-15 mg/ml ginger extract, 5-10 mg/ml resveratrol powder, 2-6 mg/ml tetrahydrocurcumin powder. Microemulsions were formed by mixing the oil-surfactant phase with one ninth - one tenth volume of water and assisted by gentle shaking and / or heating the mixture to 50 degrees Celsius.
  • An oil phase consisting of a 3:1 ratio of Crodamol GTCC to Capmul MCM C8 was prepared.
  • a surfactant/co-surfactant phase consisting of an 8:5:2 ratio of Crillet 4 (Tween 80) to Dehyton AB30 (coco-betaine; Cognis) to Crill 4 (Span 80) was also prepared.
  • An oil/surfactant phase was thus prepared by mixing the oil and surfactant / co-surfactant phases at a ratio of 6.8:2.2. Microemulsions were formed by mixing the oil-surfactant phase with one ninth - one tenth volume of water.
  • One or more of the following agents were dissolved in the microemulsions: Pueraria Mirifica root extract (to yield a final concentration of 8-10 mg/ml; Smith Naturals), Gatulene Expression (to yield a final concentration of 50 mg/ml; Trapeze), Dermican (to yield a final concentration of 30 mg/ml; Cognis) and Vialox (to yield a final concentration of 5 mg/ml; Pentapharm).
  • Pueraria Mirifica root extract to yield a final concentration of 8-10 mg/ml
  • Smith Naturals Gatulene Expression
  • Dermican to yield a final concentration of 30 mg/ml; Cognis
  • Vialox to yield a final concentration of 5 mg/ml; Pentapharm.
  • Microemulsion formation was often assisted by gentle shaking and / or heating the mixture to 50 degrees Celsius.
  • a 3:1 ratio of Crodamol GTCC to Capmul MCM C8 was prepared (oils).
  • a second surfactant mixture of an 8:5:1 ratio of Crillet 4 (Tween 80) to Dehyton AB30 (coco- betaine, Cognis) to cocamide MEA (Cognis) was also prepared.
  • the oil/surfactant phase was prepared by mixing the oil with surfactant at a ratio of 6.8:2.2.
  • Microemulsions were formed by mixing the oil-surfactant phase with one ninth - one tenth volume of water.
  • One or more of the following agents were dissolved in the microemulsions: Eyeliss (to yield a final concentration of 10-35 mg/ml; Croda), Haloxyl (to yield a final concentration of 15- 28 mg/ml; Croda) and Esculoside (to yield a final concentration of 1-4 mg/ml; Croda).
  • Eyeliss to yield a final concentration of 10-35 mg/ml; Croda
  • Haloxyl to yield a final concentration of 15- 28 mg/ml
  • Croda Croda
  • Esculoside to yield a final concentration of 1-4 mg/ml; Croda.
  • Microemulsion formation was often assisted by gentle shaking and / or heating the mixture to 50 degrees Celsius.
  • Microemulsions are prepared in accordance with one of the above-mentioned microemulsion formulation Examples 1 to 5, wherein the active agents include a 125 I- labeled-protein. Suitable proteins may include BSA, epidermal growth factor or a recombinant human TNFR chimeric protein such as that described in PCT Application No. WO 06/079176. Ionodation can be performed using a standard method known in the art. The microemulsions are then applied to a 2 x 1 cm area of the shaved skin of Balb/C mice.
  • microemulsion is left on the mice and after 180 minutes or overnight the mice are euthanased and all the organs removed and counted in a gamma counter to determine the distribution of 125 I-labeled-proteins. It is anticipated this data will show that the microemulsion is capable of delivering the applied water-soluble protein to the muscle and adjacent skin tissue.
  • Vitamin B 12 analogs including cyanocobalamin, hydroxycobalamin, cobalamin, adenosyl-cobalamin (all obtained from Sigma Pharmaceuticals), MLIF (synthesized by Auspep, Melbourne, Australia), tetrahydrocurcumin, recombinant human TNF receptor II - IgGl Fc chimera molecules, for example, molecules of which the production and purification is described in PCT Application No. WO 06/079176; are dissolved in the appropriate phase (depending upon the specific polar characteristics of the molecule) prior to the formation of separate microemulsions as described above in the microemulsion formulation examples.
  • the final concentrations range of each ingredient in the subsequent microemulsions is 2-25 mg/ml.
  • One of the standard models of acute or chronic inflammation is induced in animals of the appropriate species.
  • acute inflammation is induced in the rear right footpad of C57/B16 strain mice by an intra-pad injection of 50 ⁇ l of 1% llambda Carrageenan.
  • mice receive treatment, which consisted of the application of 20 microL of atopical composition containing one or more anti-inflammatory molecules, as described above, applied to a previously shaved area of the belly of the mouse using a flat applicator.
  • the thickness of the Carageenan-injected foot and left rear foot (non-injected control foot) is measured with a constant pressure vernier calliper at regular timepoints for a total of 14 days.
  • topical compositions are anticipated to be active in reducing inflammation within the right rear foot.
  • a chronic model of inflammation such as the collagen joint model could be used to test the efficacy of a topical composition of the present invention containing one or more anti- inflammatory molecules.
  • Expected results would, in that case, be similar to those anticipated for an acute inflammatory model such as the Carrageenan model.
  • the individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.
  • the clinical trial is subjected to rigorous controls to ensure that individuals are not unnecessarily put at risk and that they are fully informed about their role in the study.
  • volunteers are randomly assigned to topical placebo or topical treatment groups.
  • the doctors from being biased in treatments, they are not informed as to whether the medication they are administering is the topical pharmaceutical composition or topical placebo. Using this randomization approach, each volunteer has the same chance of being given either the new treatment or the placebo.
  • Volunteers receive either a topical pha ⁇ naceutical composition, for example, one of those described above in Examples 3 or 7 or topical placebo for an appropriate period, for example, a single 0.8ml application on one psoriatic lesion (total skin area of 20 cm2) with a 7 day follow-up or, alternatively, multiple 0.8ml applications on the same target lesion 9 times (every second day) over a 21 day period.
  • Biological parameters associated with the indicated disease state or condition for example, psoriasis, will be measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period. Such measurements include the levels of TNF alpha in body fluids, tissues or organs compared to pre-treatment levels.
  • measurements include, but are not limited to, indices of the disease state or condition being treated, body weight, blood pressure, serum titers of pharmacologic indicators of disease or toxicity as well as ADME (absorption, distribution, metabolism and excretion) measurements.
  • ADME absorption, distribution, metabolism and excretion
  • Information recorded for each patient includes age (years), gender, height (cm), family history of disease state or condition (yes/no), motivation rating (some/moderate/great) and number and type of previous treatment regimens for the indicated disease or condition.
  • Volunteers taking part in this study are adults aged 18 to 65 years and exhibit a particular dermalogical disorder, for example, psoriatic skin lesions. Additionally, roughly an equal number of males and females participate in the study. Volunteers with certain characteristics are equally distributed for topical placebo and topical pharmaceutical composition.
  • Evaluation of treatment is graded by an appropriate method, for example, the grading of treatment into one of four categories, namely, cured, obviously effective, effective and non-effective.
  • Cured is where the inflammatory area on the plaque is diminished completely and the pruritus disappeared.
  • Obviously effective is where the inflammatory area on the plaque is diminished by more than 60% and the pruritus is slighted and softened.
  • Effective is where the inflammatory area on the plaque is diminished by 20 to 60% and the pruritus is slighted and softened.
  • “Non-effective” is where the inflammatory area on the plaque is diminished by less than 20 % or there is exacerbation of psoriasis.
  • treatment evaluation is graded by the Local Plaque Severity Index (LPSI), whereby each target plaque is assessed and rated for erthema, induration and dessquamation using a five-point scale by the supervising clinician at the time of the specified clinic visits.
  • LPSI Local Plaque Severity Index
  • An example of an appropriate clinical visit timetable for the "multiple application" treatment regime is on days 0, 11 and 21.
  • the volunteers treated with topical placebo have little or no response to treatment, whereas the volunteers treated with a topical pharmaceutical composition of the present invention show positive trends in their disease state or condition index at the conclusion of the study.
  • the topical pharmaceutical composition of the present invention is obviously effective on most patients in the treatment group. No visible side-effects are observed.
  • Microemulsions for use as topical vaccines include a proteinaceous or non-proteinaceous antigen with or without an adjuvant.
  • ten mice per group are vaccinated with tenanus toxoid (TT; 1.8 limit of flocculation units, 20 meg; # 832-081, CSL, Melbourne, Australia) either by injection of 0.05 ml into the quadriceps muscle with a 30 gauge needle or topical application of 0.04 ml of microemulsion containing TT, using microemulsion formulation methods as described above in the examples 1 to 5, to a 2 cm 2 patch of shaved skin on the belly.
  • the control group receives a mock vaccination of saline alone.
  • mice Identical control and test groups are used in both the C57Black and BALB/C strains of mice. Mice are vaccinated every two weeks for a total of three vaccinations. Blood samples are taken by orbital bleeds 2 weeks post-vaccination. Indirect ELISA assay is used to determine the titre of serum IgG specific to tetanus toxoid using an anti-mouse whole IgG antibody alkaline phosphatase conjugate (Sigma Cat No. A3562). Cell mediated immunity is measured by the delayed type hypersensitivity reaction.
  • the right footpad of vaccinated or control mice are injected with 10 meg of tetanus toxoid in 0.05 ml of saline and the left footpad is injected with saline only as a control.
  • the increase in footpad swelling of the right footpad is measured 24, 48 and 72 hours post-injection using a caliper.
  • a microemulsion of the present invention contains one or more adjuvants selected from the following list of protein adjuvants selected from the list of: muramyl dipeptide, recombinant human C-type lectins such as CD209L, CD209L-Fc, DC-SIGN, DC-SIGN-Fc, CLEC4K and CD 207.
  • adjuvants are mixed at a suitable ratio, for example, a range of 1:1 to 1:5: of antigen : adjuvant protein.
  • a microemulsion for topical weight loss is prepared according to the methods as described above in the microemulsion formulation examples 1-5, wherein the active agents are selected from the list of: insulin, insulin-like growth factor 1 (IGFl), GHRP-6 at a final concentration range of 0.5 to 10 mg/ml.
  • a standardised volume of microemulsion is applied daily (TD) to an area of shaved skin on the belly of conscious Swiss strain mice.
  • a standardised volume of each of insulin, or insulin-like growth factor 1 (IGFl) or GHRP-6 at a final concentration range of 0.5 to 10 mg/ml in saline is injected sub cutaneously (SC) into the belly region of Swiss strain mice. Animal weights are measured 3 times per week.
  • mice Following 2 weeks of treatment the mice are euthanased and their tissues removed and weighed. The weight of the animals with time is plotted was and a decrease in body weight of animals treated with the topical insulin is anticipated. Examination of organ weights is expected to show the greatest change in the topically applied insulin group, where the weight of skin is significantly reduced whilst muscle mass increases.
  • mice (Balb/C and C57/B1) are injected intradermally or sub-cutaneously with 1 x 10 6 cells of the Bl 6 melanoma line. The tumour is allowed to grow until it is approximately 0.5 gm in mass. The mice are then treated topically with in a microemulsion, for example, a microemulsion as described above in the microemulsion formulation examples 1 to 5 wherein the active agents include one or more of the following chemotherapeutic agents: 5 -fluorouracil (10 mg/ml, 20 microlitre dose, 2 times per day); Carboplatin (10 mg/ml, 20 microlitre dose, 2 times per day); Cis-Platinum (10 mg/ml, 20 microlitre dose, 2 times per day); or a control microemulsion without chemotherapeutic agents. After 4 days of treatment, there is anticipated to be a reduction in growth rate of the tumour but no reduction in body weight of the animals. EXAMPLE 13
  • the individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.
  • the trial is conducted in a double-blinded fashion. Volunteer patients at a tertiary care center are randomly assigned to placebo or treatment groups. Furthermore, to prevent the doctors from being biased in treatments, they are not informed as to whether the medication they are administering is the topical pharmaceutical composition of the present invention or a placebo composition. Using this randomization approach, each volunteer has the same chance of being given either the new treatment or the placebo treatment.
  • Volunteers presenting with, for example, cutaneous melanoma receive either the topical pharmaceutical composition of the present invention, for example, as described above in the microemulsion formulation examples 1 to 5 wherein the active agents include one or more of the following anti-cancer agents: 5-fluorouracil, carboplatin, oxaliplatin, cis-platin, selenomethionine, chlorotoxin, L49 monoclonal antibody, MAb-LM609, antileukinate, gold-monophosphine and gold-diphosphine, pyridoxal isonicotinoyl hydrazone (PIH), amino-substituted platinum complexes, paclitaxol, defoxamine, synthetic retinoids such as tretinoin, isotretinoin, and etretinate, imiquimod (e.g.
  • the active agents include one or more of the following anti-cancer agents: 5-fluorouracil, carboplatin, oxaliplatin
  • Such measurements include the levels of each pharmaceutical agent in the composition in body fluids, tissues or organs compared to pre-treatment levels. Other measurements include, but are not limited to, evidence of metastisis in tissues such as lymph nodes. Still other measurements include, but are not limited to body weight, blood pressure, serum titers of pharmacologic indicators of disease such as specific disease indicators or toxicity as well as ADME (absorption, distribution, metabolism and excretion) measurements.
  • Information recorded for each patient includes age (years), gender, height (cm), family history of disease state or condition (yes/no), motivation rating (some/moderate/great) and number and type of previous cancer treatment regimens.
  • Recent methods indicate that cancerous disease states associated with the expression of abnormal common fusion genes can be treated by the specific silencing of those genes via the administration of siRNA (Hashimoto et al. PNAS 101(17): 6647-6652, 2004).
  • the individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.
  • the trial is conducted in a double-blinded fashion. Volunteers presenting with a cancerous condition, for example, chronic myeloid leukemia, are randomly assigned to placebo or treatment groups. Furthermore, to prevent the doctors from being biased in treatments, they are not informed as to whether the medication they are administering is the topical pharmaceutical composition of the present invention or a topical placebo. Using this randomization approach, each volunteer has the same chance of being given either the new treatment or the placebo.
  • a cancerous condition for example, chronic myeloid leukemia
  • Volunteers receive either the topical pharmaceutical composition of the present invention, for example, a microemulsion as described above in the microemulsion formulation Examples 1 to 5 , wherein the active agents consist of siRNAs directed against one or more of the two types of the common fusion genes present in chronic myeloid leukemia patients (Hashimoto et ah, supra) or placebo for an appropriate period with biological parameters associated with the indicated disease state being measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period. Such measurements include the levels of pharmaceutical agent in the composition in body fluids, tissues or organs compared to pre-treatment levels.
  • the active agents consist of siRNAs directed against one or more of the two types of the common fusion genes present in chronic myeloid leukemia patients (Hashimoto et ah, supra) or placebo for an appropriate period with biological parameters associated with the indicated disease state being measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period.
  • Other measurements include, but are not limited to, counts of abnormal white cells, other specific tests for detecting trace leukemia cells, body weight, blood pressure, serum titers of pharmacologic indicators of disease such as specific disease indicators or toxicity as well as ADME (absorption, distribution, metabolism and excretion) measurements.
  • ADME absorption, distribution, metabolism and excretion
  • Information recorded for each patient includes age (years), gender, height (cm), family history of disease state or condition (yes/no), motivation rating (some/moderate/great) and number and type of previous treatment regimens for the indicated disease or condition.
  • Volunteers taking part in this study are adults aged 18 to 65 years and roughly an equal number of males and females participate in the study. Volunteers with certain characteristics are equally distributed for placebo and the topical pharmaceutical composition of the present invention treatment. In general, the volunteers treated with placebo have little or no response to treatment, whereas the volunteers treated with the topical pharmaceutical composition of the present invention show positive trends in one or more of the following parameters: decreased counts of abnormal white cells, overall survival (OS), disease-free survival (DFS) at the conclusion of the study.
  • OS overall survival
  • DFS disease-free survival
  • the individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.
  • the trial is conducted in a double-blinded fashion. Volunteers presenting with either neuropathic or nociceptic pain are randomly assigned to placebo or treatment groups. Furthermore, to prevent the doctors from being biased in treatments, they are not informed as to whether the medication they are administering is the topical pharmaceutical composition of the present invention or a topical placebo. Using this randomization approach, each volunteer has the same chance of being given either the new treatment or the placebo.
  • Volunteers receive either the topical pharmaceutical composition of the present invention, for example, a microemulsion as described above in the formulation examples 1 to 5 , wherein the active agents consist of one or more known anti-inflammatory molecules, optionally containing one or more analgesic agents such as lidocaine (4%) or placebo for an appropriate period with biological parameters associated with the indicated disease state or condition being measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period.
  • the active agents consist of one or more known anti-inflammatory molecules, optionally containing one or more analgesic agents such as lidocaine (4%) or placebo for an appropriate period with biological parameters associated with the indicated disease state or condition being measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period.
  • Such measurements include the levels of each pharmaceutical agent in the composition in body fluids, tissues or organs compared to pre-treatment levels.
  • Other measurements include, but are not limited to, indices of the disease state or condition being treated, body weight, blood pressure, serum titers of pharmacologic indicators of disease such as specific disease indicators or toxicity as well as ADME (absorption, distribution, metabolism and excretion) measurements.
  • Information recorded for each patient includes age (years), gender, height (cm), family history of disease state or condition (yes/no), motivation rating (some/moderate/great) and number and type of previous treatment regimens for the indicated disease or condition.
  • Volunteers taking part in this study are adults aged 18 to 65 years and roughly an equal number of males and females participate in the study. Volunteers with certain characteristics are equally distributed for placebo and the topical pharmaceutical composition of the present invention treatment. In general, the volunteers treated with placebo have little or no response to treatment, whereas the volunteers treated with the topical pharmaceutical composition of the present invention show an amelioration of pain intensity at the conclusion of the study.
  • the individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.
  • the trial is conducted in a double-blinded fashion. Volunteers are randomly assigned to placebo or treatment groups. Furthermore, to prevent the participating clinicians from being biased in treatments, they are not informed as to whether the treatment being administering is the topical pharmaceutical composition of the present invention or a topical placebo composition. Using this randomization approach, each volunteer has the same chance of being given either the new treatment or the placebo.
  • Volunteers take part in a suitable exercise protocol to induce lower limb fatigue, for example, repetitive sets of jumping movements on an inclined sled apparatus on three separate days with a minimum of one week rest between each test session (presentation by S. Hughes at Australian Conference of Science and Medicine in Sport, Yanuca, Fiji, October, 2006).
  • Volunteers receive a controlled application of either the topical pharmaceutical composition of the present invention, for example, a microemulsion as described above in the formulation examples 1 to 5 , wherein the active agents are one or more anti- inflammatory moleculesor topical placebo composition applied to the lower limbs at regular time-points after the initial exercise protocol for a total of 1-14 days.
  • the topical pharmaceutical composition of the present invention for example, a microemulsion as described above in the formulation examples 1 to 5 , wherein the active agents are one or more anti- inflammatory moleculesor topical placebo composition applied to the lower limbs at regular time-points after the initial exercise protocol for a total of 1-14 days.
  • Suitable physical and biological parameters associated with DOMS are measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period. Suitable physical parameters include the level of performance between initial and follow-up test sessions of the exercise protocol. Suitable biological parameters include the levels of pharmaceutical agent or agents of the composition in body fluids, tissues or organs compared to pre-treatment levels. Other measurements include, but are not limited to, blood lactate levels, body weight, blood pressure, serum titers of pharmacologic indicators of DOMS such as creatinine levels or toxicity as well as ADME (absorption, distribution, metabolism and excretion) measurements.
  • ADME absorption, distribution, metabolism and excretion
  • Information recorded for each patient includes age (years), gender, height (cm), family history of disease state or condition (yes/no), motivation rating (some/moderate/great) and number and type of previous treatment regimens for DOMS.
  • Volunteers taking part in this study are adults aged 18 to 65 years and roughly an equal number of males and females participate in the study. Volunteers with certain characteristics are equally distributed for topical placebo composition and topical pharmaceutical composition of the present invention treatments.
  • Volunteers treated with the topical pharmaceutical composition of the present invention show positive trends in one or more of the above-mentioned physical or biological parameters in comparison to volunteers treated with the topical placebo composition.
  • the individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.
  • the trial is conducted in a double-blinded fashion. Volunteer patients at a tertiary care center are randomly assigned to placebo or treatment groups. Furthermore, to prevent the doctors from being biased in treatments, they are not informed as to whether the medication they are administering is the topical pharmaceutical composition of the present invention or a placebo composition. Using this randomization approach, each volunteer has the same chance of being given either the new treatment or the placebo treatment.
  • Volunteers presenting with a tumour for example, a cutaneous melanoma, receive either the topical pharmaceutical composition of the present invention, for example, a microemulsion as described above in the microemulsion formulation Examples 1 to 5 wherein the active agent is an imaging agent is a known imaging compound such as 99m Tc chelated to DTPA, or topical placebo composition, applied to the tumour.
  • the body region is imaged for the detection of imaging agent. It is anticipated that the accumulation of imaging agent within the sentinel lymph node will allow for a rapid identification and subsequent removal of the lymph node.
  • Other measurements include, but are not limited to body weight, blood pressure, serum titers of pharmacologic indicators of disease such as specific disease indicators or toxicity as well as ADME (absorption, distribution, metabolism and excretion) measurements.
  • Information recorded for each patient includes age (years), gender, height (cm), family history of disease state or condition (yes/no), motivation rating (some/moderate/great) and number and type of previous cancer treatment regimens.
  • Volunteers taking part in this study are adults aged 18 to 65 years. Volunteers with certain characteristics are equally distributed for topical placebo treatment and the topical pharmaceutical composition of the present invention treatment.
  • the volunteers treated with the topical pharmaceutical composition of the present invention when compared with the volunteers topically treated with the placebo composition, show positive trends in one or more of the following parameters: incidence of metastisis, overall survival (OS), disease-free survival (DFS).
  • OS overall survival
  • DFS disease-free survival
  • the individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.
  • the trial is conducted in a double-blinded fashion. Volunteers are randomly assigned to placebo or treatment groups. Furthermore, to prevent the doctors from being biased in treatments, they are not informed as to whether the medication they are administering is the topical pharmaceutical composition of the present invention, or a placebo. Using this randomization approach, each volunteer has the same chance of being given either the new treatment or the placebo.
  • Volunteers receive either the topical pharmaceutical composition of the present invention, for example, a microemulsion as described above in the microemulsion formulation examples 1 to 5 wherein the active agents include one or more of the following: insulin
  • Suitable physical measurements include skinfold caliper measurements or A- and/or B-mode ultrasound measurements of the thickness of the subcutaneous fat layer overlying the upper arm.
  • Suitable biological measurements include blood levels of insulin and glucose in body fluids, tissues or organs compared to pre-treatment levels. Other measurements include, but are not limited to, body weight and blood pressure as well as ADME (absorption, distribution, metabolism and excretion) measurements.
  • Information recorded for each patient includes age (years), gender, height (cm), family history of condition (yes/no), motivation rating (some/moderate/great) and number and type of previous treatment regimens for the indicated condition.
  • Volunteers taking part in this study are adults aged 18 to 65 years and roughly an equal number of males and females participate in the study. Volunteers with certain characteristics are equally distributed for placebo and the topical pharmaceutical composition of the present invention treatment.
  • the volunteers treated with the topical pharmaceutical composition of the present invention show reduced thickness of sub cutaneous fat deposits of the upper arm, as determined by one or more of the following methods: skinfold caliper measuremen, A-mode ultrasound measurement, B-mode ultrasound measurement; whereas the volunteers treated with placebo have little or no response to treatment. It is anticipated that blood levels of insulin, insulin-like growth factor 1 (IGFl) or GHRP-6 are not affected by treatment with the topical pharmaceutical composition of the present invention.
  • IGFl insulin-like growth factor 1
  • GHRP-6 are not affected by treatment with the topical pharmaceutical composition of the present invention.
  • the individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which include humans.
  • a standardized volume of a topical formulation of the present invention selected from Examples 4 or 5 or a placebo formulation is applied to an appropriate area of the body, for example, the forehead or the back of the hand of volunteers in a prescribed fashion for an appropriate period, for example, 6 weeks with biological parameters associated with each skin area being measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period.
  • Such measurements include :
  • Leica Stereo Explorer (5) the measurement of skin elasticity using a suitable instrument (for example, a Cutometer SEM 575 skin elasticity meter).
  • Information recorded for each patient includes age (years), gender, motivation rating (some/moderate/great) and number and type of previous treatment skin regimens.
  • Volunteers taking part in this study are adults aged 18 to 65 years. Volunteers treated with a topical formulation of the present invention show positive "anti-ageing" parameters in one or more physical or biological parameters, including skin capacitance, skin conductance, and parameters associated with skin surface topography such as wrinkle frequency, wrinkle depth, roughness and smoothing, and altered “skin-lightening” parameters such as skin color, skin brightness, time taken for pigment fade, at the conclusion of the study compared with the baseline measurements taken at the commencement of the study.
  • positive "anti-ageing" parameters in one or more physical or biological parameters, including skin capacitance, skin conductance, and parameters associated with skin surface topography such as wrinkle frequency, wrinkle depth, roughness and smoothing, and altered “skin-lightening” parameters such as skin color, skin brightness, time taken for pigment fade, at the conclusion of the study compared with the baseline measurements taken at the commencement of the study.
  • the individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.
  • the trial is conducted in a double-blinded fashion. Volunteers are randomly assigned to placebo or treatment groups. Furthermore, to prevent the researchers from being biased in treatments, they are not informed as to whether the treatments they are administering is a pharmaceutical composition of the present invention or a placebo formulation. Using this randomization approach, each volunteer has the same chance of being given either the topical formulation of the present invention or the placebo formulation.
  • a standardized volume of either the topical formulation of the present invention selected from Examples 3, 4 or 5 or a placebo formulation is applied to the face of volunteers in a prescribed fashion for an appropriate period, for example, 6 weeks with biological parameters associated with the facial skin being measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period.
  • biological parameters associated with the facial skin being measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period.
  • Such measurements include:
  • a suitable instrument for example, a Cutometer SEM 575 skin elasticity meter.
  • a suitable instrument for example, a Cutometer SEM 575 skin elasticity meter.
  • a suitable instruments for example, a Minolta skin analysis camera, software analysis and standardized color palettes (for example, the Pantone [Registered Trade Mark] system).
  • Information recorded for each patient includes age (years), gender, motivation rating (some/moderate/great) and number and type of previous treatment regimens for facial skin.
  • Volunteers taking part in this study are adults aged 18 to 65 years. Volunteers with certain characteristics are equally distributed for the topical formulation of the present invention and placebo formulation treatment.
  • the volunteers treated with placebo formulation have little or no improvement in one or more physical or biological parameters, including facial skin capacitance, facial skin conductance, parameters associated with skin surface topography such as wrinkle frequency, wrinkle depth, roughness and smoothing, and altered "skin-lightening" parameters such as skin color, skin brightness, time taken for pigment fade, whereas the volunteers treated with a topical formulation of the present invention show positive "anti-ageing" parameters in one or more of the above-mentioned physical or biological parameters at the conclusion of the study.
  • the water-in-oil microemulsion is used to formulate agents to treat erectile dysfunction.
  • the microemulsion comprises a glycerol trinitrate and sildenafil citrate.
  • the agent relaxes the smooth muscle in the corpus cavernosum of the penis.
  • the agents contemplated therein include 1) nitric oxide donors which includes nitroglycerin, alkyl nitrites, such as amyl nitrite and L-arginine; 2) The phosphodiesterase inhibitors sildenafil (Viagra), Tadalafil (Cialis), Vardenafil (Levitra) and any other cGMP specific phosphodiesterase type 5 inhibitors; and 3) The adenyl cyclase activators, prostaglandin El (Alprostidil) and forskolin.

Abstract

The present invention generally relates to a vehicle useful for delivering a pharmaceutically active compound transdermally to a subject. More particularly, the present invention provides microemulsions for transdermal delivery of active agents to a subject.

Description

TRANSDERMAL DELIVERY OF ACTIVE AGENTS
FIELD
The present invention generally relates to a vehicle useful for delivering a pharmaceutically active compound transdermally to a subject. More particularly, the present invention provides microemulsions for transdermal delivery of active agents to a subject.
BACKGROUND
Bibliographic details of references provided in the subject specification are listed at the end of the specification.
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 any country.
It is generally accepted in the art that delivery of therapeutic quantities of water-soluble macromolecules such as peptides and proteins or other such pharmaceutical agents across the skin is extremely difficult, as the skin functions as a barrier to prevent transdermal penetration of most water-soluble molecules.
However, transdermal delivery is a highly desired method by which therapeutic amounts of peptides, proteins or other pharmaceutical agents could be administered particularly for local and/or systemic therapy. This is because biologically-active macromolecules, such as certain peptides or proteins, generally have low oral bioavailability, making oral administration difficult, and often have short biological half-lives, making parenteral delivery impractical outside a hospital setting (Shahrokh et al. American Chemical Society, 1997). Additionally, there are many medical conditions and diseases of the skin, which would greatly benefit from direct local therapy, without exposing the other organs of the body to the deleterious side-effects of delivery via other routes. In this regard various pharmaceutical agents, such as non-steroidal anti-inflammatory drugs (NSAIDs), have been shown to have considerable gastric toxicity, thereby reducing their effectiveness. Additional side effects of NSAIDs include renal insufficiency and failure, gastrointestinal ulceration, bleeding or perforation, exacerbation of hypertension and congestive heart failure.
Delivery of therapeutic pharmaceutical agents across the skin addresses the above- mentioned problems by offering a number of potential advantages compared to conventional methods, such as pills and injections, including: (1) little or no degradation or modification of the pharmaceutical agents such as digestion of the proteins by the enzymes and surfactants resident in the stomach and small intestine; (2) an increase in drug effectiveness as the transdermal route avoids first pass metabolism by the liver; (3) minimization of gastrointestinal and other side effects caused by ingestion of the pharmaceutical agent; (4) an improvement in patient comfort and compliance due to a more user-friendly delivery method; and (5) the potential for prolonged and controlled drug delivery.
Current research into transdermal transport of water-soluble molecules focuses on: (1) using chemical enhancers to alter the skin's lipid environment; (2) using liposomes to facilitate transdermal transport; (3) using iontophoresis to provide an electrical driving force for transdermal transport; (4) using electroporation to create a new transdermal pathway; and (5) using ultrasound to create a new transdermal pathway. Although chemical enhancers have been shown to increase transdermal transport of small compounds, they are often shown to cause significant skin irritation and may affect drug stability (Santus et al. J Control Release 25:1-20, 1993). Introduction of electrical or ultrasonic energy into skin alters its properties as a consequence of the high energy input and issues of safety and drug stability via this method need to be further investigated.
Emulsions have been proposed as a means to enhance transdermal uptake of a small number of hydrophobic molecules. Generally, an emulsion is a composition comprising more than one phase where at least one of the phases consists of a finely divided phase domain (such as, for example, particles or droplets) distributed throughout a continuous phase domain. The finely divided domains are generally referred to as dispersed or discontinuous phase domains. A microemulsion generally is a system comprising an oil phase, a water phase and a surfactant. Some microemulsions are quaternary systems and further comprise a co- surfactant or a co-solvent. Microemulsions differ from ordinary emulsions (or macroemulsions) in that they are thermodynamically stable and form spontaneously upon mixing. The average drop size of ordinary emulsions grows continuously with time so that phase separation ultimately occurs under gravitational force, i.e. they are thermodynamically unstable and their formation requires input of work. In addition, the drops of the dispersed phase are generally large (> 0.2μm) so that ordinary emulsions often take on a milky opaque appearance. In contrast, microemulsions have an observable transparency, which is due to the fact that the maximum size of the droplets of the dispersed phase is not larger than one-half of the wavelength of visible light. On average, the droplet diameter in stable microemulsions is usually within the range of 5-200 nm. Microemulsions also possess specific physicochemical properties such as optical isotropy, low viscosity and thermodynamic stability.
Sintov et al. (Journal of Controlled Release 95: 173-183, 2004) describe a system with formulations that contain isopropyl palmitate, water, tetraglycol (as the co-surfactant) and a combination of glyceryl oleate (HLB = 2.8) and PEG-40 hydrogenated castor oil (HLB = ca. 15) at a ratio of 1 :2 This microemulsion system was loaded with 2.5% lidocaine (a highly lipophilic molecule of low molecular weight) as base or as a hydrochloride salt.
In addition, Kreilgaard et al {Journal of Controlled Release 69:421-433, 2000) compared microemulsions: (i) Labrasol (a mixture consisting of 30% mono-, di and triglycerides of C and C fatty acids, 50% of the ap- mono- and di-esters of poly(ethylene glycol) (PEG 400) and 20% of free PEG 400); (ii) Plurol Isostearique (isostearic acid ester of polyglycerol, containing 30-35% of diglycerol, 20-25% of triglycerol, 15-20% tetraglycerol, and 10% of pentaglycerol and higher oligomers); (iii) isostearylic isostearate (92% purity); and (iv) water on their transdermal delivery potential of a lipophilic (lidocaine) and a low molecular weight hydrophilic model drug (prilocaine hydrochloride). The microemulsions were applied to excised rat skin in a Franz-type diffusion cell and were found to increase transdermal flux of lidocaine through the excised rat skin by up to four times compared to a conventional oil-in- water emulsion, and that of prilocaine hydrochloride by almost 10 times compared to a hydrogel.
Li et al. (International Journal of Pharmaceutics 237:77-85, 2002) examined the use of a microemulsion for intranasal delivery of the hydrophobic drug diazepam. Diazepam, a practically water-insoluble drug, displayed a high solubility of 41 mg/ml in a microemulsion consisting of 15% ethyl laurate, 15% H2O, and 70% (w/w) surfactant/co- surfactant (Tween 80:propylene glycolethanol at 1 :1 :1 weight ratio). The study demonstrated an intranasal bioavailability of 50%.
In a study reported by Valenta et al. (Journal of Controlled Release 95:257- 265, 2004), the ability of three different microemulsions (A: 2.5 g Brij 97, 7.2 g water and 0.34 g tributyrin; B: 1.87 Brij 97, 5.17 g water and 0.45 miglyol; and C: 2.5 g Brij 97, 6.62 g water and 0.78 g soybean oil) to increase the permeability of the hydrophobic compound sodium fluorescein through porcine skin was examined.
Escribano et al. (European Journal of Pharmaceutical Sciences 7P/203-210, 2003) studied the use of microemulsion formed with 19% Transcutanol, 19.5% Plurol oleique, 30.6% water, 10.9% Isostearol isostearate, Labrasol 19% for transdermal delivery of the low molecular weight chemical diclofenac across human skin. The microemulsion was found to be less efficient than a preparation made with the absorption enhancers oleic acid and d- limonene.
However, these microemulsions have not been demonstrated to carry macromolecules, such as certain peptides or proteins, water soluble vitamins, NSAIDs and chemical compounds across the dermis. In addition, it has not been possible to generate microemulsions which contain high concentrations of active ingredients. Accordingly, there is a need to develop formulations for transdermal delivery of active molecules. Further, there is a need to develop formulations which can carry macromolecules and other water soluble compounds at a range of concentrations. SUMMARY
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.
The present invention provides a topical delivery system which assists in the penetration of a desired active agent, such as peptides, polypeptides and proteins, water soluble vitamins, NSAIDs, genetic molecules and chemical compounds into the skin, which does not necessitate the use of chemical enhancers, nor electrical or ultrasonic energy to facilitate penetration into and though the skin. Furthermore, the delivery system is easy to apply to small or large areas of the skin.
The present invention further enables the use of hydrophilic molecules which are incorporated within the submicron sized water droplets contained within the microemulsion, thereby enhancing transdermal delivery of desired active agents.
The topical delivery system of the present invention is in the form of a microemulsion. The subject microemulsions for transdermal delivery of peptides, polypeptides and proteins or other molecules such as water soluble vitamins, NSAIDs, genetic molecules and chemical compounds are able to be formed with little or no shear force applied to the molecules during the preparation of the microemulsion, which aids in the preservation of the structure and bioactivity of these molecules. The microemulsion also provides both a hydrophilic and hydrophobic environment, which can aid in the solubilization of various molecules and which additionally provides the ability to mix hydrophobic molecules and hydrophilic molecules within the same formulation. Additionally, the microemulsions of the present invention provide an environment which allows for the solubilization of various water- soluble molecules at a wide range of concentrations. Hence, in one aspect, the present invention provides a microemulsion comprising a water phase; an oil phase; an active ingredient; one or more zwitterionic surfactants; and one or more co-surfactants.
In another aspect, the present invention provides a microemulsion which is capable of transdermal delivery of active agents. The present invention further extends to a pharmaceutical composition comprising the microemulsion and an active agent wherein the pharmaceutical composition is suitable for topical application on a subject.
The present invention further provides a method suitable for the transdermal delivery of active agents in a range of therapeutic, cosmetic and research applications. The method comprises (a) preparing a water phase, including dissolving one or more selected active agents in water; (b) preparing an oil phase; (c) emulsifying the water phase and the oil phase using one or more zwitterionic surfactants and one or more co-surfactants, thereby forming a topical pharmaceutical composition; and (d) applying the topical pharmaceutical composition on a subject.
The microemulsion of the present invention is useful inter alia for the topical delivery of anti-inflammatory agents, such as small molecules, proteins, antibodies, Fc fusion molecules and soluble receptors; vaccines; imaging agents; anti-cancer agents, particularly anti-melanoma agents; skin lightening including whitening agents; UV blocking agents, agents to reduce or nullify nitric oxide, agents useful for the treatment of erectile dysfunction; agents for increasing local circulation, agents for the amelioration of facial and skin ageing, agents for the treatment of obesity, re-pigmentation agents; genetic molecules such as RNAi, DNA, virosomes and anti-sense RNA; and agents useful in treating muscle dystrophy. The microemulsions have application in human and veterinary medicine as well as for agricultural animals.
A list of agents contemplated for use in the microemulsions includes but is not limited to the list provided in Table 1. TABLE 1
Agents
Figure imgf000008_0001
Figure imgf000009_0001
Figure imgf000010_0001
Figure imgf000011_0001
Figure imgf000012_0001
-12-
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
Figure imgf000017_0001
Agents Description and/or Alternate names/ or alternative drugs
VEGF Vascular endothelial growth factor
PDE5 inhibitors phosphodiesterases type 5 inhibitors; sildenafil, tadalafil, vardenafil
Vitamin B12 See AdeCbl derivatives
Weight Modifying Insulin drugs
DETAILED DESCRIPTION
It is to be understood that unless otherwise indicated, the subject invention is not limited to specific formulations, manufacturing methods, diagnostic methods, assay protocols, nutritional protocols, or research protocols or the like as such may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.
It must be noted that, as used in the subject specification, the singular forms "a", "an" and "the" include plural aspects unless the context already dictates otherwise. Thus, for example, reference to "a pharmaceutically active agent" includes a single pharmaceutically active agent as well as two or more pharmaceutically active agents, "an agent" includes a single agent as well as two or more agents; reference to "the microemulsion" includes one or more microemulsions; and so forth.
The terms "compound", "agent", "active agent", "chemical agent", "pharmaceutical agent", "pharmacologically active agent", "medicament", "active" and "drug" are used interchangeably herein to refer to a chemical compound and in particular a protein or a chimeric molecule thereof that induces a desired pharmacological and/or physiological effect. The terms also encompass pharmaceutically acceptable and pharmacologically active ingredients of those active agents specifically mentioned herein including but not limited to salts, esters, amides, prodrugs, active metabolites, analogs and the like. When the terms "compound", "agent", "active agent", "chemical agent", "pharmaceutical agent", "pharmacologically active agent", "medicament", "active" and "drug" are used, then it is to be understood that this includes the active agent per se as well as pharmaceutically acceptable, pharmacologically active salts, esters, amides, prodrugs, metabolites, analogs, etc. The desired pharmacological effect is dependent on the agent used but includes an antiinflammatory effect, an immune response, a change in the level of gene expression, a pigmentation effect including a lightening effect, an imaging effect on a target such as a target lymph node, an anti-muscle dystrophic effect, weight loss, treatment of cancer, reduction in pain or a physiological effect such as correction of erectile dysfunction. Reference to a "compound", "agent", "active agent", "chemical agent", "pharmaceutical agent", "pharmacologically active agent", "medicament", "active" and "drug" includes combinations of two or more active agents such as two or more proteins. A "combination" also includes multi-part such as a two-part composition where the agents are provided separately and given or dispensed separately or admixed together prior to dispensation.
The terms "agents" and the like include peptides, polypeptides and proteins, large and small chemical molecules, hormones, steroids and nucleic acid molecules as well as viral components such as virosomes.
The terms "effective amount" and "therapeutically effective amount" of an agent as used herein mean a sufficient amount of the pharmaceutical agent, alone or in combination with other agents to provide the desired therapeutic or physiological effect or outcome. Undesirable effects, e.g. side effects, are sometimes manifested along with the desired therapeutic effect; hence, a practitioner balances the potential benefits against the potential risks in determining what is an appropriate "effective amount". The exact amount required will vary from subject to subject, depending on the species, age and general condition of the subject, mode of administration and the like. Thus, it may not be possible to specify an exact "effective amount". However, an appropriate "effective amount" in any individual case may be determined by one of ordinary skill in the art using only routine experimentation.
An "effective amount", for example, would include an amount of a cytokine or anti- cytokine antibody or anti-sense or RNAi molecule directed to a gene or mRNA transcript encoding an inflammatory cytokine to inhibit an inflammatory response by a reduction in aberrant skin proliferation, antibody production, cytokine production and/or immune response effector molecule production.
By "pharmaceutically acceptable" carrier, excipient or diluent is meant a pharmaceutical vehicle comprised of a material that is not biologically or otherwise undesirable, i.e. the material may be administered to a subject along with the selected active agent without causing any or a substantial adverse reaction. Carriers may include excipients and other additives such as diluents, detergents, coloring agents, wetting or emulsifying agents, pH buffering agents, preservatives, and the like.
Similarly, a "pharmacologically acceptable" salt, ester, amide, prodrug or derivative of a compound as provided herein is a salt, ester, amide, prodrug or derivative that this not biologically or otherwise undesirable.
The terms "treating" and "treatment" as used herein refer to reduction in severity and/or frequency of symptoms of the condition being treated, elimination of symptoms and/or the underlying cause, prevention of the occurrence of symptoms of the condition and/or their underlying cause and improvement or remediation or amelioration of damage following a condition.
"Treating" a subject may involve prevention of a condition or other adverse physiological event in a susceptible individual as well as treatment of a clinically symptomatic individual by ameliorating the symptoms of the condition.
A "subject" as used herein refers to an animal, in a particular embodiment, a mammal and in a further embodiment human who can benefit from the pharmaceutical formulations and methods of the present invention. There is no limitation on the type of animal that could benefit from the presently described pharmaceutical formulations and methods. A subject regardless of whether a human or non-human animal may be referred to as an individual, patient, animal, host or recipient. The compounds and methods of the present invention have applications in human medicine, veterinary medicine as well as in general, domestic or wild animal husbandry including agriculturally-relevant animals (e.g. livestock animals).
As indicated above, in a particular embodiment, the animals are humans or other primates such as orangutans, gorillas or marmosets, further contemplated are livestock animals, laboratory test animals, companion animals or captive wild animals, as well as avian species.
Examples of laboratory test animals include mice, rats, rabbits, guinea pigs and hamsters. Rabbits and rodent animals, such as rats and mice, provide a convenient test system or animal model. Livestock animals include sheep, cows, pigs, goats, horses, yaks, llamas, camels and donkeys. Companion animals include dogs and cats. Non-mammalian animals such as avian species, fish and amphibians.
The term "protein" is used interchangeably with the term "peptide" or "polypeptide" and is used in its most general sense including ligands and receptors. As used herein, reference to a particular protein should be understood to refer to the "complete" protein as well as fragments, isoforms, derivatives or homologs or chimeras thereof comprising one or more amino acid additions, deletions or substitutions, but which substantially retain the biological activity of the complete protein.
The term "polypeptide" refers to a polymer of amino acids and its equivalent but does not imply a limitation as to a specific length of the product, thus, peptides, oligopeptides, polypeptides and proteins are included within the definition of a "polypeptide". This term also includes all co- or post-translationally modified forms of a polypeptide. Also included within the definition are, for example, polypeptides containing one or more analogs of an amino acid including, unnatural amino acids or polypeptides with substituted linkages.
As used herein, the terms "co- or post-translational modifications" refer to covalent modifications occurred during or after translation of the peptide chain. Exemplary co- or post-translational modifications include, but are not limited, to acylation (including acetylation), amidation or deamidation, biotinylation, carbamylation (or carbamoylation), carboxylation or decarboxylation, disulfide bond formation, fatty acid acylation (including myristoylation, palmitoylation and stearoylation), formylation, glycation, glycosylation, hydroxylation, incorporation of selenocysteine, lipidation, lipoic acid addition, methylation, N- or C-terminal blocking, N- or C-terminal removal, nitration, oxidation of methionine, phosphorylation, proteolytic cleavage, prenylation (including farnesylation, geranyl geranylation), pyridoxal phosphate addition, sialylation or desialylation, sulfation, ubiquitinylation (or ubiquitination) or addition of ubiquitin-like proteins.
A "derivative" of a polypeptide of the present invention also encompasses a portion or a part of a full-length parent polypeptide, which retains partial transcriptional activity of the parent polypeptide and includes a variant. Such "biologically-active fragments" include deletion mutants and small peptides, for example, for at least 10, in a particular embodiment, at least 20 and in a further embodiment at least 30 contiguous amino acids, which exhibit the requisite activity. Peptides of this type may be obtained through the application of standard recombinant nucleic acid techniques or synthesized using conventional liquid or solid phase synthesis techniques. For example, reference may be made to solution synthesis or solid phase synthesis as described, for example, in Chapter 9 entitled "Peptide Synthesis" by Atherton and Shephard which is included in a publication entitled "Synthetic Vaccines" edited by Nicholson and published by Blackwell Scientific Publications. Alternatively, peptides can be produced by digestion of an amino acid sequence of the invention with proteinases such as endoLys-C, endoArg-C, endoGlu-C and staphylococcus V8-protease. The digested fragments can be purified by, for example, high performance liquid chromatographic (HPLC) techniques. Any such fragment, irrespective of its means of generation, is to be understood as being encompassed by the term "derivative" as used herein.
The active agent of the present invention may be a chimeric molecule comprising an isolated protein or a fragment thereof, such as an extra-cellular domain of a membrane bound protein, linked to the constant (Fc) or framework region of a human immunoglobulin via one or more protein linkers. Such a chimeric molecule is also referred to herein as protein-Fc. Other chimeric molecules contemplated by the present invention include the protein or protein-Fc or a fragment thereof, linked to a lipid moiety such as a polyunsaturated fatty acid molecule. Such lipid moieties may be linked to an amino acid residue in the backbone of the molecule or to a side chain of such an amino acid residue. The human immunoglobulin may be selected from IgGl, IgG2, IgG3, IgG4, IgAl, IgA2, IgM, IgE5 IgD. The active agent of the present invention may further include a chimeric molecule comprising an isolated protein or a fragment thereof, such as an extra-cellular domain of a membrane bound protein, linked to the constant (Fc) or framework region of a mammalian immunoglobulin via one or more protein linker. In another aspect, the mammal Fc or framework region of the immunoglobulin is derived from a mammal selected from the group consisting of primates, including humans, marmosets, orangutans and gorillas, livestock animals (e.g. goats, cows, sheep, pigs, horses, donkeys, asses), laboratory test animals (e.g. mice, rats, guinea pigs, hamsters, rabbits, companion animals (e.g. cats, dogs) and captured wild animals (e.g. rodents, foxes, deer, kangaroos). In another embodiment the Fc or framework region is a human immunoglobulin. In a particular embodiment the mammal is a human. Such a chimeric molecule is also referred to herein as protein-Fc. Other chimeric molecules contemplated by the present invention include the protein or protein-Fc or a fragment thereof linked to a lipid moiety such as a polyunsaturated fatty acid molecule. Such lipid moieties may be linked to an amino acid residue in the background of the molecule or to a side chain of such an amino acid residue.
The active agent has one or more of the following activities: anti -infective, antiinflammatory, anti-tumor, skin repair, skin damage prevention, skin anti-aging, skin lightening or whitening, hair growth promotion or hair growth inhibition, muscle repair, weight loss promoting, erectile dysfunction correcting, anti-acne, induction of immune response (antibody response and/or a cytotoxic T-cell and/or a T helper response) and CNS healing activities.
The active agent used in the compositions and methods of the present invention may be of any size.
In certain aspects, the active agents used in the present invention comprise molecules which are greated than 500Da in size. I a related aspect, molecules contained in the active ingredients are between 500Da and 15OkDa, such as, for example, 500, 600, 700, 800, 900,
1000, 1100. 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2100, 2200, 2300, 2400, 2500, 2600, 2700, 2800, 2900, 3000, 3100, 3200, 3300, 3400, 3500, 3600, 3700, 3800, 3900, 4000, 4100, 4200, 430O54400, 4500, 4600, 4700, 4800, 4900, 5000, 5100, 5200, 5300, 5400, 5500, 5600, 570O55800, 5900, 6000, 6100, 6200, 6300, 6400, 6500, 6600, 6700, 6800, 6900, 7000, 7100, 7200, 7300, 7400, 7500, 7600, 77.00, 7800, 7900, 8000, 8100, 8200, 8300, 8400, 8500, 8600, 8700, 8800, 8900, 9000, 9100, 9200, 9300, 9400, 9500, 9600, 9700, 9800, 9900, 10000, 10100, 10200, 10300, 10400, 10500, 10600, 10700, 10800, 10900, 11000, 11100, 11200, 11300, 11400, 11500, 11600, 11700, 11800, 11900, 12000, 12100, 12200, 12300, 12400, 12500, 12600, 12700, 12800, 12900, 13000, 13100, 13200, 13300, 13400, 13500, 13600, 13700, 13800, 13900, 14000, 14100, 14200, 14300, 14400, 14500, 14600, 14700, 14800, 14900, 15000, 15100, 15200, 15300, 15400, 15500, 15600, 15700, 15800, 15900, 16000, 16100, 16200, 16300, 16400, 16500, 16600, 16700, 16800, 16900, 17000, 17100, 17200, 17300, 17400, 17500, 17600, 17700, 17800, 17900, 18000, 18100, 18200, 18300, 18400, 18500, 18600, 18700, 18800, 18900, 19000, 19100, 19200, 19300, 19400, 19500, 19600, 19700, 19800, 19900, 20000, 20100, 20200, 20300, 20400, 20500, 20600, 20700, 20800, 20900, 21000, 21100, 21200, 21300, 21400, 21500, 21600, 21700, 21800, 21900, 22000, 22100, 22200, 22300, 22400, 22500, 22600, 22700, 22800, 22900, 23000, 23100, 23200, 23300, 23400, 23500, 23600, 23700, 23800, 23900, 24000, 24100, 24200, 24300, 24400, 24500, 24600, 24700, 24800, 24900, 25000, 25100, 25200, 25300, 25400, 25500, 25600, 25700, 25800, 25900, 26000, 26100, 26200, 26300, 26400, 26500, 26600, 26700, 26800, 26900, 27000, 27100, 27200, 27300, 27400, 27500, 27600, 27700, 27800, 27900, 28000, 28100, 28200, 28300, 28400, 28500, 28600, 28700, 28800, 28900, 29000, 29100, 29200, 29300, 29400, 29500, 29600, 29700, 29800, 29900, 30000, 30100, 30200, 30300, 30400, 30500, 30600, 30700, 30800, 30900, 31000, 31100, 31200, 31300, 31400, 31500, 31600, 31700, 31800, 31900, 32000, 32100, 32200, 32300, 32400, 32500, 32600, 32700, 32800, 32900, 33000, 33100, 33200, 33300, 33400, 33500, 33600, 33700, 33800, 33900, 34000, 34100, 34200, 34300, 34400, 34500, 34600, 34700, 34800, 34900, 35000, 35100, 35200, 35300, 35400, 35500, 35600, 35700, 35800, 35900, 36000, 36100, 36200, 36300, 36400, 36500, 36600, 36700, 36800, 36900, 37000, 37100, 37200, 37300, 37400, 37500, 37600, 37700, 37800, 37900, 38000, 38100, 38200, 38300, 38400, 38500, 3860O538700, 38800, 38900, 39000, 39100, 39200, 39300, 39400, 39500, 39600, 39700, 39800, 39900, 40000, 40100, 40200, 40300, 40400, 40500, 40600, 40700, 40800, 40900, 41000, 41100, 41200, 41300, 41400, 41500, 41600, 41700, 41800, 41900, 42000, 42100, 42200, 42300, 42400, 42500, 42600, 42700, 42800, 42900, 43000, 43100, 43200, 43300, 43400, 43500, 43600, 43700, 43800, 43900, 44000, 44100, 44200, 44300, 44400, 44500, 44600, 44700, 44800, 44900, 45000, 45100, 45200, 45300, 45400, 45500, 45600, 45700, 45800, 45900, 46000, 46100, 46200, 46300, 46400, 46500, 46600, 46700, 46800, 46900, 47000, 47100, 47200, 47300, 47400, 47500, 47600, 47700, 47800, 47900, 48000, 48100, 48200, 48300, 48400, 48500, 48600, 48700, 48800, 48900, 49000, 49100, 49200, 49300, 49400, 49500, 49600, 49700, 49800, 49900, 50000, 50100, 50200, 50300, 50400, 50500, 50600, 50700, 50800, 50900, 51000, 51100, 51200, 51300, 51400, 51500, 51600, 51700, 51800, 51900, 52000, 52100, 52200, 52300, 52400, 52500, 52600, 52700, 52800, 52900, 53000, 53100, 53200, 53300, 53400, 53500, 53600, 53700, 53800, 53900, 54000, 54100, 54200, 54300, 54400, 54500, 54600, 54700, 54800, 54900, 55000, 55100, 55200, 55300, 55400, 55500, 55600, 55700, 55800, 55900, 56000, 56100, 56200, 56300, 56400, 56500, 56600, 56700, 56800, 56900, 57000, 57100, 57200, 57300, 57400, 57500, 57600, 57700, 57800, 57900, 58000, 58100, 58200, 58300, 58400, 58500, 58600, 58700, 58800, 58900, 59000, 59100, 59200, 59300, 59400, 59500, 59600, 59700, 59800, 59900, 60000, 60100, 60200, 60300, 60400, 60500, 60600, 60700, 60800, 60900, 61000, 61100, 61200, 61300, 61400, 61500, 61600, 61700, 61800, 61900, 62000, 62100, 62200, 62300, 62400, 62500, 62600, 62700, 62800, 62900, 63000, 63100, 63200, 63300, 63400, 63500, 63600, 63700, 63800, 63900, 64000, 64100, 64200, 64300, 64400, 64500, 64600, 64700, 64800, 64900, 65000, 65100, 65200, 65300, 65400, 65500, 65600, 65700, 65800, 65900, 66000, 66100, 66200, 66300, 66400, 66500, 66600, 66700, 66800, 66900, 67000, 67100, 67200, 67300, 67400, 67500, 67600, 67700, 67800, 67900, 68000, 68100, 68200, 68300, 68400, 68500, 68600, 68700, 68800, 68900, 69000, 69100, 69200, 69300, 69400, 69500, 69600, 69700, 69800, 69900, 70000, 70100, 70200, 70300, 70400, 70500, 70600, 70700, 70800, 70900, 71000, 71100, 71200, 71300, 71400, 71500, 71600, 71700, 71800, 71900, 72000, 72100, 72200, 72300, 72400, 72500, 72600, 72700, 72800, 72900, 73000, 73100, 73200, 73300, 73400, 73500, 73600, 73700, 73800, 73900, 74000, 74100, 74200, 74300, 74400, 74500, 74600, 74700, 74800, 74900, 75000, 75100, 75200, 75300, 75400, 75500, 75600, 75700, 75800, 75900, 76000, 76100, 76200, 76300, 76400, 76500, 76600, 76700, 76800, 76900, 77000, 77100, 77200, 77300, 77400, 77500, 77600, 77700, 77800, 77900, 78000, 78100, 78200, 78300, 78400, 78500, 78600, 78700, 78800, 78900, 79000, 79100, 79200, 79300, 79400, 79500, 79600, 79700, 79800, 79900, 80000, 80100, 80200, 80300, 80400, 80500, 80600, 80700, 80800, 80900, 81000, 81100, 81200, 81300, 81400, 81500, 81600, 81700, 81800, 81900, 82000, 82100, 82200, 82300, 82400, 82500, 82600, 82700, 82800, 82900, 83000, 83100, 83200, 83300, 83400, 83500, 83600, 83700, 83800, 83900, 84000, 84100, 84200, 84300, 84400, 84500, 84600, 84700, 84800, 84900, 85000, 85100, 85200, 85300, 85400, 85500, 85600, 85700, 85800, 85900, 86000, 86100, 86200, 86300, 86400, 86500, 86600, 86700, 86800, 86900, 87000, 87100, 87200, 87300, 87400, 87500, 87600, 87700, 87800, 87900, 88000, 88100, 88200, 88300, 88400, 88500, 88600, 88700, 88800, 88900, 89000, 89100, 89200, 89300, 89400, 89500, 89600, 89700, 89800, 89900, 90000, 90100, 90200, 90300, 90400, 90500, 90600, 90700, 90800, 90900, 91000, 91100, 91200, 91300, 91400, 91500, 91600, 91700, 91800, 91900, 92000, 92100, 92200, 92300, 92400, 92500, 92600, 92700, 92800, 92900, 93000, 93100, 93200, 93300, 93400, 93500, 93600, 93700, 93800, 93900, 94000, 94100, 94200, 94300, 94400, 94500, 94600, 94700, 94800, 94900, 95000, 95100, 95200, 95300, 95400, 95500, 95600, 95700, 95800, 95900, 96000, 96100, 96200, 96300, 96400, 96500, 96600, 96700, 96800, 96900, 97000, 97100, 97200, 97300, 97400, 97500, 97600, 97700, 97800, 97900, 98000, 98100, 98200, 98300, 98400, 98500, 98600, 98700, 98800, 98900, 99000, 99100, 99200, 99300, 99400, 99500, 99600, 99700, 99800, 99900, 100000, 100100, 100200, 100300, 100400, 100500, 100600, 100700, 100800, 100900, 101000, 101100, 101200, 101300, 101400, 101500, 101600, 101700, 101800, 101900, 102000, 102100, 102200, 102300, 102400, 102500, 102600, 102700, 102800, 102900, 103000, 103100, 103200, 103300, 103400, 103500, 103600, 103700, 103800, 103900, 104000, 104100, 104200, 104300, 104400, 104500, 104600, 104700, 104800, 104900, 105000, 105100, 105200, 105300, 105400, 105500, 105600, 105700, 105800, 105900, 106000, 106100, 106200, 106300, 106400, 106500, 106600, 106700, 106800, 106900, 107000, 107100, 107200, 107300, 107400, 107500, 107600, 107700, 107800, 107900, 108000, 108100, 108200, 108300, 108400, 108500, 108600, 108700, 108800, 108900, 109000, 109100, 109200, 109300, 109400, 109500, 109600, 109700, 109800, 109900, 110000, 110100, 110200, 110300, 110400, 110500, 110600, 110700, 110800, 110900, 111000, 111100, 111200, 111300, 111400, 111500, 111600, 111700, 111800, 111900, 112000, 112100, 112200, 112300, 112400, 112500, 112600, 112700, 112800, 112900, 113000, 113100, 113200, 113300, 113400, 113500,
113600, 113700, 113800, 113900, 114000, 114100, 114200, 114300, 114400, 114500,
114600, 114700, 114.800, 114900, 115000, 115100, 115200, 115300, 115400, 115500,
115600, 115700, 115800, 115900, 116000, 116100, 116200, 116300, 116400, 116500, 116600, 116700, 116800, 116900, 117000, 117100, 117200, 117300, 117400, 117500,
117600, 117700, 117800, 117900, 118000, 118100, 118200, 118300, 118400, 118500,
118600, 118700, 118800, 118900, 119000, 119100, 119200, 119300, 119400, 119500,
119600, 119700, 119800, 119900, 120000, 120100, 120200, 120300, 120400, 120500,
120600, 120700, 120800, 120900, 121000, 121100, 121200, 121300, 121400, 121500, 121600, 121700, 121800, 121900, 122000, 122100, 122200, 122300, 122400, 122500,
122600, 122700, 122800, 122900, 123000, 123100, 123200, 123300, 123400, 123500,
123600, 123700, 123800, 123900, 124000, 124100, 124200, 124300, 124400, 124500,
124600, 124700, 124800, 124900, 125000, 125100, 125200, 125300, 125400, 125500,
125600, 125700, 125800, 125900, 126000, 126100, 126200, 126300, 126400, 126500, 126600, 126700, 126800, 126900, 127000, 127100, 127200, 127300, 127400, 127500,
127600, 127700, 127800, 127900, 128000, 128100, 128200, 128300, 128400, 128500,
128600, 128700, 128800, 128900, 129000, 129100, 129200, 129300, 129400, 129500,
129600, 129700, 129800, 129900, 130000, 130100, 130200, 130300, 130400, 130500,
130600, 130700, 130800, 130900, 131000, 131100, 131200, 131300, 131400, 131500, 131600, 131700, 131800, 131900, 132000, 132100, 132200, 132300, 132400, 132500,
132600, 132700, 132800, 132900, 133000, 133100, 133200, 133300, 133400, 133500,
133600, 133700, 133800, 133900, 134000, 134100, 134200, 134300, 134400, 134500,
134600, 134700, 134800, 134900, 135000, 135100, 135200, 135300, 135400, 135500,
135600, 13570O5135800, 135900, 136000, 136100, 136200, 136300, 136400, 136500, 136600, 136700, 136800, 136900, 137000, 137100, 137200, 137300, 137400, 137500,
137600, 137700, 137800, 137900, 138000, 138100, 138200, 138300, 138400, 138500,
138600, 138700, 138800, 138900, 139000, 139100, 139200, 139300, 139400, 139500,
139600, 139700, 139800, 139900, 140000, 140100, 140200, 140300, 140400, 140500,
140600, 140700, 140800, 140900, 141000, 141100, 141200, 141300, 141400, 141500, 141600, 14170O3141800, 141900, 142000, 142100, 142200, 142300, 142400, 142500,
142600, 142700, 142800, 142900, 143000, 143100, 143200, 143300, 143400, 143500, 143600, 143700, 143800, 143900, 144000, 144100, 144200, 144300, 144400, 144500, 144600, 144700, 144800, 144900, 145000, 145100, 145200, 145300, 145400, 145500, 145600, 145700, 145800, 145900, 146000, 146100, 146200, 146300, 146400, 146500, 146600, 146700, 146800, 146900, 147000, 147100, 147200, 147300, 147400, 147500, 147600, 147700, 147800, 147900, 148000, 148100, 148200, 148300, 148400, 148500, 148600, 148700, 148800, 148900, 149000, 149100, 149200, 149300, 149400, 149500, 149600, 149700, 149800, 149900, 150000Da. In a further embodiment, the molecules contained within the active ingredients are between 500 and 1000Da, such as, for example 500, 510 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990, 1000Da.
Examples of particular agents and applications follow:
1. Anti-inflammatory Activities for the Treatment of Skin Disorders.
The microemulsions of the present invention are useful inter alia for delivering small molecules, proteins (including peptides and polypeptides and chimeras), antibodies, other water soluble molecules as well as oil-soluble molecules useful in treating skin-related inflammatory conditions. For example, the microemulsions can be used to deliver nitric oxide and TNF alpha inhibitors. Vitamin B 12, for example, is a useful nitric oxide chelator and is useful in reducing skin-related inflammatory responses. In a another embodiment, the present invention contemplates a topical composition comprising a vitamin (VB 12) derivative (or analogs thereof) for the treatment of skin-related inflammatory conditions. Any inhibitor of i-nitric oxide synthase, e-nitric oxide synthase or n-nitric oxide synthase is contemplated by the present invention. Nicotinamide may also be used to block nitric oxide synthase. Other agents useful for treating psoriasis, dermatitis and itch include curcuminoids, COX-I and COX-2 inhibitors, vitamins such as vitamin E and vitamin C. In a related embodiment, the present invention contemplates a topical composition comprising a curcuminoid derivative (or an analog thereof) for the treatment of inflammatory conditions of the skin.
Proteins useful for treating skin-related inflammatory conditions such as psoriasis, dermatitis, systemic lupus erythematosus (SLE) and itch include antibodies directed against IL-20, C5aR, IFN alpha, TNF (e.g. infliximab, adalimumab), IL-12, IL-I, IL-6, leukotrienes and IL-4. Such proteins include receptor-Fc chimeras, for example, TNF receptor-Fc chimera proteins such as etanercept.
In a further embodiment, the vitamin B 12 derivative is adenosylcobalamin and the curcuminoid is tetrahydrocurcumin.
In yet another embodiment, an anti-TNF molecule is administered within a microemulsion containing both adenosylcobalamin and tetrahydrocurcumin.
2. Anti-inflammatory Activities for the Treatment of Bruising.
The microemulsions of the present invention are useful inter alia for delivering small molecules, proteins (including peptides and polypeptides and chimeras), antibodies, other water soluble molecules as well as oil-soluble molecules useful in treating bruising. This approach avoids the toxicity common with systemic or oral delivery of, for example, anti-ecchymosis therapies such as extracts of Arnica montana for the treatment of bruising. For example, the microemulsions can be used to deliver nitric oxide and TNF alpha inhibitors to a site of ecchymosis following cosmetic surgery such as rhytidectomy. Vitamin B 12, for example, is a useful nitric oxide chelator and is useful in reducing bruising, also known as ecchymosis. In one embodiment, the present invention contemplates a topical composition comprising a vitamin B 12 derivative (or analogs thereof) for the treatment of bruising. In a further embodiment, the present invention contemplates a topical composition comprising a vitamin B12 derivative (or an analog thereof) and a curcuminoid derivative (or an analog thereof) for the treatment of bruising. In a related embodiment the vitamin B 12 derivative is adenosylcobalamin and the curcuminoid is tetrahydrocurcumin.
3. Anti-inflammatory Activities for the Treatment of Muscular or Tendon Damage.
The microemulsions of the present invention are useful inter alia for delivering small molecules, proteins (including peptides and polypeptides and chimeras), antibodies, other water soluble molecules as well as oil-soluble molecules useful in treating musclar or tendon damage. Conditions such as delayed onset muscle soreness (DOMS) can prevent athletes from recovering following a training event or game, resulting in measurable reductions in power, flexibility or endurance. Vitamin B 12, for example, is a useful nitric oxide chelator and is useful in reducing DOMS. The microemulsions of the present invention can be used to deliver nitric oxide and TNF alpha inhibitors to muscular tissue to ameliorate conditions such as DOMS.
This approach avoids the toxicity common with systemic or oral delivery of other anti-inflammatory therapies such as COX inhibitors. In a related embodiment, the present invention contemplates a topical composition comprising a vitamin B 12 derivative (or analogs thereof) for the treatment of muscular damage such as DOMS. Additionally such treatment also be used for tendon injuries such as tenosynovitis,
Carpal Tunnel syndrome, bruising, bursitis and pain, repetitive strain injury (RSI), patellotendonitis, paratendonitis, proliferative tendonitis, degenerative tendonitis, enthesis In more particular embodiment, the present invention contemplates a topical composition comprising a vitamin B12 derivative (or an analog thereof) and a curcumioid derivative (or an analog thereof) for the treatment of muscular damage such as DOMS.
In a further embodiment the vitamin B 12 derivative is adenosylcobalamin and the curcuminoid is tetrahydrocurcumin. 4. Anti-inflammatory Activities for the Treatment of Joint Damage.
The microemulsions of the present invention are useful inter alia for delivering small molecules, proteins (including peptides and polypeptides and chimeras), antibodies, other water soluble molecules as well as oil-soluble molecules useful in treating joint damage. Joint damage may arise through chronic inflammatory disease states such as rheumatoid arthritis and systemic lupus erythematosus (SLE) or in response to acute damage such as that experienced by individuals during sporting activities (e.g. ankle sprain). Vitamin B 12, for example, is a useful nitric oxide chelator and is useful in reducing joint-related inflammation. The microemulsions of the present invention can be used to deliver nitric oxide and TNF alpha inhibitors to joint tissue to ameliorate conditions such as rheumatoid arthritis and joint damage following acute injury. This approach avoids the toxicity common with systemic or oral delivery of other anti-inflammatory therapies such as COX inhibitors. In a related embodiment, the present invention contemplates a topical composition comprising a vitamin B 12 derivative (or analogs thereof) for the treatment of rheumatoid arthritis, SLE, or acute joint damage. In a further preferred embodiment, the present invention contemplates a topical composition comprising a vitamin B 12 derivative (or an analog thereof) and a curcuminoid derivative (or an analog thereof) for the treatment of rheumatoid arthritis, SLE, or acute joint damage,
In a related embodiment the vitamin B 12 derivative is adenosylcobalamin and the curcuminoid is tetrahydrocurcumin.
5. Topical Vaccines.
The microemulsions are useful as topical vaccines such as to induce an antibody response and/or a cytotoxic T-cell and/or a T helper response including a TH1/TH2 response. Topical vaccines are particularly useful for mobilizing dendritic cells in the dermal layer and increasing numbers of activated dendritic cells. In the past, hyperdermic needles were able to deliver agents to the subcutaneous layer or muscle layer, however, the procedure was invasive and did not necessarily target the desired region. The topical microemulsion may be provided with or without an adjuvant. Adjuvants contemplated herein include microorganisms or viruses or components thereof and chemical substances which enhance the immune response and/or chemical which act as preservatives and tissue fixatives. Particularly useful adjuvants include aluminium hydroxide, aluminium phosphate and calcium phosphate. In addition, oil based emulsions and products from bacteria including synthetic derivatives and liposomes of gram-negative bacteria, endotoxins, cholesterol, fatty acids, aliphatic amines, parafmic and vegetable oils, monophosphoryl lipid A, iscoms with Quil-A and Syntex agents formulations containing threonyl derivaties or muramyl type peptides may be employed. Other adjuvants contemplated by the present invention include Freund's emulsify oil adjuvants (complete and incomplete), ARLACEL A, mineral oil, emulsified peanut oil adjuvant (adjuvant 65), mineral compounds and bacterial products from Bordetella pertussis, Corynebacterium granulosom-deήvQd P40 component, lipopolysaccaride, Mycobacterium or components thereof, inulin, cholera toxin and/or liposomes may also be employed. Still other adjuvants contemplated by the present invention include proteinacous pathogen recognition receptors or related chimeric molecules such as soluble C-type lectins, for example, CD209L, CD209L- Fc, DC-SIGN, DC-SIGN-Fc, CLEC4K, CD 207. Binding of pathogen by C-type lectins induces internalisation and degradation in lysosomes, which facilitates antigen processing and presentation and the initiation of an immune response. In particular, the use of human cell expressed recombinant human C-type proteins would be preferred as the specific post-translational modifications such as sugar moieties are known to be important in the interaction of C-type lectin with dermally located immune cells such as Langerhans cells. Specific disease states that can be topically treated with a microemulsion of the present invention include Hepatitis B virus (HBV), Hepatitis C virus (HCV), DPT, Human paploma virus (HPV), Japanese encephalitis, shigella, malaria, rabies, measles, mumps, rubella, whooping cough, rotavirus and herpes zoster. In a related embodiment, the present invention contemplates a topical composition comprising a antigen and an adjuvant within a microemulsion for the vaccination of a vertebrate host.
6. Topical Fat Weight Loss.
The microemulsions of the present invention may be used to induce weight loss via lymphodystrophy. It is proposed that topically applied insulin induces insulin- mediated lymphodystrophy. There is minimal if any entry of the insulin to the blood system and hence no increase in glucose levels. Reference to "insulin" includes insulin analogs and precursor forms of insulin which may have the ability to interact with insulin receptors but lack signalling capability. In a related embodiment, the present invention contemplates a topical composition comprising insulin for the amelioration of sub-cutaneuous fat deposits. In another embodiment of the present invention there is described a topical composition comprising growth factor administered within a microemulsion for the amelioration of obesity. In yet another embodiment of the present invention there is described a topical composition comprising a growth hormone releasing hormone, or growth hormone releasing peptide for the amelioration of obesity. Other examples of agents which are useful in sub-cutaneuous fat deposits include Coleus Forskohlii Root Extract and Bodyfit.
7. Imaging and Treatment of Skin Cancer and Breast Cancer.
According to this aspect of the present invention, the microemulsions are able to access or introduce imaging reporter molecules to the sentinel lymph node in relation to particular cancers such as melanoma and breast carcinoma. The sentinel lymph node is the first lymph node to which cancer is likely to spread from the primary tumor. Cancer cells may appear first in the sentinel node before spreading to other lymph nodes and other places in the body. In accordance with the present invention, imaging prior to the removal of the sentinel lymph node increases the likelihood that the correct lymph node is removed in its entirety. Any imaging agent may be employed, such as a radio-active agents, chelates, fluorescent compounds and metals. The identification of the sentinel lymph node is improved and can be successfully removed thus reducing the risk of spread of a cancer. In chelate methods a radionuclide is bound to the targeting molecule indirectly, through a bifunctional chelating agent (BFCA)
Examples of radioactive agents which may be admixed with the microemulsions include the following: Antimony-124, Antimony-125, Arsenic-74, Barium-103, Barium- 140, Beryllium-7, Bismuth-206, Bismuth-207, Cadmium- 109, Cadmium- HSm, Calcium-45, Cerium-139, Cerium-141, Cerium-144, Cesium-137, Chromium-51, Cobalt-56, Cobalt-57, Cobalt-58, Cobalt-60, Cobalt-64, Erbium-169,
Europium-152, Gadolinium-153, Gold-195, Gold-199, Hafnium-175, HafniumO175- 181, Indium-I l l, Iridium-192, Iron-55, Iron-59, Krypton-85, Lead-210, Manganese- 54, Mercury- 197, Mercury-203, Molybdenum-99, Neodymium-147, Neptunium- 237, Nickel-63, Niobium-95, Osmium-185+191, Palladium- 103, Platinum- 195m, Preseodymium-143, Promethium-147, Protactinium-233, RadiumO226, Rhenium-
186, Rubidium-86, Ruthenium- 103, Ruthenium- 106, Scandium-44, Scandium-46, Selenium-75, Silver-l lOm, Silver-Ill, Sodium-22, Strontium-85, Strontium-89, Strontium-90, Sulfur-35, Tantalum-82, Technetium-99m, Tellurium- 125, Tellurium- 132, Thallium-204, Thorium-228, Throium-232, Thallium-170, Tin-113, Titanium- 44, Tungsten-185, Vanadium-48, Vanadium-49, Ytterbium- 169, Yttrium-88,
Yttrium-90, Yttrium-91, Zinc-65 or Zirconium-95.
Examples of chelating groups which may be admixed with the microemulsions include the following: 1, 3, 5 triaminocyclohexane; 1, 3, 5 triaminocyclohexane N-pyridine; 1,1- cyclobutanedicarboxylic acid; l,2-Dimethyl-3-hydroxypyridin-4-one; 1 ,2-dimethyl- 3-hydroxypyridin-4-one (Deferiprone); 1, 4,7,10-tetraaza-4,7, 10- tris(carboxymethyl)- 1 -cyclododecylacetyl-i?-(+)-R-methylbenzylamine (DOTA- MBA); l,4,7,10-tetraazacyclododecane-N,Λ'',N5Λ''-tetraacetic acid (DOTA); 1,6- dimethyl-2-(l-hydroxyethyl)-3-hydroxypyridin-4-one; l-ethyl-2- (1-hydroxyethyl)-
3-hydroxypyridin-4-one; l-hydroxypryidin-2one; l-hydroxypryidin-2one; 2- Deoxy-2-(N-carbamoylmethyl-[Λr9-29-methyl-39-hydroxypyrid-49-one])-D- glucopyranose; 2-furoylcarboxaldehyde isonicotinoyl hydrazone (FIH); 2-hydroxy- 1-naphthylaldehyde benzoyl hydrazone; 2-hydroxy-l-naphthylaldehyde isonicotinoyl hydrazone; 2-hydroxy-l-napthylaldehyde isonicotinoyl hydrazone; 2- methyl-3-hydroxy-4H-benzopyran-4-one (MCOH); 2-pyridylcarboxaldehyde 2- thiophenecarboxyl hydrazone (PCTH); 2-pyridylcarboxaldehyde benzoyl hydrazone (PCBH); 2-pyridylcarboxaldehyde isonicotinoyl hydrazone (PCIH); 2- pyridylcarboxaldehyde isonicotinoyl hydrazone (PCIH); 2-pyridylcarboxaldehyde m-bromobenzoyl hydrazone (PCBBH); 2-pyridylcarboxaldehyde m-bromobenzoyl hydrazone (PCBBH); 2-pyridylcarboxaldehyde p-aminobenzoyl hydrazone
(PCAH); 2-pyridylcarboxaldehyde p-hydroxybenzoyl hydrazone (PCHH); 2- pyridylcarboxaldehyde thiophenecarboxyl hydrazone (PCTH); 311 2-hydroxy-l- napthylaldehyde isonicotinoyl hydrazone; 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (Triapine®) ;3-aminopyridine-2-carboxyaldehyde thiosemicarbazone; 3-hydroxypyridin-2-one; 3-hydroxypyridin-4-one; 4-[3,5-bis-
(hydroxyphenyl)-l,2,4-triazol-l-yl] -benzoic acid (ICL670A), aminocarboxylates, B APTA/ AM (1 ,2-bis(2-aminophenoxy)ethane-N,N,N9,N9-tetraacetic acid acetoxymethyl ester); Catechols; CDTA cyclohexanediaminetetraacetic acid; cis- 1,3,5-triaminocyclohexane; clioquinol; DDC diethyldithiocarbamate; Defarasirox; Deferiprone (l,2-dimethyl-3-hydroxypyrindin-4-one); Deferoxamine; Demercaptol;
DFO desferoxamine; DFOA; Diaminocyclohexane; diethylenetriaminepentaacetic acid; DMB; DMPA dimercaptopropionic acid; DMPS; DMSA dimercaptosuccinic acid;DPA (D-penicillamine); DTPA (diethylene triamine pentaacetic acid); EDTA, (ethylendiaminetetraacetic acid); Ferroportin-1; Hydroxamates; Hydroxycarboxylates; hydroxypyridinones ; IDA iminodiacetic acid; MECAM;
N,iV_-bis-(2-hydroxybenzyl) ethylenediamine-iV,jV_-diacetic acid (HBED); N,Nφ- bis(2-hydroxybenzyl)ethylenediamine-iV,iV-diacetic acid (HBED); N,Nfi-Bis(3,4,5- trimethoxybenzytyethylenediamine-ΛfJV-diacetic acid; N,N-dimethyl-2,3- dihydroxybenzoic acid; N4,NR,Λ'R5N_y/V_-pentakis[[((iV-hydroxy-Λ''- methyl]carbonyl)methyl]-2, 6-diamino-4-azahexanoic hydrazide; NAPA iV-acetyl-
D-penicillamine; N-ethyl N, N5 N-tris(pyridylmethyl)-cis,cis, 1,3,5,- triaminocyclohexane; NOTP ( 1 ,4,7-triazacyclononane- 1 ,4,7- tris(methylenephosphonate)); NOTPME (1 ,4,7-triazacyclononane-l ,4,7- tris(methylenephosphonatemonoethylester)); N-pyridine; NTA nitrilotriacetic acid; oxalic acid; pyridoxal hydrochloride; pyridoxal isonicotinoyl hydrazone; Pyridoxal isoonicotinoyl hydrazone (PIH); pyridoxal metachlorobenzoyl hydrazone; pyridoxal metafluorobenzoyl hydrazone; pyridoxal paramethoxybenzoyl hydrazone; rhizoferrin; salicylaldehyde benzoyl hydrazone; staphloferrin; staphloferrin; succinic acid; tachpyridine; TETA (triethylenetetra- amine); tetraaza-4,7, 10-tris(carboxymethyl)- 1 -cyclododecylacetylbenzylamine (DOTA-BA); TREN-(Me-3,2-hydroxipyridonate) (HOPO); TRENCAM; Triapine,
3~aminopyridine-2-carboxyaldehyde thiosemicarbazone; TTD tetratethythiuramdisulfide; TTHA triethylenetetraminehexaacetic acid; Deferiprone (1 ,2-dimethyl-3-hydroxypyridin-4-one); HYNIC (6-hydrazinonicotinamide); HYNIC-Kp-DPPB and HYNIC-Ko-DPPB where (HYNIC ) 6- hydrazinonicotinamide, where K is lysine and DPPB is diphenylphosphine-benzoic acid; HPO 3-hydroxypyridin-4-one; 1 -(2' -carboxyethyl)-2 -methyl-Shy droxypyridin-4-one; l-(3'-hydroxypropyl)-2-methyl-3-hydroxypyridin-4-one; or 1 -(2' -hydroxyethyl)-2-ethyl-3-hydroxypyridin-4-one.
Chelates for use during imaging are readily formed by mixing the appropriate radionuclide (formulated within the microemulsion) with an equimolar amount of the particular chelating moiety (also formulated within the microemulsion). The resultant radionuclide-chelate forms spontaneously within the microemulsion and can be used almost immediately for topical application.
Microemulsions can also be used to topically introduce chemotherapeutic agents and cytotoxic compounds to a site close to the tumor. This approach avoids the toxicity common with the systemic or oral delivery of most chemotherapies. Particular cancers that may be treated in this way include skin cancers such as melanoma, squamous cell carcinoma, basal cell carcinoma, solar keratosis as well as breast carcinoma. Examples of chemotherapeutic agents which may be admixed with the microemulsions include the following:
Antimetabolites: substances that interfere with the body's chemical processes, such as creating proteins, DNA, and other chemicals needed for cell growth and reproduction; in cancer treatment, antimetabolite drugs disrupt DNA production, which in turn prevents cell division. Examples include Azaserine, D-Cycloserine, Mycophenolic acid, Trimethoprim, 5-fluorouracil, capecitabine, methotrexate, gemcitabine, cytarabine (ara-C) and fludarabine.
Antitumor antibiotics which interfere with DNA by stopping enzymes and mitosis or altering the membranes that surround cells. These agents work in all phases of the cell cycle. Thus, they are widely used for a variety of cancers. Examples of antitumor antibiotics include dactinomycin, daunorubicin, doxorubicin
(Adriamycin), idarubicin, and mitoxantrone.
Mitotic inhibitors which are plant alkaloids and other compounds derived from natural products. They can inhibit, or stop, mitosis or inhibit enzymes for making proteins needed for reproduction of the cell. These work during the M phase of the cell cycle. Examples of mitotic inhibitors include paclitaxel, docetaxel, etoposide (VP- 16), vinblastine, vincristine, and vinorelbine.
Steroids which are natural hormones and hormone-like drugs that are useful in treating some types of cancer (lymphoma, leukemias, and multiple myeloma) as well as other illnesses. When these drugs are used to kill cancer cells or slow their growth, they are considered chemotherapy drugs. They are often combined with other types of chemotherapy drugs to increase their effectiveness. Examples include prednisone and dexamethasone. Sex hormones, or hormone-like drugs, which alter the action or production of female or male hormones. They are used to slow the growth of breast, prostate, and endometrial (lining of the uterus) cancers, which normally grow in response to hormone levels in the body. These hormones do not work in the same ways as standard chemotherapy drugs. Examples include anti-estrogens (tamoxifen, fulvestrant), aromatase inhibitors (anastrozole, letrozole), progestins (megestrol acetate), anti-androgens (bicalutamide, flutamide), and LHRH agonists (leuprolide, goserelin, histerillin, nafarelin, buserelin).
Alkylating agents which work directly on DNA to prevent the cancer cell from reproducing. As a class of drugs, these agents are not phase-specific (in other words, they work in all phases of the cell cycle). These drugs are active against chronic leukemias, non-Hodgkin's lymphoma, Hodgkin's disease, multiple myeloma, and certain cancers of the lung, breast, and ovary. Examples of alkylating agents include busulfan, chlorambucil, cyclophosphamide, ifosfamide, dacarbazine (DTIC), mechlorethamine (nitrogen mustard), and melphalan.
Platinum and other heavy metals may also be used in the treatment of cancers. Examples of platinum compounds and other compounds that may be used in the treatment of cancers include but are not limited to Cisplatin, oxaliplatin, carboplatin, diaminocyclohexylplatinin (DACH-platinum), nedaplatin, cycloplatan, SKI2053R, Loboplatin, enloplatin, zeniplatin, Miboplatin, L-NDDP, TRK-710, MBA (bis monobromo acetato trans 1 ,2-diaminocyclohexane platinum II), PYP (Bis-pyruvato 1,2-diaminocyclohexaneplatinum II), Sulphato 1,2 diaminocyclohexane platinum II, Malonato 1,2 diaminocyclohexane platinum II, Isocitrato 1,2-diaminocyclohexane platinum II, 4-Carboxyphtalato 1,2-diaminocyclohexane platinum II, bis- monobromoacetato trans 1,2-diaminocyclooctane platinum II, 1,1 diaminomethyl cyclohexane sulphate platinum II, JM-216 (bis-acetato-ammine-dichloro- cyclohexylamine platinum IV, Iproplatin (cis-dichloro-trans-dihydroxy-bis- isopropylamino platinum IV, CHIP, JM-9), Iproplatin (cis-dichloro-trans-dihydroxy- bis-isopropylamino platinum IV, CHIP, JM-9), 5-fluorouracil, leucovorin, Capecitabine, Gemcitabine, Topotecan, 9-aminocamtotecin, Rubitecan, Irinotecan, Exatecan, Docetaxol, Doxifluridine, Carmofur, UFT, Eniluracil, ZD-9331, MMI- 166, Eniluracil, 6-hydroxymethylacylfulvene, Nediplatin, Satroplatin, Lipoplatin, BBR3464, ZD0473, lobaplatin, Spirogermanium, Gallium trinitrate, etoposide, gallium chloride, Budotitane, titanocene dichloride, trans-indazolium-
[tetrachlorobis(2H-indazole)ruthenate(III,Nl )] (HInd[RuInd2C14])3 Trans- Na[RuInd2C14], ruthenium dimethylsulfoxide, thioguanidine, hexamethylmelamine), 5_nor-anhydro-vinblastine (Vinorelbine), Avastin (bevacizumab), BEC2 (mitomomab), Tositumomab (Bexxar),Campath (alemtuzumab), CeaVab, herceptin (trastuzumab), IMC-C225 (centuximab),
Lymphocide (epratuzumab), MDX-210, Mylotarg (Gemtuzumab), Panorex (Edorcolomab), Rituxan (Rituximab), Theragyn (pemtumomab), Zamyl, Zevalin (Ibritumomab), Gefitinib (Iressa), Erlotinib, ABX-EGF, GSK 572016 (lapatinibditosylate), CI-1033 (canertinib diHCl), Bortezomib, Arsenic trioxide (Trisenox), CC-394 (Actimid), CDC-501 (Revlimid), Thalidomide (Thalomid),
Oblimersen (Genasense), Abarelix (Plenaxis), Zevalin, Gemtuzumab, 2-MPPA, CPI-0004Na.
Nitrogen mustard in the form of its crystalline hydrochloride which is used as a drug in the treatment of Hodgkin's disease, non-Hodgkin's lymphomas, and brain tumors.
Nitrogen mustards cause mutations in the genetic material of cells, thereby disrupting mitosis, or cell division. Cells vary in their susceptibility to nitrogen mustards, with rapidly proliferating tumor and cancer cells most sensitive; bone marrow, which produces red blood cells, is also sensitive, and depression of red blood cell production is a frequent side effect of nitrogen mustard therapy. The nitrogen mustards also suppress the immune response (see immunity). Other types include the aromatic mustards melphalan and chlorambucil, cyclophosphamide,
FTNl, έ/s-(2-chloroethyl), ethylamine; HN2, &w-(2-chloroethyl), methylamine and
HN3, tλ7J-(2-chloroethyl), amine. Nitrosoureas act in a similar way to alkylating agents. They interfere with enzymes that help repair DNA. These agents are able to travel to the brain so they are used to treat brain tumors as well as non-Hodgkin's lymphomas, multiple myeloma, and malignant melanoma. Examples of nitrosoureas include carmustine (BCNU) and lomustine (CCNU).
Hormone agonists such as Leuprolide (Lupron, Viadur, Eligard) for prostate cancer, Goserelin (Zoladex) for breast and prostate cancers and Triptorelin (Trelstar) for ovarian and prostate cancers and nafarelin acetate (Synarel).
Microtubule inhibitors include "Vinca" alkaloids, taxoids and benzimidazoles.
Curcuminoid derivatives such as tetrahydrocurcumin (THC), sodium circuminate, bisdemethoxycurcumin, demethoxycurcumin, 5'- methoxycurcumin and dihydrocurcumin, tetrahydrodemethoxycurcumin (INCI: tetrahydrodemethoxy- diferuloymethane), and tetrahydrobisdemethoxycurcumin (INCI: tetrahydro- bisdemethoxydif eruloylmethane .
Trichostatin A (TSA), an inhibitor of the enzyme deacetylase.
In one embodiment, the present invention contemplates a topical composition comprising 99mTc chelated to DTPA for the identification of a sentinel lymph node in relation to melanoma or breast carcinoma.
In an embodiment, the present invention contemplates a topical composition comprising one or more chemotherapeutic agents for the treatment of skin or breast cancer.
In another embodiment, the present invention contemplates a topical composition comprising a curcuminoid derivative for the treatment of melanoma or breast carcinoma. In a related embodiment, the present invention contemplates a topical composition comprising TSA for the treatment of melanoma or breast carcinoma.
In yet another embodiment, the present invention contemplates a topical composition comprising an anthracyclin for the treatment of breast carcinoma. This composition may optimally also contain 5-fluorouracil and chlorambucil.
In a further embodiment, the present invention contemplates a topical composition comprising a chelate between an imaging agent and a chelating agent. Examples include, but are not limited to 99m-Tc-HYNIC, 99m-Tc-DTPA, ""^-ciprofloxacin, 99mTc-methylene diphosphonate (MDP), indium or technetium-99"1 hexamethylpropylene amine oxime (HMPAO), 11 1In-DTP A-Folate, 99mTc diadenosine tetraphosphate (Ap4A; AppppA, -Pl,.P4-di(adenosme-5*)- tetraphosphate) and its analog 99mTc AppCHClppA, 99mTc-HYNIC-IL-8., 99mTc-
TRODAT-I, 99mTc-M-TRODAT, 99mTc-RP128, Gd(III) DOTA, 99mTc-H YNIC- folate, [99mTc(SG38)(tricine)(TPPTS)] (RP517), natCu-D OTADYl -TATE and DOTA-natl-DYl-TATE, 61Cu-DOTA-DYl -TATE, 99mTc labeled [(iV-[2-((39-N9- propyl-[3,3,l]aza-bicyclononan-3a-yl)(20-methoxy-5-methyl-phenylcarbamate)(2- mercaptoethyl)amino)acetyl]-2-aminoethanethiolato] technetium(V) oxide),,
Albumin-(biotin)10-(gadopentetate)25, 99mTc0(BAT-NI), (131 I, 123I, "1In, (67Ga, 66Ga, 64Cu, 67Ga-deferoxamine-folate, mIn-octadentate-DTPA-folate (g), 1 1 1In- DTPA-NOON-pteroate., 99mTc(CO)3-DTPA-folate (g), 99mTc-EC20 (99mTc-Cys- Asp-Dap-D-Glu-Pte), 99mTc-oxa-PnAO-(a and g)-folatel l55, 99mTc-ethylene dicysteine (99mTc-L,L-EC), Co-56, Co-57, Co-58, and Co-60-labelled vitamin B 12,
DCTA, ! ' 'indium-DTPA-vitamin B 12.
In a still further embodiment, the present invention contemplates a topical composition comprising a chemotherapeutic agent linked to a targeting agent. Such targeting agents selected from Amphiregulin, Antibodies including Avastin
(bevacizumab), BEC2 (mitomomab), Tositumomab (Bexxar),Campath (alemtuzumab), CeaVab, herceptin (trastuzumab), IMC-C225 (centuximab), Lymphocide (epratuzumab), MDX-210, Mylotarg (Gemtuzumab), Panorex (Edorcolomab),Rituxan (Rituximab), Theragyn (pemtumomab), Zamyl, Zevalin (Ibritumomab),, BAFF, BDNF, bFGF, BMP, BMP-4, BMP-7, CD209L, CSF, EGF, Fas-Ligand, Fc fragments, FGF, FGF2, FGFRl, FGFR4, Flt3, Flt3-Ligand, G-CSF,
GM-CSF, hGH, hGHR, Growth Hormone Releasing Hormone and analogs, Growth Hormone Releasing Peptides, IFN, IgG, IGF-I, IGFBP-3, Interleukins, Langerin, LHRH analogs, LIF, L-Selectin, Lymphotoxin-a, MC-148, MCP-I, MIP-Ia, MIP- Ib, MLIF, NGF, NGFR, OSM, OX-40, PDGF, RANTES, Tf, TGF, TGF-bl, TNF, TNF-a, TPO, TrkA, TrkB, VEGF, Vitamin derivatives, including derivatives of vitamin B 12, biotin, folate and/or riboflavin.
8. Skin Lightening and Re-Pigmentation.
The microemulsions of the present invention can be used to treat melasma and other areas where the skin contains no pigment or to lighten the skin where there areas of colored blotches. In one example, the microemulsions are used to introduce nucleic acid molecules to induce gene expression in order to produce pigment. Melanin and other pigmentation molecules may also be introduced.
Examples of skin lightening, skin whitening and re-pigmentation molecules include estrogen-like compounds such as chromenes, isoflavones and coumestran groups of phyloestrogens (coumestan glycosides). Examples of chromenes include microestrol and deoxymicroestrol. Examples of isoflavones include diadzen, genisten, mirificin, puerarin and puerarin-6"-monoacetate. Examples of coumestran glycosides include moumestrol, mirificoumestan, mirificoumestan glycol and mirificoumestran hydrate.
A useful source of estrogen-like compounds is from the plant genus Pueraria such as Pueraria lobata and Pueraria muriflca and hence the present invention extends to the estrogen-like compound contained within an extract of a Pueraria plant such as an extract of P. muriflca and/or P lobata.
Collagen fragment mimetics include dermaxyl (a palmitoyl oligopeptide), ceramide 2 (a natural skin lipid) and matrixyl 3000 which is a mixture of palmitoyl oligopeptides (e.g. palmitoyl-GHK and palmitoyl GQPR in a ratio of 2:1) may also be administered with the microemulsion alone or with an anti-oxidant.
Examples of suitable anti-oxidants include retinyl palmitate (Vitamin A)5 allantoin (also called 5-ureidohydantoin, glyoxyldiureide and 5-ureidohydantoin; it is a diureide of glycoxylic acid) and D-α tocopherol (Vitamin E).
9. Gene Therapy.
The microemulsions of the present invention are a useful vehicle to introduce nucleic acid molecules such as DNA, siRNA, anti-sense RNA, effector RNA molecules and microRNAs to inhibit gene expression or to facilitate gene expression. Reference to gene therapy in this regard includes the introduction of virosomes or constructs to facilitate gene expression. Gene therapy is useful inter alia to the treatment of cancer, diabetes and inflammatory conditions. In an embodiment, the present invention contemplates a topical composition comprising a siRNA for the treatment of cancer.
In another embodiment a microemulsion of the present invention may be used in the formation of DNA/Ca++ nanoparticles. In an extension of the previous embodiment the DNA/Ca++ nanoparticles of the present invention can be used to increase the frequency of in vitro cellular transfection.
10. Muscle Dystrophy.
The microemulsions of the present invention are useful inter alia for delivering small molecules, proteins (including peptides and polypeptides and chimeras), antibodies, other water soluble molecules as well as oil-soluble molecules useful in the treatment of muscular dystrophy. An example of a small molecule with therapeutic potential in the treatment of muscular dystrophy is trichostatin A (TSA), an inhibitor of the enzyme deacetylase. In an embodiment, the present invention contemplates a topical composition comprising TSA for the treatment of muscular dystrophy.
11. Treatment of Neuropathic and Nociceptive Pain.
The microemulsions of the present invention may be used to introduce analgesic agents such as to include but not limited to fentanyl, oxycodone, codeine, dihydrocodeine, dihydrocodeinone enol acetate, morphine, desomorphine, apomorphine, diamorphine, pethidine, methadone, dextropropoxyphene, pentazocine, dextromoramide, oxymorphone, hydromorphone, dihydromorphine, noscapine, papverine, papveretum, alfentanil, buprenorphine and tramadol and pharmaceutically acceptable salts, derivatives, homologs or analogs thereof as well as opioid agonists. Both neuropathic and nociceptic pain may be treated and agents useful for either neuropathic or nociceptic pain may be introduced via the microemulsion. Pain includes the treatment of sport injuries, such as but not limited to, inflammatory conditions, planta fasciatis, bursitis of the knee, elbow, shoulder, subcutaneous prepatella bursa, olecranon bursa, tenosynovitis, Carpal Tunnel syndrome, bruising, Repetitive Strain Injury (RSI), patellotendonitis, paratendonitis, proliferative tendonitis, Degenerative tendonitis, enthesis and other conditions such as delayed onset muscle soreness (DOMS) and stings. In an embodiment, the present invention contemplates a topical composition comprising one or more analgesic agents for the treatment of neuropathic or nociceptic pain. In yet another embodiment of the invention the treatment of pain involves the use of a microemulsion containing a nitric oxide scavenger or an inhibitor of NOS such as Niacinamide, vitamin B 12 and derivatives, curcumin and derivatives L-NAME5JV0- nitro-L-arginine methyl ester; L-NMA, JV^-methyl-L-arginine, L-NMMA, JV0- 8 000908
- 45 -
monomethyl-L-arginine, S-Methylisothiourea sulphate, Aminoguanidine. Inhibitor of iNOS, 7-nitro indazole or N-nitro-L-arginine.
12. Erectile Dysfunction.
The microemulsion of the present invention can be used to topically apply agents which are useful in treating erectile dysfunction including inhibitors of cGMP specific phosphodiesterases type 5 (PDE5). These molecules act by inhibiting the breakdown of nitric oxide (NO). Examples of PDE5 inhibitors include sildenafil, tadalafil and vardenafil. It is proposed that the microemulsion be topically applied to overcome erectile dysfunction. This approach avoids the side effects often associated with oral delivery of PDE5 inhibitors, including headache, lightheadedness, dizziness, flushing and change in vision. In a related embodiment, the present invention contemplates a topical composition comprising a PDE5 inhibitor for the treatment of erectile dysfunction.
Persistent, painful erection of the penis (priapism) is relatively common in young males with sickle cell anaemia but can also result following oral delivery of PDE5 inhibitors. It is therefore contemplated that the microemulsion of the present invention can be used to topically apply agents which enhance the break-down of
NO for the treatment of persistent erection. Example of such molecules include NO chelators such as vitamin B12 derivatives as well as inhibitors of NO synthase (e.g. Niacinamide, curcumin and derivatives, L-NAME^-nitro-L-arginine methyl ester; L-NMA, Λ^-methyl-L-arginine, L-NMMA, JV°-monomethyl-L-arginine, S- Methylisothiourea sulphate, Aminoguanidine. 7-nitro indazole, N-nitro-L-arginine).
In a preferred embodiment, the present invention contemplates a topical composition comprising an NO chelator for the treatment of persistent erection. In another embodiment, the present invention contemplates a topical composition comprising an NO synthase inhibitor for the treatment of persistent erection. 13. Acne Treatment.
The microemulsion of the present invention can be used to topically treat acne. Examples of agents which are useful in treating acne include curcumin, tetrahydrocurcumin, sodium circuminate, bisdemethoxycurcumin, demethoxycurcumin, 5'- methoxycurcumin and dihydrocurcumin, vitamin E, selenium, zinc, Vitamin C, azaleic acid, pantothenic acid, benzyl peroxide and extract of tea tree oil. In an embodiment, the present invention contemplates a topical composition comprising one or more compounds selected from the list of: Curcumin, tetrahydrocurcumin, sodium circuminate, bisdemethoxycurcumin, demethoxycurcumin, 5'- methoxycurcumin and dihydrocurcumin, vitamin E, selenium, zinc, vitamin C, azaleic acid, pantothenic acid, benzyl peroxide and extract of tea tree oil for the treatment of acne.
14. Treatment of Neurological Conditions.
The microemulsion of the present invention can be used to topically treat oxidative damage associated with neurological disease. Such diseases include neurodegenerative conditions like Parkinson's disease, Alzheimer's disease and other dementias as well as amyotrophic lateral sclerosis and multiple sclerosis. Other neurological diseases include those following acute injury such as stroke and spinal cord damage. Examples of agents which are useful in treating oxidative damage associated with neurological disease include NO chelators such as vitamin B 12 derivatives as well as inhibitors of NO synthase (e.g. Niacinamide, vitamin B12 and derivatives, curcumin and derivatives L-NAME5N0 -nitro-L-arginine methyl ester; L-
NMA, JV^-methyl-L-arginine, L-NMMA, JV^-monomethyl-L-arginine, S- Methylisothiourea sulphate, Aminoguanidine. 7-nitro indazole Inhibitro of nNOS, N-nitro-L-arginine). In a related embodiment, the present invention contemplates a topical composition comprising an NO synthase inhibitor for the treatment of acute CNS injuries such as stroke and spinal cord damage. 15. Treatment of Abrasions.
The microemulsion of the present invention can be used to topically treat skin damage associated with cuts and abrasions. Such skin damage is susceptible to microbial infection. Povidone-iodine type compounds such as Betadine are commonly used broad-spectrum topical microbicides as treatments for abrasions but can display poor capacity to penetrate within the wound. In an embodiment, the present invention contemplates a topical composition comprising a povidone-iodine type compound for the treatment of skin damage associated with cuts and abrasions.
16. Amelioration of aging symptoms.
The microemulsion of the present invention can be used topically to ameliorate the symptoms of skin aging, especially in relation to the face. Symptoms of facial (and other non facial) skin ageing include wrinkles, a lowering of the collagen content of the dermis, facial "puffiness", the appearance of hyperpigmentation adjacent to the eyes, photo-aging of skin such as by free radical scavenging and an increase in trans- epidermal water loss from the skin. Examples of agents which are useful in counteracting the effects of skin ageing include collagen stimulating agents such as Dermican, Pueraria Mirifica Extract; anti-wrinkle agents such as Vialox, SynAke,
Gatuline Expression; melanin inhibitors and/or tyrosinase inhibitors including Haloxyl, phytoestrogens, Melaclear2, Lumiskin, Sepiwhite and stilbene compounds such as resveratrol; compounds that affect skin "puffiness including Haloxyl, Eyeliss, Esculoside and other mixtures containg esculin, anti-bacterial compounds suitable for application to skin including Triclosan; exfoliation compounds suitable for application to skin including bromelain and papain.
The present invention provides, therefore, a microemulsion that is capable of transdermal delivery of pharmaceutical or physiologically useful or active agents. As used herein, the term "microemulsion" refers to a system comprising an oil phase, a water phase, one or more zwitterionic surfactants; one or more co-surfactants and an active agent wherein the microemulsion is a single optically isotropic and thermodynamically stable liquid solution. The term "transdermal delivery" is not to be construed as passage of an agent solely through the dermal layer although such passage is also contemplated by the present invention. The term provides passage of agents through all layers of the skin.
The microemulsion of the present invention comprises an oil phase, a water phase and one or more zwitterionic surfactantsand an active agent, wherein the microemulsion is capable of transdermal delivery of the active agents. In one embodiment, the microemulsions further comprises a co-surfactant, a co-solvent, or a combination thereof.
The microemulsion of the present invention may be produced by methods known in the art. In general, microemulsions are produced by emulsifying components under conditions including typically sufficient force or the required temperature to generate the required dispersion level, conductivity, viscosity, percolativity or other dispersion characteristics.
Microemulsion formation can be assessed using scattering and spectroscopic techniques such as neutron scattering, time-average scattering, quasi-electric light scattering i.e., high- resolution ultrasonic spectroscopy or photon correlation spectroscopy. The partition coefficients of microemulsions may also be measured chromatographically.
The selection of particular formulations is based on a number of different paradigms depending upon the desired application. Illustrative paradigms include the hydrophilic- lipophilic balance, the phase-inversion temperature, or the cohesive-energy ratio.
Microemulsions may be formulated using a wide range of immiscible liquids and other additional agents.
The microemulsion of the present invention may comprise an oil phase of between 50 and
99% by weight, most preferably between 50 and 90% by weight; a water phase of between 2 and 50% by weight, most preferably between 1 and 50% by weight; 0.1 to 90% by weight a zwitterionic surfactant, preferably 1 to 90% by weight zwitterionic surfactant. The microemulsion may further comprise 0.1 to 90% by weight co-surfactant or cosolvent; preferably 1 to 90% by weight co-surfactant or cosolvent.
The oil phase may comprise natural oils derived from plants or animals, such as vegetable oils, sunflower oils, coconut oils, almond oils; purified synthetic or natural di or triglycerides (such as Crodamol GTCC™ and Capmul MCM™); phospholipids and their derivatives (such as lecithin or lysolecithin); fatty acid esters (such as isopropyl myristate, isopropyl palmitate, ethyl oleate, oleic acid ethyl ester); hydrocarbons (such as hexane, the n-decane through n-octadecane series); and/or glycerolysed fats and oils (such as glyceryl monooleate, glyceryl monocaprylate, glycerol monocaprate, propylene glycol monocaprylate, propyleme glycol monolaurate).
Other oil phase ingredients include, but are not limited to, Labrafil M 1944 CS™, benzene, tetrahydrofuran, and n-methyl pyrrolidone, or halogenated hydrocarbons, such as methylene chloride, or chloroform. In a particular embodiment, the oil phase comprises Crodamol GTCC™ and Capmul MCM™, at 3: 1 ratio.
The oil component is either used alone or in combination with other oil component. Each oil or unique mixture of oils may require a different surfactant or mixture of surfactants or surfactants and co-surfactants to form a microemulsion with the water phase, as can routinely be determined by those of skill in the art.
Water phase ingredients may comprise water and any water-soluble components in water, including one or more active agents.
Zwitterionic surfactants are used in the compositions and methods of the present invention. These surfactants are very mild and can be anionic (negatively charged), cationic (positively charged) or non-ionic (no charge) in solution, depending on the acidity or pH of the water.
They are compatible with all other classes of surfactants and are soluble. Zwitterionic surfactants may contain two charged groups of different sign. Whereas the positive charge is almost always ammonium, the source of the negative charge may vary (carboxylate, sulphate, sulphonate).
Zwitterionic surfactants include the betaines, sulfobetaines, i.e. sultaines and fatty acid ethanolamides. Examples of betaines include dodecyldimethylammonium acetate, tetradecyldimethylammonium acetate, hexadecyldimethylammonium acetate, alkyldimethylammonium acetate wherein the alkyl group averages about 14.8 carbon atoms in length, dodecyldimethylammonium butanoate, tetradecyldimethylammonium butanoate, hexadecyldimethylammonium butanoate, dodecyldimethylammonium hexanoate, hexadecyldimethylammonium hexanoate, tetradecyldiethylammonium pentanoate and tetradecyldipropyl ammonium pentanoate, and so forth. Examples of fatty acid betaines include coco-betaine. Examples of fatty acid ethanolamides include cocamide monoethanolamide. Still other examples of zwitterionic surfactants include 3-(N,N- dimethyl-N-hexadecylammonio)-propane-l -sulfonate; 3-(N,N-dimethyl-N- hexadecylammonio)-2-hydroxypropane- 1 -sulfonate; N,N-dimethyl-N-dodecylammonio acetate; 3-(N,N-dimethyl-N-dodecylammonio)propionate; 2-(N,N-dimethyl-N- octadecylammonio)ethyl sulfate; 3-(P,P-dimethyl-P-dodecylphosphonio)propane-l- sulfonate; 2-(S-methyl-S-tert-hexadecylsulfo)ethane-l -sulfonate; 3-(S-methyl-S- dodecylsulfonio)propionate; N,N-bis(oleylamidopropyl-N-methyl-N- carboxymethylammonium betaine; N,N-bis(stearamidoproρyl)-N-methyl-N- carboxymethylammonium betaine; N-(stearamidopropyl)-N-dimethyl-N- carboxymethylammonium betaine; 3-(N-4-n-dodecylbenzyl-N,N- dimethylammonio)propane-l -sulfonate; 3-(N-dodecylphenyl-N,N-dimethylammonio)- propane- 1 -sulfonate.
Co-surfactants used according to the subject invention are known surface-active agents in the art that act, in addition to surfactants, to further lower the interfacial energy of a microemulsion. Co-surfactants include, but are not limited to non-toxic amphiphilic molecules; alcohols (including aliphatic alcohols, shorter chain alcohols, such as ethanol); fatty acid alcohols (such as n-alkane-l,2-diols); acids (such as acetic acid); esters (such as butyl lactate); anionic co-surfactants, cationic co-surfactants and non-ioninc co-surfactants; any surfactants as herein listed or mixtures thereof. Alcohol content may range, for example, from 0 to about 30% by volume in the microemulsion.
Anionic co-surfactants include fatty acid soaps (including sodium oleate, sodium palmitate, sodium myristate, sodium sterate, potassium oleate and triethanolamine oleate); alkyl sulfates (including sodium dodecyl sulfate, ammonium lauryl sulfate, triethanolamine lauryl sulfate and sodium alkyl sulfate); alkyl lactylates (including calcium stearoxyl-2-lactylate), alkyl lactates (including sodium-0-stearyllactate and sodium stearoyllactylate) alkyl benzenesulfonates (including calcium dodecyl benzene sulfonate); alkyl sulfonates (including alkyl aryl sulfonate); alkyl phosphates; alkyl oleates; alkyl stearates (including self-emulsifying glycerol monostearate); alkyl esters (including dioctyl ester of sodium sulphosuccininc acid (AOT, Aerosol OT)); acyl sulfates; or acyl sulfosuccinates.
Cationic co-surfactants include alkyl primary, secondary, tertiary, or quaternary amines; high-molecular-weight amine and fatty amine blends; polyoxyethylene fatty amines (including tallow amine); alkyl sulfates (including N-cetyl-N-ethyl morpholinium ethyl sulfate (35%)); alkyl pyridinium and quaternary ammonium salts.
Non-ionic co-surfactants include alcohol ethoxylate, alkylphenol ethoxylate, fatty acids (such as oleic acid), lanolin alcohols (such as polyoxyethylene (5) lanolin alcohol (ether and ester), polyoxyethylene (50) lanolin (ether and ester), acetylated polyoxyethylene (10) lanolin, polyoxyethylene (16) lanolin alcohol, acetylated polyoxyethylene (9) lanolin), alkyl polyglycosides, mono-, di- or glyceride esters (such as diglycerine sesquioleate), acetylated monoglycerides, polyglycerols, polyglycerol esters (such as decaglycerol decaoleate, decaglycerol octaoleate, decaglycerol tetraoleate), phospholipids (such as lecithin), mono- or diglyceride esters of citric acid, tartaric acid and lactic acid, sorbitan fatty acid esters (such as sorbitan monostearate (Span 60, Crill 3), sorbitan monooleate (Arlacel 80, Span 80, Crill 4), sorbitan isosterate (Crill 6), sorbitan monolaurate (Arlacel 20, Span 20, Crill 1), sorbitan trioleate (Span 85, Crill 45), sorbitan tristearate (Span 65), sorbitan sesquioleate (Arlacel 83, Crill 43), sorbitan monopalmitate (Span 40, Crill 2)), polyol fatty acid esters (such as ethylene glycol distearate, ethylene glycol monostearate, diethylene glycol monostearate, propylene glycol monostearate, propylene glycol monolaurate, polyoxyethylene (1.5) nonylphenol, polyoxyethylene (4) nonylphenol, polyoxyethylene (5) nonylphenol, polyoxyethylene (6) nonylphenol, polyoxyethylene (8) nonylphenol, polyoxyethylene (20) nonylphenol, polyoxyethylene (30) nonylphenol, polyoxyethylene (10) nonylphenol, poly(ethylene glycol) 200 distearate, poly(ethylene glycol) 300 dilaurate, poly(ethylene glycol) 400 distearate, polyoxyethylene octylphenol, poly(ethylene glycol) 400 dilaurate, poly(ethylene glycol) 400 monostearate, poly(ethylene glycol) 400 monolaurate, poly(ethylene glycol) 4000 distearate, polyoxyethylene (10) octylphenol, poly(ethylene glycol) 600 monostearate, Polyoxyethylene (14) nonylphenol, polyoxyethylene (24) cholesterol, polyoxyethylene (25) soyasterol, poly(ethylene glycol) 1000 monooleate, polyoxyethylene (25) propylene glycol monostearate, poly(ethylene glycol) 1000 monolaurate, polyoxyethylene (70) dinonylphenol), glycerol fatty acid esters (such as glycerol dioleate, glycerol monoleate, glycerol monostearate, glycerol monolaurate, polyoxyethylene (20) glycerol monostearate), sucrose fatty acid esters (such as sucrose distearate, sucrose monolaurate), polyoxyethylene sorbitan fatty acid esters (polysorbates) (such as polyoxyethylene (4) sorbitan monolaurate, polyoxyethylene (5) sorbitan monooleate, polyoxyethylene (20) sorbitan monooleate (Tween 80), polyoxyethylene (40) sorbitol hexaoleate, polyoxyethylene (50) sorbitol hexaoleate, polyoxyethylene (20) sorbitan tristearate, polyoxyethylene (20) sorbitan trioleate, polyoxyethylene (20) sorbitan monostearate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monolaurate (Tween 20), polysorbate 20 NF, EP, JP, poly(ethylene glycol)-20 sorbitan isostearate, poly(ethylene glycol) (20) sorbitan trioleate (Crillet 45), poly(ethylene glycol) (20) sorbitan stearate (Crillet 3 Super, Polysorbate 60), poly(ethylene glycol) (20) sorbitan oleate (Crillet 4 Super, Polysorbate 80), poly(ethylene glycol) (20) sorbitan laurate (Crillet 2 Super, Polysorbate 40)), monoesters (such as polyoxyethylene (4) stearic acid, polyoxyethylene (8) stearic acid, polyoxyethylene (8) lauric acid, polyoxyethylene (40) stearic acid, polyoxyethylene (50) stearic acid), polyethoxylated esters of acyl acids (such as polyoxyethylene (2) octyl alcohol, polyoxyethylene (4) tridecyl alcohol, polyoxyethylene (6) tridecyl alcohol, polyoxyethylene (8) tridecyl alcohol), copolymers of polyethylene oxide and polypropylene oxide, polyoxyethylene fatty ethers (such as polyoxyethylene fatty ethers derived from lauryl, cetyl, stearyl and oleyl alcohols, polyoxyethylene (4) lauryl ether, polyoxyethylene (23) lauryl ether (Brij 35), polyoxyethylene (2) cetyl ether (Brij 52), polyoxyethylene (10) cetyl ether, polyoxyethylene (20) cetyl ether (Brij 58), polyoxyethylene (2) stearyl ether (Brij 72), polyoxyethylene (10) stearyl ether, polyoxyethylene (20) stearyl ether, polyoxyethylene (2) oleyl ether, polyoxyethylene (10) oleyl ether (Brij 97), polyoxyethylene (20) oleyl ether, polyoxyethylene (21) stearyl ether, polyoxyethylene (12) lauryl ether), fatty amides (such as N,N,-Dimethylstearamide), Polyethylene glycol ether of linear alcohol, polyoxyethylene (15) tall oil fatty acids (ester), acetylated sucrose diesters, isopropyl ester of lanolin fatty acids, polyoxyethylene sorbitol beeswax derivative, Polyoxypropylene/Polyoxyethylene condensate, sodium oleate, polyoxyethylene (20) castor oil (ether, ester), glycerol oleate & propylene glycol (Arlacel 186) and Cremophor.
Zwitterionic surfactants in the microemulsion of the present invention may be used alone or in combination with each other.
The microemulsion of the present invention may further comprise solvents or other agents to enhance emulsion formation or stability. Other agents may be introduced to provide functions such as pH, ionic content, polymerisation, taste, smell, sterility, colour, viscosity etc.
The microemulsions of the present invention may also be generated using any suitable synthetic plastic or polymeric, monomeric or hybrid colloidal material.
The present invention further extends to a pharmaceutical composition comprising the microemulsion and one of more active or pharmaceutical agents as described herein, wherein such pharmaceutical composition is suitable for topical application on a subject.
Suitable pharmaceutical agents for use in the present invention include any water soluble solute, including, but not limited to peptides, proteins, polysaccharides, oligonucleotides, salts, sugars, nutrients, vitamins, nucleic acids minerals, acids, anti-oxidants, or any biological active compounds for administration to a subject, such as a human, animal or other mammal. The pharmaceutical agent is selected based upon the intended application or therapy, wherein the effect of the pharmaceutical agent is suitable to treat a particular condition.
Suitable peptides and proteins that can be delivered via the microemulsion of the present invention include molecules, protein chimeric molecules, other chimeric molecules or fragments selected from the TNF superfamily (including TNF-α, TNFRl, TNFR2, BAFF, BAFFR, OX-40, Lymphotoxin-α, Fas-ligand); chemokines (including MCP-I, MIP-Ia, MIP-Ib3 RANTES, IL-8 and viral like chemokine antagonist MC148); interleukins, interleukin receptors and antagonist (including IL-Ia IL-Ib5 IL-2, IL-3, IL-4, IL-5, IL-6, IL- 7, IL-8, IL-10, IL-I l5 IL-12, IL-13, IL-15, IL-16, IL-18, IL-20, IL-21, IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, their respective receptors including IL-IRa, IL- 2Ra, IL-2Rb, IL-2Rg, IL-3Ra, IL-4Ra, IL-5Ra, IL-6Ra, IL-7Ra, IL-IORa, IL-I lRa, IL- 13Ra5 IL-15Ra as well as IL-IR Antagonist); the interferon family (including IFN-a2B, IFN-bl, IFN-g, IFN-y, IFN-aR2, IFN-aRa, IFN-aRb, IFN-gRa, IFN-gRb); lectins (including CD209 type I and II, E-Selectin, L-Selectin5 P-Selectin, Langerin, DC-SIGN); growth factors and their receptors (including Amphiregulin, Angiopoietin, BDNF, beta-cellulin, BMPs (including BMP-2, BMP-4, BMP-7), CNTF, cripto, ECGF-I5 EGF5 EGFR5 EPO5 FGFs and their receptors (including FGF-I5 FGF-2, FGF-5, FGF-7, FGF-9, FGF-11, FGF- 12, FGF-13, FGF-14, FGF-14 FLAG5 FGF-18, FGF-19, FGF-21, FGFRl5 FGFRl, FGFR4, FGFR5), Flt3-Ligand and its receptor (including Flt3)5 G-CSF, GDNF, GM-CSF, GM- CSF-R, hGH and its receptor (including hGHR), IGF-I, IGFBP-3, M-CSF, Neuregulin, NGFs and its receptor (including NGF-b, NGFR, NGFR), NT-35 PDGFs5 TGFs and their receptors (including TGF-a, TGF-b, TGFbR2)5 Trk-A, Trk-B , TPO, VEGFs and their receptors (including VEGF-A5 VEGF-B5 VEGF-C5 VEGF-D5 VEGF-165, VEGFR); embryonic growth factors (such as Noggin, Nodal, SCF5 Wnts, Wnt-2, Wnt-3, Wnt-3A, Wnt-4, Wnt-5A, Wnt-5 A-FLAG-C5 Wnt-5B5 Wnt-65 Wnt-7A5 Wnt-7B5 Wnt-IOA, Wnt- 10B5 Wnt-1 OB-FLAG-C, Wnt-11); adhesion molecules (such as adiponectin, ICAM)5 other cytokines and proteins, such as LIF5 OSM5 transferring and its receptor, hormones (such as insulin, calcitonin, adrenocorticotropin (ACTH), glucagon, somatostatin, somatotropin, thymosin, parathyroid hormone, pigmentary hormones, somatomedin, lutenizing hormone and agonists and antagonists thereof, chorionic gonadotropin, hypothalmic releasing factors, antidiuretic hormones, vasopressin, thyroid stimulating hormone, endorphins, enkephalins, biphalin and prolactin.); antibiotics (such as gentamycin, amikacin, neomycin, penicillin, streptomycin), enzymes (such as activin A asparaginase, adenosine deaminase, BACE-I, caspase-1, fucosyltransferase, furin, mTACE, sialyltransferase ) Factor VIII, LH-RH analogs, anticoagulants such as Heparin, Warfarin, Herbal extracts such as those derived from Danshen .Devil's Claw, Eleuthero, Garlic, Ginger, Ginkgo, Horse Chestnut, Panax Ginseng, Papain, Red Clover, Saw Palmetto, capsaicin and vaccines (for instance, vaccines for Hepatitis 'B' surface antigen, typhoid, tetanus and cholera vaccines) and plasminogen activator inhibitors, and small peptides such as MLIF (Met-Gln-Cys-Asn-Ser), or analogs thereof.
Suitable chimeric proteins that can be delivered via the microemulsion of the present invention include TNFRl-Fc, TNFR2-Fc, OX-40-Fc, MC148-Fc, IL-IRa-Fc, IL-2Ra-Fc,
IL-2Rb-Fc, IL-2Rg-Fc, IL-3Ra-Fc, IL-4Ra-Fc, IL-5Ra-Fc, IL-6Ra-Fc, IL-7Ra-Fc, IL-IORa-
Fc, IL-I lRa-Fc, IL- 13Ra-Fc, IL- 15Ra-Fc, IFN-aRa-Fc, IFN-aRb-Fc, IFN-gRa-Fc, IFN- gRb-Fc, CD209L-Fc, DC-SIGN, E-Selectin-Fc, L-Selectin-Fc, P-Selectin-Fc, Langerin-
FLAG, EGFR-Fc, FGFRl-Fc, FGFR4-Fc, FGFR5-Fc, Flt3-Fc, hGHR-Fc, NGFR-Fc, TGFbR2-Fc, Trk-A-Fc, Trk-B-Fc, VEGFR-Fc, Wnt-5A-FLAG-C, WnM OB-FLAG-C and, most preferably, TNFRl-Fc, TNFR2-Fc.
In another embodiment, the active agent of the present invention comprises MLIF or analogs thereof, such as iodacetylated MLIF.
Additional suitable proteins include monoclonal and polyclonal antibodies, single-chain antibodies, other antibody fragments, analogs and derivatives thereof. Polynucleotides, including antisense oligonucleotides, aptamers and therapeutic genes can also be delivered using the methods and compositions of the present invention. In a further embodiment, suitable antibodies that can be delivered via the microeniulsion of the present invention include antibodies directed against TNF alpha, IL-I, IL-8, IL-6 or other cytokines involved in the inflammatory response and, most preferably, anti-TNF alpha antibodies with the technical names of infliximab and adalimumab.
Anticoagulants, such as Heparin, Warfarin, Herbal extracts such as those derived from Danshen Devil's Claw, Eleuthero, Garlic, Ginger, Ginkgo, Horse Chestnut, Panax Ginseng, Papain, Red Clover, Saw Palmetto, capsaicin, also can be delivered using the methods and compositions of the invention. Still other suitable therapeutic agents for use in the present invention include bioactive molecules, such as anticancer drugs, e.g., Curcumin derivatives, Glycyrrhizin, Glycyrrhetinic acid, antracycline, platinum drugs, methotrexate derivatives, heary metals, genistein, chlorambucil, cyclophosphamide, melphalan, cyclopropane, doxorubicin, daunomycin, adriamycin, mitomycin C, [2-(hydroxymethyl)anthraquinone], methotrexate, dichloromethatrexate, cisplatin, carboplatin, metallopeptides containing platinum, copper, vanadium, iron, cobalt, gold, cadmium, zinc and nickel, DON, thymidine, pentamethylmelamin, dianhydrogalactitol, 5-Methyl-THF, anguidine, maytansine, neocarzinostatin, chlorozotocin, AZQ, 2'deoxycoformycin, PALA, AD-32, rø-AMSA and misonidazole, doxorubicin, epirubicin and daunorubicin, vincristine, vinblastin methotrexate, paclitaxol, taxol, camptothecin and camptothecin analogs, antipsychotics, antidepressants, and drugs for the treatment of diabetes and cardiovascular disease.
Other agents that can be delivered via the microemusltion of the present invention are ones which have anti-inflammatory properties, such as NSAIDs. Exemplary NSAIDs include salicylates (including aspirin, choline magnesium trisalicylate, dilfunisal, salasalate, benorylate); phenylalkanoic acids (including carpofen, fenoprofen, fluribiprofen, ibuprofen, ketoprofen, naproxen, naproxen Na, suprofen, oxaprozin,); acetic acids or indoles (including alclofenac, diclofenac, fenclofenac, indomethacin, sulindac, tolemetin); enolic acids (including isoxicam, meloxicam, piroxicam, tenoxicam); fenamic acids (including flufenamic acid, meclofenamate, mefenamic acid); napthylalkanones (including nabumetone); niflumic acid; pyranocarboxylic acids (including etodolan); pyrazolones (including phenylbutazone, oxyphenobutazone); pyrroles (including ketorolac); COX-2 inhibitors (including celecoxib, parecoxib, valdecoxib, rofecoxib). Some brand names of NSAIDs are shown in Table 2.
TABLE 2
Brand Names of NSAIDs
Figure imgf000058_0001
Figure imgf000059_0001
In another embodiment, the active agent suitable to be delivered via the microemulsion of the present invention comprises copper complexes of NSAIDs. Formation of copper complexes of NSAIDs reduces toxicity and may enhance the anti-inflammatory properties of the complex due to the anti-inflammatory property of copper. Such pharmaceutical agents include copper complexes of salicylates (including aspirin, choline magnesium trisalicylate, dilfunisal, salasalate, benorylate); copper complexes of phenylalkanoic acids (including carpofen, fenoprofen, fluribiprofen, ibuprofen, ketoprofen, naproxen, naproxen Na, suprofen, oxaprozin); copper complexes of acetic acids or indoles (including alclofenac, diclofenac, fenclofenac, indomthacin, sulindac, tolemetin); copper complexes of enolic acids (including isoxicam, meloxicam, piroxicam, tenoxicam); copper complexes of fenamic acids (including flufenamic acid, meclofenamate, mefenamic acid); copper complexes of napthylalkanones (including nabumetone); copper complexes of niflumic acid; copper complexes of pyranocarboxylic acids (including etodolan); copper complexes of pyrazolones (including phenylbutazone, oxyphenobutazone); copper complexes of pyrroles (including ketorolac); copper complexes of COX-2 inhibitors (including celecoxib, parecoxib, valdecoxib, rofecoxib). In yet another embodiment, the active agent may be selected from copper complexes of tyrosine, lysine, valproic acid, 2, 2,-bypyridine, 1, 10-phenanthroline, 2, 9-dimethyl-l, 10- phenanthroline, glycylglycine, cimetidine, sulfathiazole, tetraanhydroaminmobenzaldehyde, metronidazole and imidazole.
Other potential complexes suitable for delivery as active agents include copper complexes with amino acids, aromatic carboxylic acid, corticoids, tetrazoles, histamines, penicillamines, anti-ulcer histamine antagonists, rantidine and cimetidine.
The aforementioned copper complexes may further comprise one of more molecules of
DMF, DMSO, Pyridine, Caffeine, Methylimidazole, Imidazole, 2-methylbenzimidazole, metronidazole, 2-methylimidazole, 3-picoline, 4-picoline, imidazole, 1 -methylimidazole, diethylamine, nicotinamide, papaverine, N, N-dimethylacetamide, N-methyl-2- pyrrolidone, ethanol and methanol.
The pharmaceutical composition of the present invention comprising one or more copper- NSAID complexes may be formed by mixing two microemulsions of the present invention containing NSAID and copper respectively.
Suitable vitamins include but are not limited to water soluble vitamins such as vitamin C, analogs or derivatives thereof; folic acid and analogs or derivatives thereof (including but not limited to methotrexate, aminopterin, 10-deazaminopterin, 10-ethyl-lO- deazaaminopterin, 5,10-dideazatetrahydrofolate, folinic acid, 7-hydroxyaminopterin); niacin (nicotinic acid, vitamin B3) and analogs or derivatives thereof (including but not limited to beta-hydroxybutyrate, acipimox, niceritrol, nicotinamide (niacin)); thiamine (vitamin B-I), analogs and derivatives thereof; riboflavin (vitamin B2) and its analogs or derivatives thereof (including but not limited to 7-nor-7-chlororiboflavin, 8-nor-8- chlororiboflavin, 7-nor-7-bromoriboflavin, 8-nor-8-bromoriboflavin, 7 -methylribofiavin, 8 -methylribofiavin, 7 ,8 -dimethylriboflavin, 7-nor-7-bromo-8 -methylribofiavin, 7 -methyl- 8-nor-8-bromoriboflavin, 7-nor-7-chloro-8 -methylribofiavin, 7 -methyl-8~nor-8- chlororiboflavin, 8-nor-8-fluororiboflavin, 7-nor-7-chloro-8-nor-8-chlororiboflavin, 8-nor- 8-aminoriboflavin, N(3)-methylriboflavin and 5-deaza-5-carbariboflavin); pyridoxine (vitamin B6), analogs or derivatives thereof; cyanocobalamin (vitamin B 12), analogs or derivatives thereof; pantothenic acid (vitamin B5), analogs or derivatices thereof; biotin, analogs or derivatives thereof; and the vitamin E derivative Trolox. Suitable vitamins also include but are not limited to oil soluble vitamins such as vitamin A, analogs or derivatives thereof, such as acitretin (Soriatane); vitamin E (alpha tocopherol) analogs or derivatives thereof.
Suitable vitamin B 12 (VB 12) analogs according to the invention include descobaltocorrinoids such as alpha (5,-6-dimethylbenzymidazolyl)-hydrogenobamide, hydroxocobalamin (OH-CbI), methylcobalamin, adenosylcobalamin (AdeCbl), aquocobalamin, methylcobalamin, cyanocobalamin, carbanalide, 5 methoxybenzylcyanocobalamin [(5-MeO)CN-CbI], as well as the desdimethyl, monoethylamide and the methylamide analogs of all of the above VB 12 molecules. Other suitable VB 12 analogs include all alkyl cobalamins in which the alkyl chain is linked to the corrin nucleus by a direct CoC covalent bond. Still other suitable VB 12 analogs include chlorocobalamin, sulfitocobalamin, nitrocobalamin, thiocyanatocobalamin, benzimidazolecyanocobalamin derivatives such as 5,6 dichlorobenzimidazole, 5- hydroxybenzimidazole, trimethylbenzimidazole, as well as adenosylcyanocobalamin [(Ade)CN-Cbl], cobalamin lactone, cobalamin lactam and the anilide, ethylamide, monocarboxylic and dicarboxylic acid derivatives of VB 12 or its analogs. Other VB 12 derivatives include adeninylalkylcobalamin, adeninylethylcobalamin (AdeEtCbl), adeninylpropylcobalamin (AdePrCbl) and adeninylpentylcobalamin (AdePeCbl)). Alternative derivatives of VB 12 include the mono, di- and tricarboxylic acid derivatives or the propionamide derivatives of VB 12. Alternatively, suitable analogs of VB 12 include those in which the cobalt ion is replaced by either a zinc or nickel ion. Other derivatives and analogs of vitamin B12 are discussed in Schneider and Stroinski, Comprehensive B 12, Walter De Gruyter, Berlin, NY, 1987, the disclosure of which is incorporated herein by reference. In a related embodiment, the active agent of the present invention comprises aquocobalamin, adenosylcobalamin, methylcobalamin, hydroxycobalamin, cyanocobalamin, carbanalide, or 5 methoxybenzylcyanocobalamin [(5-MeO)CN-CbI].
Without limiting the scope of the invention, VB 12 analogs or derivatives of the present invention are involved in the sequestration and/or neutralization of nitric oxide and other reactive nitrogen species. Thus, nitric oxide and nitric superoxide rapidly combine to form a toxic reaction product, peroxynitrite anion (ONOO). The ratio of superoxide to NO is important in determining the reactivity of peroxynitrite: excess NO or excess superoxide reduces the oxidation elicited by peroxynitrite. The oxidant reactivity of peroxynitrite is mediated by an intermediate with the biological activity of the hydroxyl radical. Activated macrophages have been shown to secrete high levels of NO due to inducible NO synthase (iNOS). Molecules such as vitamin B12 have been shown to reduce the activity of NO by complexation to the CN group. Additionally, the skin contains many dendritic cells, which have also been shown to produce iNOS when stimulated. Such stimulation may occur in clinical conditions such as psoriasis.
Suitable curcumin analogs include tetrahydrocurcumin, bisdemethoxycircumin, extracts from Curcuma xanthorrhiza or C. domestica such as 25 P54FP and extracts from C. phaeocaulis valeton. Suitable curcumin derivatives include 5'- methoxycurcumin, dihydrocurcumin, cyclocurcumin and demethoxy curcumin, as well as curcumins and curcuminoids such as those described in the Chinese Pharmacopoeia; Yujin (from the tubers of C. wenyujin, C. longa, C. kwangsiensis or C. phaeocaulis); Jianghuang (the rhizome of C. longa); Pian-Jianghuang (the rhizome of C. wenyujin) and Ezhu (the rhizomes of C. phaeocaulis, C. kwangsiensis or C. wenyujin) as well as in the rhizomes of C. zedoaria and C. aromatica. Additional molecules with anti-inflammatory properties include those that are structurally related to curcumin, such as diarylheptanoids obtained from Alpinia oxyphylla, several sesquiterpenes, germacrone, turmerone, ar-(+)-, a-, β- turmerones; β- bisabolene; a-curcumene; zingiberene; β- sesquiphellandene, bisacurone; curcumenone; dehydrocurdione; procurcumadiol; bis-acumol; curcumenol; isoprocurcumenol epiprocurcumenol; procurcumenol; zedoaronediol; curlone; and turmeronol A and turmeronol B, as well as four polysaccharides-ukonans - having activity on the Reticuloendothelial system, along with stigmasterol, β-sitosterol, cholesterol and 2- hydroxymethyl anthraquinone.
In a further embodiment, the active agent of the present invention comprises a curcumin analog and, in particular, tetrahydrocurcumin.
In a further embodiment, the pharmaceutical composition of the present invention comprises a microemulsion, an emulsion or a cream and one of more active agent as described herein, wherein such pharmaceutical composition is suitable for topical application on a subject.
The present invention further provides a method suitable for the transdermal delivery of active agents in a range of therapeutic, cosmetic and research applications. The method comprises first preparing a water phase, wherein any water-soluble components, including any active agents, are dissolved in water. Oil soluble components are dissolved in the oil phase as appropriate. Any undissolved material is separated by centrifugation or filtration. The oil and water phases and one or more suitable zwitterionic surfactants and co- surfactants are then mixed in a suitable vessel, and given time to equilibrate. Standard emulsification techniques, like stirring, use of membranes, applying shear, ultrasound and the like, can be used to facilitate the mixing of the two phases. The pharmaceutical composition is then applied on a subject.
Examples of therapeutic, cosmetic and research applications of the present invention include any skin conditions, or any dermatologic disorders, including dermatitis, bacterial, fungal, parasitic, viral infections of the skin, disorders of hair follicles and sebaceous glands, scaling papular diseases, psoriasis, inflammatory reactions, reaction to sunlight, bullous diseases, disorders of cornification, pressure sores, pigmentation disorders, disorders of sweating, benign and malignant tumours. Examples of dermatitis include contact dermatitis, atopic dermatitis, seborrheic dermatitis, nummular dermatitis, chronic dermatitis of hands and feet, generalized exfoliative dermatitis, stasis dermatitis, lichen simplex chronicus.
Examples of bacterial infections of the skin include cellulitis, acute lymphangitis, lymphadenitis, erysipelas, cutaneous abscesses, necrotizing subcutaneous infections, staphylococcal scalded skin syndrome, folliculitis, furuncles, hidrandenitis suppurativa, carbuncles, paronychial infections, erythrasma.
Examples of fungal skin infections include dermatophyte infections including tinea corporis, tinea pedis, tinea unguium, tinea capitis, tinea cruris, tinea barbae, dermatiphytids or Id Eruptions; yeast infections including candidiasis, tinea versicolor.
Examples of parasitic skin infections include scabies, pediculosis, creeping eruption.
Examples of viral skin infections include warts and molluscum contagiosum.
Examples of hair follicles and sebaceous glands disorders include acne, rosacea, perioral dermatitis, hypertrichosis, alopecia, pseudofolliculitis barbae, keratinous cyst.
Examples of scaling papular diseases include psoriasis, pityriasis rosea, lichen planus, pityriasis rubra pilaris.
Examples of inflammatory reactions of the skin include drug eruptions, toxic epidermal necrolysis, erythema multiforme, erythema nodosum, granuloma annulare.
Examples of skin reaction to sunlight include sunburn, chronic effects of sunlight, such as actinic keratoses, squamous and basal cell carcinoma and malignant melanomas, and photosensitivity. Examples of bullous diseases include pemphigus, bullous pemphigoid, dermatitis herpetiformis, linear immunoglobulin A disease.
Examples of cornification disorders include ichthyosis, keratosus pilaris, calluses and corns.
Pressure sores results from ischemic necrosis and ulceration of tissues overlying a bony prominence that has been subjected to prolonged pressure against an external object, such as bed, wheelchair, cast or splint. Examples of pressure sores include bedsores, decubitus ulcers, trophic ulcers.
Examples of pigmentation disorders include hypopigmentation and hyperpigmentation.
Examples of sweating disorders include miliaria and hyperhidrosis.
Examples of benign tumours include moles, dysplastic nevi, skin tags, lipomas, angiomas, pyogenic granuloma, seborrheic keratoses, dermatofibroma, keratoacanthoma, keloid.
Examples of malignant tumours include basal cell carcinoma, squamous cell carcinoma, Bowen's disease, malignant melanoma, breast cancer, Paget' s disease of the nipples, and Kaposi's sarcoma.
Examples of cosmetic applications of the present invention include the amelioration of skin wrinkles, the lightening of skin colour, the amelioration of facial "puffiness", the cleansing of the skin of bacteria and skin exfoliation.
Other examples of therapeutic, cosmetic and research applications of the present invention include any musculoskeletal and connective tissue disorders, including rheumatoid arthritis, Sjogren's syndrome, Behcet's syndrome, relapsing polychondritis, systemic lupus erythematosus, discoid lupus erythematousis, systemic sclerosis, eosinophilic fasciitis, polymyositis, dermatomyositis, polymyalgia rheumatica, vasculitis, termporal arteritis, polyarteritis nodosa, Wegener's granulomatosis, mixed connective tissue disease, ankylosing spondylitis, Reiter's syndrome, psoriatic arthritis, osteoarthritis, neurogenic arthropathy, avascular necrosis, infectious arthritis, ostemyelitis, gout, idiopathic hyperuricemia, calcium pyrophosphate dihydrate crystal deposition disease, basic calcium phosphate and other crystal disorders, muscular dystrophies, benign and malignant tumors of bone, osteoporosis, Paget's disease of bone, nonarticular rheumatism (such as spasmodic torticollis, low back pain, bursitis, tendinitis, tenosynovitis, iϊbromyaglia, ankle sprains, disorders associated with heel pain (such as calcaneal spur syndrome, epiphysitis of the calcaneus, posterior achilles tendon bursitis, fracture of the posterolateral talar tubercle, anterior achilles tendon bursitis, posterial tibial nerve neuralgia), disorders associated with metatarsalgia (such as interdigital nerve pain, metatarsophalangeal articulation pain, hallux rigidus), common hand disorders including deformities (such as Mallet finger, swan-neck deformity, Boutonniere deformity, erosive (inflammatory) osteoarthritis, Dupuytren's contracture), neurovascular syndromes (such as carpal tunnel syndrome, cubital tunnel syndrome, radial tunnel syndrome, Kienbock's disease, ganglia, reflex sympathetic dystrophy), trauma, infections, congenital deformities, tendon problem; and common sports injuries including metatarsal stress fracture, shin splints, popliteus tendinits, achilles tendinitis, patellofemoral pain, posterior femoral muscle strain, piriformis syndrome, lumbar strain, lateral epicondylitis, medial epicondylitis and rotator cuff tendinitis.
Other examples of therapeutic, cosmetic and research applications of the present invention include inflammatory diseases not related to skin or musculoskeletal and connective tissue such as multiple sclerosis, Crohn's disease and ulcerative colitis.
The present invention is further described by the following non-limiting examples. EXAMPLE 1 Preparation of a Pharmaceutical Composition with a Water-in-Oil Microemulsion
An oil phase of a 3:1 ratio of Crodamol GTCC to Capmul MCM C8 is prepared. A surfactant/co-surfactant phase of a 3:2:0.75 ratio of Crillet 4 (Tween 80) to Crill 4 (Span
80) to AB30 (coco-betaine) is prepared. An oil-surfactant phase was then prepared by mixing the oil and surfactent phases at a ratio of 7.6:1.4. A water phase containing concentrated tetanus toxoid protein (44 mg/ml, concentrated stock solution CSL
Laboratories) is added to oil-surfactant phase at a 1:10 ratio and mixed by gentle shaking and/or mild heating.
EXAMPLE 2 Preparation of a Pharmaceutical Composition with a Water-in-Oil Microemulsion
An oil phase of a 3:1 ratio of Crodamol GTCC to Capmul MCM C8 was prepared. A surfactant/co-surfactant phase of an 8:5:2 ratio of Crillet 4 (Tween 80) to Dehyton AB30 (coco-betaine) to Crill 4 (Span 80) was also prepared. An oil-surfactant phase was then prepared by mixing the oil and surfactant phases at a ratio of 6.8:2.2. Microemulsions were formed by mixing the oil-surfactant phase with one ninth - one tenth volume of water. One or more of the following agents were dissolved in the microemulsions: forskolin powder (to yield a final concentration of 3-5 mg/ml), resveratrol powder (to yield a final ME concentration of 8-15 mg/ml) and tetrahydrocurcumin powder (to yield a final ME concentration of 6-15 mg/ml). Microemulsion formation was often assisted by gentle shaking and/or heating the mixture to 50 degrees Celsius.
EXAMPLE 3 Preparation of a Pharmaceutical Composition with a Water-in-Oil Microemulsion
An oil phase of a 3:1:0.9 ratio of Crodamol GTCC to Capmul MCM C8 to phytosqualene (Sophim) was prepared. A surfactant/co-surfactant phase of an 6:5:5 ratio of Crillet 4
(Tween 80) to Dehyton AB30 (coco-betaine) to Crill 4 (Span 80) was also prepared. An oil-surfactant phase was then prepared by mixing the oil and surfactant phases at a ratio of 6.8:2.2. The following preparations were added to the oils/surfactant: 15-20 mg/ml salicylic acid, 5-15 mg/ml ginger extract, 5-10 mg/ml resveratrol powder, 2-6 mg/ml tetrahydrocurcumin powder. Microemulsions were formed by mixing the oil-surfactant phase with one ninth - one tenth volume of water and assisted by gentle shaking and / or heating the mixture to 50 degrees Celsius.
EXAMPLE 4 Preparation of a Pharmaceutical Composition with a Water-in~Oil Microemulsion
An oil phase consisting of a 3:1 ratio of Crodamol GTCC to Capmul MCM C8 was prepared. A surfactant/co-surfactant phase consisting of an 8:5:2 ratio of Crillet 4 (Tween 80) to Dehyton AB30 (coco-betaine; Cognis) to Crill 4 (Span 80) was also prepared. An oil/surfactant phase was thus prepared by mixing the oil and surfactant / co-surfactant phases at a ratio of 6.8:2.2. Microemulsions were formed by mixing the oil-surfactant phase with one ninth - one tenth volume of water. One or more of the following agents were dissolved in the microemulsions: Pueraria Mirifica root extract (to yield a final concentration of 8-10 mg/ml; Smith Naturals), Gatulene Expression (to yield a final concentration of 50 mg/ml; Trapeze), Dermican (to yield a final concentration of 30 mg/ml; Cognis) and Vialox (to yield a final concentration of 5 mg/ml; Pentapharm). Microemulsion formation was often assisted by gentle shaking and / or heating the mixture to 50 degrees Celsius.
EXAMPLE 5 Preparation of a Pharmaceutical Composition with a Water-in-Oil Microemulsion
A 3:1 ratio of Crodamol GTCC to Capmul MCM C8 was prepared (oils). A second surfactant mixture of an 8:5:1 ratio of Crillet 4 (Tween 80) to Dehyton AB30 (coco- betaine, Cognis) to cocamide MEA (Cognis) was also prepared. The oil/surfactant phase was prepared by mixing the oil with surfactant at a ratio of 6.8:2.2. Microemulsions were formed by mixing the oil-surfactant phase with one ninth - one tenth volume of water. One or more of the following agents were dissolved in the microemulsions: Eyeliss (to yield a final concentration of 10-35 mg/ml; Croda), Haloxyl (to yield a final concentration of 15- 28 mg/ml; Croda) and Esculoside (to yield a final concentration of 1-4 mg/ml; Croda). Microemulsion formation was often assisted by gentle shaking and / or heating the mixture to 50 degrees Celsius.
EXAMPLE 6 Transdermal Delivery of125 Labeled Proteins Applied in a Microemulsion
Microemulsions are prepared in accordance with one of the above-mentioned microemulsion formulation Examples 1 to 5, wherein the active agents include a 125I- labeled-protein. Suitable proteins may include BSA, epidermal growth factor or a recombinant human TNFR chimeric protein such as that described in PCT Application No. WO 06/079176. Ionodation can be performed using a standard method known in the art. The microemulsions are then applied to a 2 x 1 cm area of the shaved skin of Balb/C mice. The microemulsion is left on the mice and after 180 minutes or overnight the mice are euthanased and all the organs removed and counted in a gamma counter to determine the distribution of 125I-labeled-proteins. It is anticipated this data will show that the microemulsion is capable of delivering the applied water-soluble protein to the muscle and adjacent skin tissue.
EXAMPLE 7
Preparation of Pharmaceutical Compositions containing Anti-Inflammatory Molecules in a Microemulsion
One or more agents selected from the list of: Vitamin B 12 analogs, including cyanocobalamin, hydroxycobalamin, cobalamin, adenosyl-cobalamin (all obtained from Sigma Pharmaceuticals), MLIF (synthesized by Auspep, Melbourne, Australia), tetrahydrocurcumin, recombinant human TNF receptor II - IgGl Fc chimera molecules, for example, molecules of which the production and purification is described in PCT Application No. WO 06/079176; are dissolved in the appropriate phase (depending upon the specific polar characteristics of the molecule) prior to the formation of separate microemulsions as described above in the microemulsion formulation examples. The final concentrations range of each ingredient in the subsequent microemulsions is 2-25 mg/ml.
EXAMPLE 8
Topical Treatment of Inflammation in Mice Using a Topical Composition of the Present Invention containing one or more Anti-Inflammatory Molecules
One of the standard models of acute or chronic inflammation is induced in animals of the appropriate species. For example, acute inflammation is induced in the rear right footpad of C57/B16 strain mice by an intra-pad injection of 50 μl of 1% llambda Carrageenan. Immediately following injection of Carrageenan into the foot-pad, mice receive treatment, which consisted of the application of 20 microL of atopical composition containing one or more anti-inflammatory molecules, as described above, applied to a previously shaved area of the belly of the mouse using a flat applicator. The thickness of the Carageenan-injected foot and left rear foot (non-injected control foot) is measured with a constant pressure vernier calliper at regular timepoints for a total of 14 days. The topical compositions are anticipated to be active in reducing inflammation within the right rear foot. Alternatively, a chronic model of inflammation such as the collagen joint model could be used to test the efficacy of a topical composition of the present invention containing one or more anti- inflammatory molecules. Expected results would, in that case, be similar to those anticipated for an acute inflammatory model such as the Carrageenan model.
EXAMPLE 9
Treatment of a D ermalogical Disorder Using a Topical Pharmaceutical Composition of the Present Invention
The individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans. The clinical trial is subjected to rigorous controls to ensure that individuals are not unnecessarily put at risk and that they are fully informed about their role in the study. To account for the psychological effects of receiving treatments, volunteers are randomly assigned to topical placebo or topical treatment groups. Furthermore, to prevent the doctors from being biased in treatments, they are not informed as to whether the medication they are administering is the topical pharmaceutical composition or topical placebo. Using this randomization approach, each volunteer has the same chance of being given either the new treatment or the placebo.
Volunteers receive either a topical phaπnaceutical composition, for example, one of those described above in Examples 3 or 7 or topical placebo for an appropriate period, for example, a single 0.8ml application on one psoriatic lesion (total skin area of 20 cm2) with a 7 day follow-up or, alternatively, multiple 0.8ml applications on the same target lesion 9 times (every second day) over a 21 day period. Biological parameters associated with the indicated disease state or condition, for example, psoriasis, will be measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period. Such measurements include the levels of TNF alpha in body fluids, tissues or organs compared to pre-treatment levels. Other measurements include, but are not limited to, indices of the disease state or condition being treated, body weight, blood pressure, serum titers of pharmacologic indicators of disease or toxicity as well as ADME (absorption, distribution, metabolism and excretion) measurements.
Information recorded for each patient includes age (years), gender, height (cm), family history of disease state or condition (yes/no), motivation rating (some/moderate/great) and number and type of previous treatment regimens for the indicated disease or condition.
Volunteers taking part in this study are adults aged 18 to 65 years and exhibit a particular dermalogical disorder, for example, psoriatic skin lesions. Additionally, roughly an equal number of males and females participate in the study. Volunteers with certain characteristics are equally distributed for topical placebo and topical pharmaceutical composition.
Evaluation of treatment is graded by an appropriate method, for example, the grading of treatment into one of four categories, namely, cured, obviously effective, effective and non-effective. "Cured" is where the inflammatory area on the plaque is diminished completely and the pruritus disappeared. "Obviously effective" is where the inflammatory area on the plaque is diminished by more than 60% and the pruritus is slighted and softened. "Effective" is where the inflammatory area on the plaque is diminished by 20 to 60% and the pruritus is slighted and softened. "Non-effective" is where the inflammatory area on the plaque is diminished by less than 20 % or there is exacerbation of psoriasis.
Alternatively, treatment evaluation is graded by the Local Plaque Severity Index (LPSI), whereby each target plaque is assessed and rated for erthema, induration and dessquamation using a five-point scale by the supervising clinician at the time of the specified clinic visits. An example of an appropriate clinical visit timetable for the "multiple application" treatment regime is on days 0, 11 and 21. The five-point scale is defined as follows: 0 = no symptoms; 1 = slight; 2 = moderate; 3 = marked; 4 = very marked. Scores for erthema, induration and desquamation are totalled. LPSI ranges from 0 to 12 with the higest score representing the more severe disease state.
In general, the volunteers treated with topical placebo have little or no response to treatment, whereas the volunteers treated with a topical pharmaceutical composition of the present invention show positive trends in their disease state or condition index at the conclusion of the study. In particular, the topical pharmaceutical composition of the present invention is obviously effective on most patients in the treatment group. No visible side-effects are observed. EXAMPLE 10
Topical vaccination of Mice using a Microemulsion of the Present Invention
Microemulsions for use as topical vaccines include a proteinaceous or non-proteinaceous antigen with or without an adjuvant. For example, ten mice per group are vaccinated with tenanus toxoid (TT; 1.8 limit of flocculation units, 20 meg; # 832-081, CSL, Melbourne, Australia) either by injection of 0.05 ml into the quadriceps muscle with a 30 gauge needle or topical application of 0.04 ml of microemulsion containing TT, using microemulsion formulation methods as described above in the examples 1 to 5, to a 2 cm2 patch of shaved skin on the belly. The control group receives a mock vaccination of saline alone. Identical control and test groups are used in both the C57Black and BALB/C strains of mice. Mice are vaccinated every two weeks for a total of three vaccinations. Blood samples are taken by orbital bleeds 2 weeks post-vaccination. Indirect ELISA assay is used to determine the titre of serum IgG specific to tetanus toxoid using an anti-mouse whole IgG antibody alkaline phosphatase conjugate (Sigma Cat No. A3562). Cell mediated immunity is measured by the delayed type hypersensitivity reaction. The right footpad of vaccinated or control mice are injected with 10 meg of tetanus toxoid in 0.05 ml of saline and the left footpad is injected with saline only as a control. The increase in footpad swelling of the right footpad is measured 24, 48 and 72 hours post-injection using a caliper.
It is anticipated that topical administration of antigen in a microemulsion will generate both an antibody and cellular response similar to that achieved when antigen is administered via traditional vaccination methods. A similar increase in efficacy of topical vaccination is anticipated where a microemulsion of the present invention contains one or more adjuvants selected from the following list of protein adjuvants selected from the list of: muramyl dipeptide, recombinant human C-type lectins such as CD209L, CD209L-Fc, DC-SIGN, DC-SIGN-Fc, CLEC4K and CD 207. In such cases, adjuvants are mixed at a suitable ratio, for example, a range of 1:1 to 1:5: of antigen : adjuvant protein. EXAMPLE 11 Weight loss microemulsion
A microemulsion for topical weight loss is prepared according to the methods as described above in the microemulsion formulation examples 1-5, wherein the active agents are selected from the list of: insulin, insulin-like growth factor 1 (IGFl), GHRP-6 at a final concentration range of 0.5 to 10 mg/ml. A standardised volume of microemulsion is applied daily (TD) to an area of shaved skin on the belly of conscious Swiss strain mice. A standardised volume of each of insulin, or insulin-like growth factor 1 (IGFl) or GHRP-6 at a final concentration range of 0.5 to 10 mg/ml in saline is injected sub cutaneously (SC) into the belly region of Swiss strain mice. Animal weights are measured 3 times per week. Following 2 weeks of treatment the mice are euthanased and their tissues removed and weighed. The weight of the animals with time is plotted was and a decrease in body weight of animals treated with the topical insulin is anticipated. Examination of organ weights is expected to show the greatest change in the topically applied insulin group, where the weight of skin is significantly reduced whilst muscle mass increases.
EXAMPLE 12
Treatment of Intradermal Melanoma in Mice Using a Topical Pharmaceutical Composition of the Present Invention
Mice (Balb/C and C57/B1) are injected intradermally or sub-cutaneously with 1 x 106 cells of the Bl 6 melanoma line. The tumour is allowed to grow until it is approximately 0.5 gm in mass. The mice are then treated topically with in a microemulsion, for example, a microemulsion as described above in the microemulsion formulation examples 1 to 5 wherein the active agents include one or more of the following chemotherapeutic agents: 5 -fluorouracil (10 mg/ml, 20 microlitre dose, 2 times per day); Carboplatin (10 mg/ml, 20 microlitre dose, 2 times per day); Cis-Platinum (10 mg/ml, 20 microlitre dose, 2 times per day); or a control microemulsion without chemotherapeutic agents. After 4 days of treatment, there is anticipated to be a reduction in growth rate of the tumour but no reduction in body weight of the animals. EXAMPLE 13
Treatment of Carcinoma Using a Topical Pharmaceutical Composition of the Present
Invention
The individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.
Preferably to account for the psychological effects of receiving treatments, the trial is conducted in a double-blinded fashion. Volunteer patients at a tertiary care center are randomly assigned to placebo or treatment groups. Furthermore, to prevent the doctors from being biased in treatments, they are not informed as to whether the medication they are administering is the topical pharmaceutical composition of the present invention or a placebo composition. Using this randomization approach, each volunteer has the same chance of being given either the new treatment or the placebo treatment.
Volunteers presenting with, for example, cutaneous melanoma, receive either the topical pharmaceutical composition of the present invention, for example, as described above in the microemulsion formulation examples 1 to 5 wherein the active agents include one or more of the following anti-cancer agents: 5-fluorouracil, carboplatin, oxaliplatin, cis-platin, selenomethionine, chlorotoxin, L49 monoclonal antibody, MAb-LM609, antileukinate, gold-monophosphine and gold-diphosphine, pyridoxal isonicotinoyl hydrazone (PIH), amino-substituted platinum complexes, paclitaxol, defoxamine, synthetic retinoids such as tretinoin, isotretinoin, and etretinate, imiquimod (e.g. 5%), calcitriol, temozolomide, interferon beta, betulinic acid (e.g. 20%), a curcuminoid derivative, TSA, or topical placebo composition, for an appropriate period thereafter with biological parameters associated with the condition being measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period. Such measurements include the levels of each pharmaceutical agent in the composition in body fluids, tissues or organs compared to pre-treatment levels. Other measurements include, but are not limited to, evidence of metastisis in tissues such as lymph nodes. Still other measurements include, but are not limited to body weight, blood pressure, serum titers of pharmacologic indicators of disease such as specific disease indicators or toxicity as well as ADME (absorption, distribution, metabolism and excretion) measurements.
Information recorded for each patient includes age (years), gender, height (cm), family history of disease state or condition (yes/no), motivation rating (some/moderate/great) and number and type of previous cancer treatment regimens.
Volunteers taking part in this study are adults aged 18 to 65 years. Volunteers with certain characteristics are equally distributed for topical placebo treatment and the topical pharmaceutical composition of the present invention treatment. In general, at the conclusion of the study the volunteers topically treated with the placebo composition have little or no response to treatment, whereas the volunteers treated with the topical pharmaceutical composition of the present invention show positive trends in one or more of the following parameters: overall survival (OS), disease-free survival (DFS).
EXAMPLE 14
Gene Therapy Treatment of Cancer Using a Topical Pharmaceutical Composition of the Present Invention containing Nucleic Acid Molecules
Recent methods indicate that cancerous disease states associated with the expression of abnormal common fusion genes can be treated by the specific silencing of those genes via the administration of siRNA (Hashimoto et al. PNAS 101(17): 6647-6652, 2004).
The individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.
Preferably to account for the psychological effects of receiving treatments, the trial is conducted in a double-blinded fashion. Volunteers presenting with a cancerous condition, for example, chronic myeloid leukemia, are randomly assigned to placebo or treatment groups. Furthermore, to prevent the doctors from being biased in treatments, they are not informed as to whether the medication they are administering is the topical pharmaceutical composition of the present invention or a topical placebo. Using this randomization approach, each volunteer has the same chance of being given either the new treatment or the placebo.
Volunteers receive either the topical pharmaceutical composition of the present invention, for example, a microemulsion as described above in the microemulsion formulation Examples 1 to 5 , wherein the active agents consist of siRNAs directed against one or more of the two types of the common fusion genes present in chronic myeloid leukemia patients (Hashimoto et ah, supra) or placebo for an appropriate period with biological parameters associated with the indicated disease state being measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period. Such measurements include the levels of pharmaceutical agent in the composition in body fluids, tissues or organs compared to pre-treatment levels. Other measurements include, but are not limited to, counts of abnormal white cells, other specific tests for detecting trace leukemia cells, body weight, blood pressure, serum titers of pharmacologic indicators of disease such as specific disease indicators or toxicity as well as ADME (absorption, distribution, metabolism and excretion) measurements.
Information recorded for each patient includes age (years), gender, height (cm), family history of disease state or condition (yes/no), motivation rating (some/moderate/great) and number and type of previous treatment regimens for the indicated disease or condition.
Volunteers taking part in this study are adults aged 18 to 65 years and roughly an equal number of males and females participate in the study. Volunteers with certain characteristics are equally distributed for placebo and the topical pharmaceutical composition of the present invention treatment. In general, the volunteers treated with placebo have little or no response to treatment, whereas the volunteers treated with the topical pharmaceutical composition of the present invention show positive trends in one or more of the following parameters: decreased counts of abnormal white cells, overall survival (OS), disease-free survival (DFS) at the conclusion of the study.
EXAMPLE 15 Treatment of Neuropathic or Nociceptic Pain Using a Topical Pharmaceutical
Composition of the Present Invention containing Analgesic Agents
The individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.
Preferably to account for the psychological effects of receiving treatments, the trial is conducted in a double-blinded fashion. Volunteers presenting with either neuropathic or nociceptic pain are randomly assigned to placebo or treatment groups. Furthermore, to prevent the doctors from being biased in treatments, they are not informed as to whether the medication they are administering is the topical pharmaceutical composition of the present invention or a topical placebo. Using this randomization approach, each volunteer has the same chance of being given either the new treatment or the placebo.
Volunteers receive either the topical pharmaceutical composition of the present invention, for example, a microemulsion as described above in the formulation examples 1 to 5 , wherein the active agents consist of one or more known anti-inflammatory molecules, optionally containing one or more analgesic agents such as lidocaine (4%) or placebo for an appropriate period with biological parameters associated with the indicated disease state or condition being measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period.
Such measurements include the levels of each pharmaceutical agent in the composition in body fluids, tissues or organs compared to pre-treatment levels. Other measurements include, but are not limited to, indices of the disease state or condition being treated, body weight, blood pressure, serum titers of pharmacologic indicators of disease such as specific disease indicators or toxicity as well as ADME (absorption, distribution, metabolism and excretion) measurements. Information recorded for each patient includes age (years), gender, height (cm), family history of disease state or condition (yes/no), motivation rating (some/moderate/great) and number and type of previous treatment regimens for the indicated disease or condition.
Volunteers taking part in this study are adults aged 18 to 65 years and roughly an equal number of males and females participate in the study. Volunteers with certain characteristics are equally distributed for placebo and the topical pharmaceutical composition of the present invention treatment. In general, the volunteers treated with placebo have little or no response to treatment, whereas the volunteers treated with the topical pharmaceutical composition of the present invention show an amelioration of pain intensity at the conclusion of the study.
EXAMPLE 16 Treatment of Delayed Onset Muscle Soreness (DOMS) Using a Topical Pharmaceutical
Composition of the Present Invention
The individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.
Preferably to account for the psychological effects of receiving treatments, the trial is conducted in a double-blinded fashion. Volunteers are randomly assigned to placebo or treatment groups. Furthermore, to prevent the participating clinicians from being biased in treatments, they are not informed as to whether the treatment being administering is the topical pharmaceutical composition of the present invention or a topical placebo composition. Using this randomization approach, each volunteer has the same chance of being given either the new treatment or the placebo.
Volunteers take part in a suitable exercise protocol to induce lower limb fatigue, for example, repetitive sets of jumping movements on an inclined sled apparatus on three separate days with a minimum of one week rest between each test session (presentation by S. Hughes at Australian Conference of Science and Medicine in Sport, Yanuca, Fiji, October, 2006).
Volunteers receive a controlled application of either the topical pharmaceutical composition of the present invention, for example, a microemulsion as described above in the formulation examples 1 to 5 , wherein the active agents are one or more anti- inflammatory moleculesor topical placebo composition applied to the lower limbs at regular time-points after the initial exercise protocol for a total of 1-14 days.
Physical and biological parameters associated with DOMS are measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period. Suitable physical parameters include the level of performance between initial and follow-up test sessions of the exercise protocol. Suitable biological parameters include the levels of pharmaceutical agent or agents of the composition in body fluids, tissues or organs compared to pre-treatment levels. Other measurements include, but are not limited to, blood lactate levels, body weight, blood pressure, serum titers of pharmacologic indicators of DOMS such as creatinine levels or toxicity as well as ADME (absorption, distribution, metabolism and excretion) measurements.
Information recorded for each patient includes age (years), gender, height (cm), family history of disease state or condition (yes/no), motivation rating (some/moderate/great) and number and type of previous treatment regimens for DOMS.
Volunteers taking part in this study are adults aged 18 to 65 years and roughly an equal number of males and females participate in the study. Volunteers with certain characteristics are equally distributed for topical placebo composition and topical pharmaceutical composition of the present invention treatments. In general, at the conclusion of the study, volunteers treated with the topical pharmaceutical composition of the present invention show positive trends in one or more of the above-mentioned physical or biological parameters in comparison to volunteers treated with the topical placebo composition.
EXAMPLE 17 Detection of Sentinel Lymph Nodes in Patients with an Identified Primary Tumour
Using a Topical Pharmaceutical Composition of the Present Invention
The individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.
Preferably to account for the psychological effects of receiving treatments, the trial is conducted in a double-blinded fashion. Volunteer patients at a tertiary care center are randomly assigned to placebo or treatment groups. Furthermore, to prevent the doctors from being biased in treatments, they are not informed as to whether the medication they are administering is the topical pharmaceutical composition of the present invention or a placebo composition. Using this randomization approach, each volunteer has the same chance of being given either the new treatment or the placebo treatment.
Volunteers presenting with a tumour, for example, a cutaneous melanoma, receive either the topical pharmaceutical composition of the present invention, for example, a microemulsion as described above in the microemulsion formulation Examples 1 to 5 wherein the active agent is an imaging agent is a known imaging compound such as 99mTc chelated to DTPA, or topical placebo composition, applied to the tumour. At an appropriate time after the application of the composition, the body region is imaged for the detection of imaging agent. It is anticipated that the accumulation of imaging agent within the sentinel lymph node will allow for a rapid identification and subsequent removal of the lymph node. Other measurements include, but are not limited to body weight, blood pressure, serum titers of pharmacologic indicators of disease such as specific disease indicators or toxicity as well as ADME (absorption, distribution, metabolism and excretion) measurements. Information recorded for each patient includes age (years), gender, height (cm), family history of disease state or condition (yes/no), motivation rating (some/moderate/great) and number and type of previous cancer treatment regimens.
Volunteers taking part in this study are adults aged 18 to 65 years. Volunteers with certain characteristics are equally distributed for topical placebo treatment and the topical pharmaceutical composition of the present invention treatment. In general, at the conclusion of the study the volunteers treated with the topical pharmaceutical composition of the present invention, when compared with the volunteers topically treated with the placebo composition, show positive trends in one or more of the following parameters: incidence of metastisis, overall survival (OS), disease-free survival (DFS).
EXAMPLE 18
Sub Cutaneous Fat Loss in Volunteers following Topical Application of the Pharmaceutical Composition of the Present Invention containing Insulin
The individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.
Preferably to account for the psychological effects of receiving treatments, the trial is conducted in a double-blinded fashion. Volunteers are randomly assigned to placebo or treatment groups. Furthermore, to prevent the doctors from being biased in treatments, they are not informed as to whether the medication they are administering is the topical pharmaceutical composition of the present invention, or a placebo. Using this randomization approach, each volunteer has the same chance of being given either the new treatment or the placebo.
Volunteers receive either the topical pharmaceutical composition of the present invention, for example, a microemulsion as described above in the microemulsion formulation examples 1 to 5 wherein the active agents include one or more of the following: insulin
(final concentration of 0.5-2 mg/ml), insulin-like growth factor 1 (IGFl) or GHRP-6 or placebo onto a suitable part of the body, for example, the anterior and posterior aspects of the upper arm for an appropriate period with particular physical and biological parameters associated with the condition being measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period. Suitable physical measurements include skinfold caliper measurements or A- and/or B-mode ultrasound measurements of the thickness of the subcutaneous fat layer overlying the upper arm. Suitable biological measurements include blood levels of insulin and glucose in body fluids, tissues or organs compared to pre-treatment levels. Other measurements include, but are not limited to, body weight and blood pressure as well as ADME (absorption, distribution, metabolism and excretion) measurements.
Information recorded for each patient includes age (years), gender, height (cm), family history of condition (yes/no), motivation rating (some/moderate/great) and number and type of previous treatment regimens for the indicated condition.
Volunteers taking part in this study are adults aged 18 to 65 years and roughly an equal number of males and females participate in the study. Volunteers with certain characteristics are equally distributed for placebo and the topical pharmaceutical composition of the present invention treatment. At the conclusion of the study, the volunteers treated with the topical pharmaceutical composition of the present invention show reduced thickness of sub cutaneous fat deposits of the upper arm, as determined by one or more of the following methods: skinfold caliper measuremen, A-mode ultrasound measurement, B-mode ultrasound measurement; whereas the volunteers treated with placebo have little or no response to treatment. It is anticipated that blood levels of insulin, insulin-like growth factor 1 (IGFl) or GHRP-6 are not affected by treatment with the topical pharmaceutical composition of the present invention. EXAMPLE 19
In vivo determination of physical and biological parameters of the skin of human subjects following topical application of a Pharmaceutical Composition with a Water-in-
OiI Microemulsion
The individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which include humans.
A standardized volume of a topical formulation of the present invention selected from Examples 4 or 5 or a placebo formulation is applied to an appropriate area of the body, for example, the forehead or the back of the hand of volunteers in a prescribed fashion for an appropriate period, for example, 6 weeks with biological parameters associated with each skin area being measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period. Such measurements include :
(1) the measurement of skin capacitance using a suitable instrument (for example, a Corneometer CM 825; Courage Khazaka Electronic, KoIn, Germany);
(2) the measurement of skin conductance using a suitable instrument (for example, a Skicon-200; ISBS Co., Hamamatsu, Japan);
(3) the measurement of skin surface topography using suitable methods and instruments (for example, the generation of skin replicas of the skin area followed by analysis with a suitable instrument, for example, an optical profϊlometer, a laser profilemeter and/or a transparency profϊlometer);
(4) the measurement of skin surface topography using high resolution photography using a suitable instruments and software analysis (for example, the Visia System (Proctor & Gamble), the Primos System (GF Messtechnik GmbH, Germany), the
Leica Stereo Explorer; (5) the measurement of skin elasticity using a suitable instrument (for example, a Cutometer SEM 575 skin elasticity meter).
(6) the measurement of skin color using high resolution photography using a suitable instruments (for example, a Minolta skin analysis camera), software analysis and standardized color palettes (for example, the Pantone [Registered Trade Mark] system).
Information recorded for each patient includes age (years), gender, motivation rating (some/moderate/great) and number and type of previous treatment skin regimens.
Volunteers taking part in this study are adults aged 18 to 65 years. Volunteers treated with a topical formulation of the present invention show positive "anti-ageing" parameters in one or more physical or biological parameters, including skin capacitance, skin conductance, and parameters associated with skin surface topography such as wrinkle frequency, wrinkle depth, roughness and smoothing, and altered "skin-lightening" parameters such as skin color, skin brightness, time taken for pigment fade, at the conclusion of the study compared with the baseline measurements taken at the commencement of the study.
EXAMPLE 20
In vivo determination of physical and biological parameters of the facial skin of human subjects following topical application of a Pharmaceutical Composition of the Present Invention and various commercially available anti-ageing formulations
The individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.
Preferably to account for the psychological effects of receiving treatments, the trial is conducted in a double-blinded fashion. Volunteers are randomly assigned to placebo or treatment groups. Furthermore, to prevent the researchers from being biased in treatments, they are not informed as to whether the treatments they are administering is a pharmaceutical composition of the present invention or a placebo formulation. Using this randomization approach, each volunteer has the same chance of being given either the topical formulation of the present invention or the placebo formulation.
A standardized volume of either the topical formulation of the present invention selected from Examples 3, 4 or 5 or a placebo formulation is applied to the face of volunteers in a prescribed fashion for an appropriate period, for example, 6 weeks with biological parameters associated with the facial skin being measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period. Such measurements include:
(1) the measurement of facial skin capacitance using a suitable instrument (for example, a Corneometer CM 825; Courage Khazaka Electronic, KoIn, Germany);
(2) the measurement of facial skin conductance using a suitable instrument (for example, a Skicon-200; ISBS Co., Hamamatsu, Japan);
(3) the measurement of skin surface topography using suitable methods and instruments (for example, the generation of skin replicas of the facial area followed by analysis with a suitable instrument, for example, an optical profilometer, a laser profilometer and/or a transparency profilometer);
(4) the measurement of skin surface topography using high resolution photography using a suitable instruments and software analysis (for example, the Visia System (Proctor & Gamble), the Primos System (GF Messtechnik GmbH, Germany), the Leica Stereo Explorer;
(5) the measurement of facial skin elasticity using a suitable instrument (for example, a Cutometer SEM 575 skin elasticity meter). (6) the measurement of skin color using high resolution photography using a suitable instruments (for example, a Minolta skin analysis camera), software analysis and standardized color palettes (for example, the Pantone [Registered Trade Mark] system).
Information recorded for each patient includes age (years), gender, motivation rating (some/moderate/great) and number and type of previous treatment regimens for facial skin.
Volunteers taking part in this study are adults aged 18 to 65 years. Volunteers with certain characteristics are equally distributed for the topical formulation of the present invention and placebo formulation treatment. In general, the volunteers treated with placebo formulation have little or no improvement in one or more physical or biological parameters, including facial skin capacitance, facial skin conductance, parameters associated with skin surface topography such as wrinkle frequency, wrinkle depth, roughness and smoothing, and altered "skin-lightening" parameters such as skin color, skin brightness, time taken for pigment fade, whereas the volunteers treated with a topical formulation of the present invention show positive "anti-ageing" parameters in one or more of the above-mentioned physical or biological parameters at the conclusion of the study.
EXAMPLE 21
Transdermal delivery of agents to treat erectile dysfunction
The water-in-oil microemulsion is used to formulate agents to treat erectile dysfunction.
In one embodiment the microemulsion comprises a glycerol trinitrate and sildenafil citrate. In particular, the agent relaxes the smooth muscle in the corpus cavernosum of the penis. The agents contemplated therein include 1) nitric oxide donors which includes nitroglycerin, alkyl nitrites, such as amyl nitrite and L-arginine; 2) The phosphodiesterase inhibitors sildenafil (Viagra), Tadalafil (Cialis), Vardenafil (Levitra) and any other cGMP specific phosphodiesterase type 5 inhibitors; and 3) The adenyl cyclase activators, prostaglandin El (Alprostidil) and forskolin.
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.
BIBLIOGRAPHY
Atherton and Shephard Chapter 9 entitled "Peptide Synthesis" by which is included in a publication entitled "Synthetic Vaccines"
Escribano et al. European Journal of Pharmaceutical Sciences 79:203-210, 2003
Hashimoto et al. PNAS 101(17): 6647-6652, 2004
Kreilgaard et al. Journal of Controlled Release 69:421-433, 2000
Li et al. International Journal of Pharmaceutics 237:77-85, 2002
S. Hughes at Australian Conference of Science and Medicine in Sport, Yanuca, Fiji, October, 2006
Santus et al. J Control Release 25:1-20, 1993
Shahrokh et al. American Chemical Society, 1997
Sintov et al. Journal of Controlled Release 95: 173-183, 2004
Valenta et al. Journal of Controlled Release 95:257- 265, 2004
WO 06/079176

Claims

CLAIMS:
1. A microemulsion comprising a water phase; an oil phase; an active ingredient; one or more zwitterionic surfactants; and one or more co-surfactants.
2. The microemulsion of Claim 1 for use as a topical delivery formulation.
3. The microemulsion of Claim 2 wherein the topical delivery is transdermal delivery.
4. The microemulsion of Claim 3 wherein the active ingredient is a protein.
5. The microemulsion of Claim 3 wherein the active ingredient is an antibody, Fc fusion molecule, imaging agent, anti-cancer agent, re-pigmentation agent or an agent listed in Table 1.
6. A method for the transdermal delivery of active agents comprising (a) preparing a water phase, including dissolving one or more selected active agents in water; (b) preparing an oil phase; (c) emulsifying the water phase and the oil phase using one or more zwitterionic surfactants and one or more co-surfactants, thereby forming a topical pharmaceutical composition; and (d) applying the topical pharmaceutical composition on a subject.
7. The method of Claim 6 wherein the active agent is selected from anti-inflammatory agents, proteins, antibodies, Fc fusion molecules, soluble receptors, vaccines, imaging agents, anti-cancer agents, skin lightening including whitening agents, UV blocking agents, agents to reduce or nullify nitric oxide, agents useful for the treatment of erectile dysfunction; agents for increasing local circulation, agents for the amelioration of facial and skin ageing, agents for the treatment of obesity, re-pigmentation agents, genetic molecules such as RNAi, DNA, virosomes and anti-sense RNA and agents useful in treating muscle dystrophy.
8. The method of Claim 6 or 7 wherein the subject is human.
9. The method of Claim 8 wherein the subject is a non-human animal.
10. A method of producing a water-in-oil microemulsion comprising preparing an oil phase, preparing a surfactant/co-surfactant phase and then preparing an oil-surfactant/co- surfactant phase.
11. The method of Claim 10 wherein the oil phase comprises crodamol GTCC and Capmul MCM C8.
12. The method of Claim 10 wherein the surfactant/co-surfactant phase comprises Tween 80, Span 80 and Coco-betaine.
13. The method of Claim 10 or 11 or 12 wherein the oil-surfactant/co-surfactant phase is in a ratio of from about 5:1 to about 10:2, respectively.
14. The method of Claim 13 wherein the oil-surfactant/co-surfactant phase is a ratio of about 7.6:1.4, respectively.
15. The method of Claim 10 further comprising an active agent selected from antiinflammatory agents, proteins, antibodies, Fc fusion molecules, soluble receptors, vaccines, imaging agents, anti-cancer agents, skin lightening including whitening agents, UV blocking agents, agents to reduce or nullify nitric oxide, agents useful for the treatment of erectile dysfunction; agents for increasing local circulation, agents for the amelioration of facial and skin ageing, agents for the treatment of obesity, re-pigmentation agents, genetic molecules such as RNAi, DNA, virosomes and anti-sense RNA and agents useful in treating muscle dystrophy.
16. Use of an oil-in- water microemulsion in the manufacture of a transdermal delivery formulation.
17. A method of treating erectile dysfunction comprising the transdermal delivery of an erectile dysfunction treating agent in the microemulsion of Claim 1.
18. A method of Claim 17 wherein the agent is sildenafil citrate.
PCT/AU2008/000908 2007-06-20 2008-06-20 Transdermal delivery of active agents WO2008154705A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
AU2007903316 2007-06-20
AU2007903316A AU2007903316A0 (en) 2007-06-20 Transdermal delivery of active agents

Publications (1)

Publication Number Publication Date
WO2008154705A1 true WO2008154705A1 (en) 2008-12-24

Family

ID=40155827

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2008/000908 WO2008154705A1 (en) 2007-06-20 2008-06-20 Transdermal delivery of active agents

Country Status (1)

Country Link
WO (1) WO2008154705A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103169641A (en) * 2013-03-14 2013-06-26 金红叶纸业集团有限公司 Acne-removing and skin-clearing composition and tissue applying same
US8835509B2 (en) 2010-05-31 2014-09-16 Arbro Pharmaceuticals Ltd. Self emulsifying drug delivery system for a curcuminoid based composition
CN112280765A (en) * 2020-09-27 2021-01-29 扬州大学 Application of sulfobetaine surfactant in improving bromelinase activity
US10980865B2 (en) 2012-08-10 2021-04-20 Aquavit Pharmaceuticals, Inc. Direct application system and method for the delivery of bioactive compositions and formulations

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999060997A1 (en) * 1998-05-22 1999-12-02 Hewlett Healthcare Limited Pharmaceutical compositions comprising an amphoteric surfactant an alkoxylated cetyl alcohol and a polar drug
WO2001028555A1 (en) * 1999-10-18 2001-04-26 Lipocine, Inc. Emulsion compositions for polyfunctional active ingredients
US20050266085A1 (en) * 2004-05-28 2005-12-01 Warner Kevin S Gelled emulsion and microemulsion formulations for dermal drug delivery
WO2006024095A1 (en) * 2004-08-31 2006-03-09 Connetics Australia Pty Ltd Microemulsion & sub-micron emulsion process & compositions
US20060109575A1 (en) * 2004-11-22 2006-05-25 Bogdan Radu Vehicle mirror assembly

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999060997A1 (en) * 1998-05-22 1999-12-02 Hewlett Healthcare Limited Pharmaceutical compositions comprising an amphoteric surfactant an alkoxylated cetyl alcohol and a polar drug
WO2001028555A1 (en) * 1999-10-18 2001-04-26 Lipocine, Inc. Emulsion compositions for polyfunctional active ingredients
US20050266085A1 (en) * 2004-05-28 2005-12-01 Warner Kevin S Gelled emulsion and microemulsion formulations for dermal drug delivery
WO2006024095A1 (en) * 2004-08-31 2006-03-09 Connetics Australia Pty Ltd Microemulsion & sub-micron emulsion process & compositions
US20060109575A1 (en) * 2004-11-22 2006-05-25 Bogdan Radu Vehicle mirror assembly

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8835509B2 (en) 2010-05-31 2014-09-16 Arbro Pharmaceuticals Ltd. Self emulsifying drug delivery system for a curcuminoid based composition
US10980865B2 (en) 2012-08-10 2021-04-20 Aquavit Pharmaceuticals, Inc. Direct application system and method for the delivery of bioactive compositions and formulations
CN103169641A (en) * 2013-03-14 2013-06-26 金红叶纸业集团有限公司 Acne-removing and skin-clearing composition and tissue applying same
CN103169641B (en) * 2013-03-14 2016-01-13 金红叶纸业集团有限公司 The light print compositions of anti-acne and apply its napkin
CN112280765A (en) * 2020-09-27 2021-01-29 扬州大学 Application of sulfobetaine surfactant in improving bromelinase activity
CN112280765B (en) * 2020-09-27 2021-10-08 扬州大学 Application of sulfobetaine surfactant in improving bromelinase activity

Similar Documents

Publication Publication Date Title
WO2007070983A1 (en) Transdermal delivery of pharmaceutical agents
KR101238703B1 (en) Carrier for enteral administration
US8685440B2 (en) Nanoliposome using esterified lecithin and method for preparing the same, and composition for preventing or treating skin diseases comprising the same
FI118296B (en) Oral liquid alendronate compositions
US6558712B1 (en) Delivery system for pharmaceutical, nutritional and cosmetic ingredients
MX2007015949A (en) A carrier comprising one or more di and/or mono-(electron transfer agent) phosphate derivatives or complexes thereof.
TWI228512B (en) Transcobalamin binding conjugates useful for treating abnormal cellular proliferation
Veerareddy et al. Formulation and evaluation of oil-in-water emulsions of piperine in visceral leishmaniasis
WO2008154705A1 (en) Transdermal delivery of active agents
KR950000770B1 (en) Calcitonin-containing emulsion for nasal administration
Itin et al. Controlled delivery of apomorphine through buccal mucosa, towards a noninvasive administration method in Parkinson's disease: a preclinical mechanistic study
JP6196847B2 (en) Anti-containing composition
CN1294990C (en) Composition of nanometer SOD and ginseng or its extract and its preparation method
WO2021077111A1 (en) Compositions and methods for delivering cannabinoids using microneedle devices to the skin
WO2019122220A1 (en) Liposomes comprising sphingomyelin
Lu et al. Oral ionic liquid for transdermal delivery and obesity treatment
JP2002543209A (en) Antioxidant vitamin B6 analog
JP2010260808A (en) Oral medicinal composition for improving skin condition
CN101247834A (en) A carrier comprising one or more di and/or mono-(electron transfer agent) phosphate derivatives or complexes thereof
CN109700853A (en) A kind of composition and its application in the drug of preparation prevention and treatment epilepsy
RU2434643C2 (en) Transporting filler, which contains one or more di- and/or mono-(electronic transmitting agent) phosphate derivatives or their compounds
RU2026688C1 (en) Method of preparation preparing for directed delivery of antitumor drugs in malignant cell
CN109803547A (en) Composition and its application method containing apo- aequorin and vitamin D
CN108719998A (en) A kind of oral diet supplement medicament for treating osteoarthritis
Balasubramaniyam et al. Development of Ethosomal Gel Loaded with Terminalia chebula for Effective Treatment of Arthritis

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08756988

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08756988

Country of ref document: EP

Kind code of ref document: A1