WO2004080478A1 - Therapeutic and prophylactic compositions and uses therefor - Google Patents

Therapeutic and prophylactic compositions and uses therefor Download PDF

Info

Publication number
WO2004080478A1
WO2004080478A1 PCT/AU2004/000300 AU2004000300W WO2004080478A1 WO 2004080478 A1 WO2004080478 A1 WO 2004080478A1 AU 2004000300 W AU2004000300 W AU 2004000300W WO 2004080478 A1 WO2004080478 A1 WO 2004080478A1
Authority
WO
WIPO (PCT)
Prior art keywords
rel
receptor
acid
tnf
compound
Prior art date
Application number
PCT/AU2004/000300
Other languages
French (fr)
Inventor
George Varigos
Steve Gerondakis
Raffi Gugasyan
Original Assignee
The Walter And Eliza Hall Institute Of Medical Research
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Walter And Eliza Hall Institute Of Medical Research filed Critical The Walter And Eliza Hall Institute Of Medical Research
Publication of WO2004080478A1 publication Critical patent/WO2004080478A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/191Tumor necrosis factors [TNF], e.g. lymphotoxin [LT], i.e. TNF-beta
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents

Definitions

  • the present invention relates generally to compositions and their use in the treatment and/or prophylaxis of inflammatory conditions in an animal such as a mammal including a human. More particularly, the compositions comprise agents which potentiate the function of the Rel/NF- ⁇ B signal transduction pathway. The compositions may also comprise agents which inhibit aspects of the immune system. Alternatively, multiple compositions may be employed which separately activate the NF- ⁇ B signaling pathway and inhibit systemically or locally an immune response.
  • the present invention further contemplates methods of treatment and/or prophylaxis in an animal such as a mammal including a human by the administration of a potentiator of Rel/NF- ⁇ B signal transduction or a composition comprising such a potentiator, optionally together with an inhibitor of an immune response.
  • a potentiator of Rel/NF- ⁇ B signal transduction or a composition comprising such a potentiator optionally together with an inhibitor of an immune response.
  • Such potentiators are useful in the treatment or prophylaxis of inflammatory conditions such as but not limited to conditions affecting non-immune organs such as the skin, heart, kidney, liver, brain, pancreas, gall bladder, lung, adrenal gland, gastrointestinal tract, amongst other tissues.
  • Rel/NF- ⁇ B family of transcription factors are expressed as dimeric proteins comprising related subunits including c-Rel, RelA, RelB, F- ⁇ Bl and NF- ⁇ B2.
  • the Rel/NF- ⁇ B signaling pathways are conserved and have been implicated in the control of gene expression in many key physiological process including cell growth, division, survival and differentiation.
  • Rel/NF- ⁇ B family members are thought to induce the transcription of genes involved in mediating stress responses as well as innate and adaptive immunity.
  • Rel/NF- ⁇ B signaling is well suited to mediate such responses which optimally require rapid deployment because the Rel/NF- ⁇ B signaling pathway exists in a latent form within the cell and signaling can be rapidly induced.
  • Rel/NF- ⁇ B transcription factors are retained in a latent state in the cytoplasm through association with inhibitory proteins (I ⁇ B proteins).
  • I ⁇ B proteins A specific kinase complex (IKK complex) is activated in response to a wide range of signals and phosphorylates the I ⁇ B, targeting it for degradation.
  • the released Rel/NF- ⁇ B transcription factors may then be transported to the nucleus where they recognize and bind to specific sequences in the regulatory region of target genes.
  • target genes include regulators of immune and inflammatory response such as inflammatory cytokines (i.e. TNF, IL-2, IL- 6, IL-8), cytokine receptors (i.e. IL-2R ⁇ ), chemokines, chemokine receptors, adhesion molecules, free radical regulatory molecules, innate immune regulatory proteins such as defensins and the like.
  • Rel/NF ⁇ B signaling pathways has been implicated in most if not all immune-mediated pathogenesis including diseases such as asthma, multiple sclerosis, inflammatory bowel disease and diabetes.
  • the present invention provides, therefore, potentiators of the Rel/NF- ⁇ B signal transduction pathway for use in the treatment and prophylaxis of inflammation or inflammatory conditions.
  • the inflammatory condition is in a non-immune organ such as the skin, heart, kidney, liver, brain, pancreas, gall bladder, lung, adrenal gland, gastrointestinal tract, amongst other tissues.
  • the potentiators are conveniently in a composition comprising the potentiator and one or more pharmaceutically acceptable carriers, diluents and/or excipients.
  • compositions or two- or multi-part pharmaceutical compositions comprising at least one potentiator of the Rel/NF- ⁇ B pathway and at least one inhibitor of an immune response or an immune component or portion of an inhibitor of an immune response pathway.
  • a potentiator of the Rel/NF- ⁇ B pathway may be a chemical agent such as a chemical molecule or peptide, polypeptide or protein or chemical analogs thereof or may be a genetic agent such as a sense or antisense molecule, ribozyme, DNAzyme or ribonuclease- type complex.
  • potentiators of Rel/NF- ⁇ B signaling and compositions comprising same of the present invention may be used systemically or locally such as topically.
  • Figure 1 is a photographic representation showing embryonic death resulting from the combined loss of NF-kB 1 and RelA or c-Rel and RelA is overcome by blocking TNF-a signalling.
  • B-D H&E stained sections of dorsal skin from El 8 fetuses.
  • FIG. 2 is a photographic representation showing c-rel mRNA is expressed in fetal skin.
  • a c-rel cRNA probe was incubated with sections of El 8.5 skin.
  • Panels A and C are dark- field images of the corresponding bright-field images in panels B and D.
  • Sense probe controls are shown in panels C and D.
  • E, F silver grains overlay the sheath cells (arrow in E) and the dermal papilla (arrowhead in E) of hair follicles, plus basal cells (arrow in F) and differentiating keratinocytes in the epidermis (bracket in E).
  • HF hair follicle
  • SC sheath cells
  • DP dermal papilla
  • BL basal layer
  • K keratinocytes.
  • Scale bars 43 ⁇ m in A, B, C, D and 22 ⁇ m in E, F. Color versions of this Figure are available from the patentee.
  • Figure 3 is a photographic representation showing Hair follicle morphogenesis and tooth development is impaired in the absence of c-Rel and RelA.
  • A-C Hair follicle development in control (rel ⁇ +/+c-rel-/-tnf a-/-) and mutant (rel ⁇ -Z-crel-/- tnf a-/-) fetal sections stained with H&E (scale bar: 425 ⁇ m).
  • A E16: Hair placodes (tylotrich) are evident in control (black arrows) but not mutant skin.
  • Figure 4 is a photographic representation showing epidermal differentiation is impaired in rela-/-c-rel-/-tnf a-/- embryos.
  • Biochemical markers were used to analyse keratinocyte differentiation in control (rela+/+c-rel-/-tnf a-/-) and mutant (rela-/-c-rel-/-tnf a-/-) El 8 dorsal skin.
  • H&E stained sections of (A) control and (B) mutant dorsal skin depict the basal layer (B), the stratum spinosum (S), the stratum granulosum (G) and stratum corneum (C) (scale bar: 225 ⁇ m).
  • FIG. 5 is a graphical representation showing Flow cytometric analysis of epidermal basal cells lacking c-Rel and RelA.
  • Basal keratinocytes from El 8 control (rela+/-c-rel-/- tnf a-/-) and mutant (rela-/ -c-rel-/ -tnf a-/-) fetuses were stained with a FITC conjugated anti-integrin-bl (shaded histogram) or control anti-CD4 (open histogram) antibody (A).
  • B Two-colour FACS analysis was performed by staining with FITC conjugated anti-integrin- a6 antibody and PE-conjugated anti-CD71 antibody.
  • FIG. 1 Three phenotypically distinct populations were identified by flow cytometry: quiescent stem cells, a6hi CD711o (gate 1); transit amplifying cells, a ⁇ hi CD71hi (gate 2) and post-mitotic differentiating cells, a ⁇ lo CD71hi (gate 3). Cells with high CD71 expression in the control profile (asterisk) were markedly reduced in the mutant.
  • Figure 6 is a graphical and photographic represenation showing cell cycle defect in TA cells lacking RelA and c-Rel.
  • Figure 7 is a photographic representation showing keratinocytes lacking RelA and c-Rel fail to form colonies in culture. Equivalent numbers of viable keratinocytes (106) were grown for 7 days.
  • Control (rel ⁇ +/+c-rel-/-tnf a-/-) and mutant (rel ⁇ -/-c-rel-/-tnf a-/-) keratinocyte cultures were stained with anti-keratin- 14 (+) or isotype matched negative control (-) antibodies.
  • Keratinocytes of differing genotypes (rel ⁇ +/+c-rel+/+tnf a-/-, rel ⁇ +/+c-rel-/-tnf a-/-, rel ⁇ -/-c-rel+/+tnf a-/-, rel ⁇ -/-c-rel-/-tnf a-/-) axe, shown by phase contrast. Color versions of this Figure are available from the patentee.
  • Figure 8 is a graphical representation showing relA/c-rel deficient skin grafts develop
  • Ci-iv or B6r ⁇ g-l-/-tnfa-/- mice (Di-v).
  • A Appearance of representative donor skin grafts over a 4 week period.
  • B, C Histological analysis of skin grafts on ⁇ rag-1-/- recipients. H&E stained sections from control (Bi) and mutant (Bii) skin grafts were analysed under low power at 4 weeks (scale bar: l ⁇ OO ⁇ m). Staining for K6, was confined to hair follicles in the control graft (Biii) and the epidermis in the mutant graft (Biv) (Scale bar: 425 ⁇ m).
  • Figure 9 is a schematic representation of how Rel/NF- ⁇ B transcription factors are activated by two distinct pathways engaged by distinct extracellular signals.
  • the "classical” pathway involves the activation of cytoplasmic c-Rel and RelA complexes via the IKK ⁇ /IKK ⁇ -dependent phosphorylation and degradation of I ⁇ B- -like proteins.
  • the "alternative” pathway involves the activation of NF- ⁇ B2/RelB complexes via the IKK ⁇ dependent phosphorylation and degradation of NF- ⁇ B2 pi 00.
  • the present invention is predicated in part on the surprising elucidation that the Rel/NF- ⁇ B signal transduction pathway needs to be up-regulated in certain organs or tissues in order to prevent or at least reduce inflammatory responses.
  • the inflammatory response being prevented or treated affects a non-immune organ such as but not limited to the skin, heart, kidney, liver, brain, pancreas, gall bladder, lung, adrenal gland, gastrointestinal tract, amongst other tissues.
  • a non-immune organ such as but not limited to the skin, heart, kidney, liver, brain, pancreas, gall bladder, lung, adrenal gland, gastrointestinal tract, amongst other tissues.
  • Reference to the "gastrointestinal tract” includes the oesophagus, stomach and small and large bowel.
  • the present invention provides, therefore, potentiators of the Rel/NF- ⁇ B signal transduction pathway and compositions comprising the potentiators.
  • the potentiators may elevate levels of NF- ⁇ B or various components of the Rel/NF- ⁇ B pathway directly or via inhibition of components which antagonize NF- ⁇ B function such as IKBS.
  • compound used interchangeably herein to refer to a chemical compound 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.
  • compound When the terms “compound”, “active 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.
  • compound is not to be construed as a chemical compound only but extends to peptides, polypeptides and proteins as well as genetic molecules such as RNA, DNA and chemical analogs thereof.
  • potentiator is an example of a compound, active agent, pharmacologically active agent, medicament, active and drug which up-regulates the Rel/NF- ⁇ B signal transduction pathway.
  • up- regulates encompasses inducing function of the pathway to correspondingly reduce an inflammatory response or the risk of an inflammatory response being elicited.
  • the present invention contemplates, therefore, compounds useful in up-regulating the function or activity of the Rel/NF- ⁇ B signaling pathway.
  • the compounds have an effect on reducing or preventing or treating inflammatory conditions such as in non-immune organs including skin, heart, kidney, liver, brain, pancreas, gall bladder, lung, adrenal gland, gastrointestinal tract, amongst other tissues.
  • Reference to a "compound”, “active agent”, “pharmacologically active agent”, “medicament”, “active” and “drug” includes combinations of two or more actives such as a potentiator of Rel/NF- ⁇ B signaling and an inhibitor of an immune response or immune response pathway.
  • a “combination” also includes a two- or multi-part pharmaceutical composition where the agents are provided separately and given or dispensed separately or admixed together prior to dispensation.
  • the terms "effective amount” and “therapeutically effective amount” of an agent as used herein mean a sufficient amount of the agent to provide the desired therapeutic or physiological effect.
  • an "effective Rel/NF- ⁇ B potentiating amount” of an agent is a sufficient amount of the agent to directly or indirectly up-regulate the function of the Rel/NF- ⁇ B signaling pathway.
  • agents acting as an agonist of signaling pathway components by the agents which are or mimic components of the signaling pathway, by agents which induce the pathway via cellular receptors or by the agents antagonizing inhibitors of signaling componets.
  • 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.
  • 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 emusifying agents, pH buffering agents, preservatives, and the like.
  • a "pharmacologically acceptable" salt, ester, emide, 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, elimination of symptoms and/or underlying cause, prevention of the occurrence of symptoms and/or their underlying cause, and improvement or remediation of damage.
  • “treating” a patient involves prevention of a particular disorder or adverse physiological event in a susceptible individual as well as treatment of a clinically symptomatic individual by inhibiting or causing regression of a disorder or disease.
  • a condition or disorder is an inflammatory response or mediates or facilitates an inflammatory response or is a downstream product of an inflammatory response.
  • the present method of "treating" a patient with an inflammatory condition or with a propensity for one to develop encompasses both prevention of the condition, disease or disorder as well as treating the condition, disease or disorder.
  • the present invention contemplates the treatment or prophylaxis of any inflammatory-type condition.
  • Inflammatory conditions contemplated herein include but are not limited to skin inflammatory conditions such as psoriasis, ichthyosis, pityriasis, rubra pilaris, seborrhoea, keloids, keratoses, neoplasias and scleroderma, warts, benign growths and cancers of the skin.
  • inflammatory conditions contemplated herein are those induced by microorganism and viruses such as enteropathogenic E. coli., Gardnerella vaginalis, Helicobacter plylori, Lactobacilli, Listeria monocytogenes, Micoplasma fermentans, Mycobacteria tuberculosis, Neisseria gonorrhoeae, Rickettsia ricettsii, Salmonella dublin, Salmonella typhimurium, Shigella flexneri, Staphyllococcus aureus, Rhodobacter sphaeroides, adenovirus, cytomegalovirus, ⁇ pstein-Barr virus ( ⁇ BV), hepatitis B virus, herpes virus saimiri, human herpesvirus 6, HIV-1, herpex simplex virus- 1, HTLV-1, influenza virus, measles virus, molony murine leukemia virus, Newcastle disease virus, respiratory syncytial virus, Sendai para
  • ⁇ ukaryotic organisms causing inflammation include Theileria pai-va.
  • Other inflammatory conditions include those caused or exacerbated by drug abuse or use of ethical drugs and physical stress including radiation exposure.
  • Inflammatory conditions exacerbated by immunological mediators are also contemplated such as multiple sclerosis, asthma, systemic lupus, nephritis, diabetes, Chrohn's disease and other inflammatory bowel diseaess, arthritis, atherosclerosis and autoimmune myocarditis.
  • Patient refers to an animal, preferably a mammal and more preferably 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 patient regardless of whether a human or non-human animal may be referred to as an individual, subject, 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.
  • an "animal” includes an avian species such as a poultry bird, an aviary bird or game bird.
  • the compounds of the present invention may be large or small molecules, nucleic acid molecules (including antisense or sense molecules), peptides, polypeptides or proteins or hybrid molecules such as RNAi- or siRNA-complexes, ribozymes or DNAzymes.
  • the compounds may need to be modified so as to facilitate entry into a cell. This is not a requirement if the compound interacts with an extracellular receptor.
  • the preferred animals are humans or other primates, livestock animals, laboratory test animals, companion animals or captive wild animals.
  • laboratory test animals examples 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 and donkeys. Non-mammalian animals such as avian species, zebrafish, amphibians (including cane toads) and Drosophila species such as Drosophila melanogaster are also contemplated.
  • the present invention provides, therefore, drugs which potentiate, activate or otherwise up- regulate the Rel/NF- ⁇ B signaling pathway including agents which antagonize inhibitors of NF- ⁇ B such as I ⁇ Bs.
  • the present invention contemplates, therefore, methods of screening for drugs comprising, for example, contacting a candidate drug with an Rel/NF- ⁇ B pathway component including an extracellular receptor.
  • a candidate drug with an Rel/NF- ⁇ B pathway component including an extracellular receptor.
  • Such a molecule is referred to herein as a "target” or “target molecule”.
  • the screening procedure includes assaying (i) for the presence of a complex between the drug and the target, or (ii) an alteration in the expression levels of nucleic acid molecules encoding the target.
  • One form of assay involves competitive binding assays. In such competitive binding assays, the target is typically labeled. Free target is separated from any putative complex and the amount of free (i.e. uncomplexed) label is a measure of the binding of the agent being tested to target molecule.
  • Another technique for drug screening provides high throughput screening for compounds having suitable binding affinity to a target and is described in detail in Geysen (International Patent Publication No. WO 84/03564). Briefly stated, large numbers of different small peptide test compounds are synthesized on a solid substrate, such as plastic pins or some other surface. The peptide test compounds are reacted with a target and washed. Bound target molecule is then detected by methods well known in the art. This method may be adapted for screening for non-peptide, chemical entities. This aspect, therefore, extends to combinatorial approaches to screening for target antagonists or agonists.
  • Purified target can be coated directly onto plates for use in the aforementioned drug screening techniques.
  • non-neutralizing antibodies to the target may also be used to immobilize the target on the solid phase.
  • the present invention also contemplates the use of competitive drug screening assays in which neutralizing antibodies capable of specifically binding the target compete with a test compound for binding to the target or fragments thereof. In this manner, the antibodies can be used to detect the presence of any peptide which shares one or more antigenic determinants of the target.
  • Yet another useful source of compounds is a chemically modified cytokine or other activator of an extracellular receptor which then in turn activates the Rel/NF- ⁇ B.
  • Analogs contemplated herein include but are not limited to modification to side chains, incorporating of unnatural amino acids and/or their derivatives during peptide, polypeptide or protein synthesis and the use of crosslinkers and other methods which impose conformational constraints on the proteinaceous molecule or their analogs.
  • side chain modifications contemplated by the present invention include modifications of amino groups such as by reductive alkylation by reaction with an aldehyde followed by reduction with NaBH 4 ; amidination with mefhylacetimidate; acylation with acetic anhydride; carbamoylation of amino groups with cyanate; trinitrobenzylation of amino groups with 2, 4, 6-trinitrobenzene sulphonic acid (TNBS); acylation of amino groups with succinic anhydride and tetrahydrophthalic anhydride; and pyridoxylation of lysine with pyridoxal-5-phosphate followed by reduction with NaBH 4 .
  • amino groups such as by reductive alkylation by reaction with an aldehyde followed by reduction with NaBH 4 ; amidination with mefhylacetimidate; acylation with acetic anhydride; carbamoylation of amino groups with cyanate; trinitrobenzylation of amino groups with 2, 4, 6-trinitrobenzene sulphonic acid (TN
  • the guanidine group of arginine residues may be modified by the formation of heterocyclic condensation products with reagents such as 2,3-butanedione, phenylglyoxal and glyoxal.
  • the carboxyl group may be modified by carbodiimide activation via O-acylisourea formation followed by subsequent derivitization, for example, to a corresponding amide.
  • Sulphydryl groups may be modified by methods such as carboxymethylation with iodoacetic acid or iodoacetamide; performic acid oxidation to cysteic acid; formation of a mixed disulphides with other thiol compounds; reaction with maleimide, maleic anhydride or other substituted maleimide; formation of mercurial derivatives using 4- chloromercuribenzoate, 4-chloromercuriphenylsulphonic acid, phenylmercury chloride, 2- chloromercuri-4-nitrophenol and other mercurials; carbamoylation with cyanate at alkaline pH.
  • Tryptophan residues may be modified by, for example, oxidation with N- bromosuccinimide or alkylation of the indole ring with 2-hydroxy-5-nitrobenzyl bromide or sulphenyl halides.
  • Tyrosine residues on the other hand, may be altered by nitration with tetranitromethane to form a 3-nitrotyrosine derivative.
  • Modification of the imidazole ring of a histidine residue may be accomplished by alkylation with iodoacetic acid derivatives or N-carbethoxylation with diethylpyrocarbonate .
  • Examples of incorporating unnatural amino acids and derivatives during peptide synthesis include, but are not limited to, use of norleucine, 4-amino butyric acid, 4-amino-3- hydroxy-5-phenylpentanoic acid, 6-aminohexanoic acid, t-butylglycine, norvaline, phenylglycine, ornithine, sarcosine, 4-amino-3-hydroxy-6-methylheptanoic acid, 2-thienyl alanine and/or D-isomers of amino acids.
  • a list of unnatural amino acid, contemplated herein is shown in Table 1.
  • Non-conventional Code Non-conventional Code amino acid amino acid
  • D- ⁇ -methylcysteine Dmcys N-(4-aminobutyl)glycine Nglu D- ⁇ -methylglutamine Dmgln N-(2-aminoethyl)glycine Naeg
  • peptides can be conformationally constrained by, for example, incorporation of C ⁇ and N ⁇ -methylamino acids, introduction of double bonds between C ⁇ and Cp atoms of amino acids and the formation of cyclic peptides or analogs by introducing covalent bonds such as forming an amide bond between the N and C termini, between two side chains or between a side chain and the N or C terminus.
  • one aspect of the present invention contemplates any compound which binds or otherwise interacts with a component of the Rel/NF- ⁇ B pathway resulting in potentiation, activation or general up-regulation of the Rel/NF- ⁇ B signaling transduction pathway.
  • the present invention is also useful for screening for other compounds which reduce expression of a gene encoding an NF- ⁇ B inhibitor or which up-regulates expression of genes encoding Rel/NF- ⁇ B components such as NF- ⁇ B itself or a receptor linked to the Rel/NF- ⁇ B pathway.
  • targets may be used in any of a variety of drug screening techniques, such as those described herein and in International Publication No. WO 97/02048.
  • a target antagonist or agonist includes a variant of the target molecule.
  • the target is a polypeptide.
  • polypeptide refers to a polymer of amino acids and its equivalent and does not refer to a specific length of the product, thus, peptides, oligopeptides and proteins are included within the definition of a polypeptide. This term also does not exclude modifications of the polypeptide, for example, glycosylations, aceylations, phosphorylations and the like. Included within the definition are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids such as those given in Table 1) or polypeptides with substituted linkages.
  • the NF- ⁇ B pathway is potentiated via an external receptor such as BCR, IL-IR, TNFR, LTBR, CDl lb/CD18, CD28, CD2, CD35, CD3, CD40, CD4, fc-2a-receptor (IgG2a), Flt- 1 , Ly6A/E, N-CAM, trail receptor- 1 , trail-receptor-2 or trail-receptor-4.
  • an external receptor such as BCR, IL-IR, TNFR, LTBR, CDl lb/CD18, CD28, CD2, CD35, CD3, CD40, CD4, fc-2a-receptor (IgG2a), Flt- 1 , Ly6A/E, N-CAM, trail receptor- 1 , trail-receptor-2 or trail-receptor-4.
  • Rel/NF- ⁇ B signal transduction potentiators include IL-1, IL-2, IL-12, IL-15, IL-17, IL-18, LIF, THANK, TNF- ⁇ , TNF- ⁇ as well as any of the TNF-receptor superfamily ligands, 1- ⁇ -D-arabinofuranosyl-cytosine (ara-C), anthralin, azidothymidine (AZT), camptothecin, ciprofibrate, cisplatin, daunomycin, daunorubicin, doxorubicin, etoposide, haloperidol, methamphetamine, phenobarbital, tamoxifen, taxol (paclitaxel), vinblastine, vincristine, advanced glycated end products (AGEs), amyloid protein fragment ( ⁇ A4), maleylated BSA, modified (oxidized) LDL, bone morphogenic protein 2, bone morphogenic protein 4, folicle stimulating hormone
  • peptide mimetic Another useful group of compounds is a mimetic.
  • the terms "peptide mimetic”, “target mimetic” or “mimetic” are intended to refer to a substance which has some chemical similarity to the target but which antagonizes or agonizes or mimics the target.
  • a peptide mimetic may be a peptide-containing molecule that mimics elements of protein secondary structure (Johnson et al, "Peptide Turn Mimetics” in Biotechnology and Pharmacy, Pezzuto et al, Eds., Chapman and Hall, New York, 1993).
  • peptide mimetics The underlying rationale behind the use of peptide mimetics is that the peptide backbone of proteins exists chiefly to orient amino acid side chains in such a way as to facilitate molecular interactions such as those of antibody and antigen, enzyme and substrate or scaffolding proteins.
  • a peptide mimetic is designed to permit molecular interactions similar to the natural molecule.
  • Peptide or non- peptide mimetics may be useful, for example, to activate the NF- ⁇ B signaling pathway via an external receptor or enter the cell and agonize or antagonize a particular component.
  • the compounds of the present invention may be selected to interact with a target alone or single or multiple compounds may be used to affect multiple targets.
  • multiple Rel/NF- ⁇ B components may be targeted with or without certain immune response components being inhibited.
  • the target polypeptide or fragment employed in such a test may either be free in solution, affixed to a solid support, or borne on a cell surface.
  • One method of drug screening utilizes eukaryotic or prokaryotic host cells which are stably transformed with recombinant polynucleotides expressing the polypeptide or fragment, preferably in competitive binding assays. Such cells, either in viable or fixed form, can be used for standard binding assays.
  • One may measure, for example, the formation of complexes between a target or fragment and the agent being tested, or examine the degree to which the formation of a complex between a target or fragment and a known ligand is aided or interfered with by the agent being tested.
  • a substance identified as a modulator of target function or gene activity may be a peptide or non-peptide in nature.
  • Non-peptide "small molecules" are often preferred for many in vivo pharmaceutical uses. Accordingly, a mimetic or mimic of the substance (particularly if a peptide) may be designed for pharmaceutical use.
  • the designing of mimetics to a pharmaceutically active compound is a known approach to the development of pharmaceuticals based on a "lead" compound. This might be desirable where the active compound is difficult or expensive to synthesize or where it is unsuitable for a particular method of administration, e.g. peptides are unsuitable active agents for oral compositions as they tend to he quickly degraded by proteases in the alimentary canal.
  • Mimetic design, synthesis and testing is generally used to avoid randomly screening large numbers of molecules for a target property.
  • the mimetic is of a cytokine or other molecule which interacts with a cell surface receptor and stimulates the Rel/NF- ⁇ B pathway.
  • the pharmacophore Once the pharmacophore has been found, its structure is modeled according to its physical properties, e.g. stereochemistry, bonding, size and/or charge, using data from a range of sources, e.g. spectroscopic techniques, x-ray diffraction data and NMR. Computational analysis, similarity mapping (which models the charge and/or volume of a pharmacophore, rather than the bonding between atoms) and other techniques can be used in this modeling process.
  • a range of sources e.g. spectroscopic techniques, x-ray diffraction data and NMR.
  • Computational analysis, similarity mapping which models the charge and/or volume of a pharmacophore, rather than the bonding between atoms
  • other techniques can be used in this modeling process.
  • the three-dimensional structure of the ligand and its binding partner are modeled. This can be especially useful where the ligand and/or binding partner change conformation on binding, allowing the model to take account of this in the design of the mimetic. Modeling can be used to generate inhibitors which interact with the linear sequence or a three-dimensional configuration.
  • a template molecule is then selected onto which chemical groups which mimic the pharmacophore can be grafted.
  • the template molecule and the chemical groups grafted onto it can conveniently be selected so that the mimetic is easy to synthesize, is likely to be pharmacologically acceptable, and does not degrade in vivo, while retaining the biological activity of the lead compound.
  • the mimetic is peptide-based
  • further stability can be achieved by cyclizing the peptide, increasing its rigidity.
  • the mimetic or mimetics found by this approach can then be screened to see whether they have the target property, or to what extent they exhibit it. Further optimization or modification can then be carried out to arrive at one or more final mimetics for in vivo or clinical testing.
  • the goal of rational drug design is to produce structural analogs of biologically active polypeptides of interest or of small molecules with which they interact (e.g. agonists, antagonists, inhibitors or enhancers) in order to fashion drugs which are, for example, more active or stable forms of the polypeptide, or which, e.g. enhance or interfere with the function of a polypeptide in vivo. See, e.g. Hodgson (Bio/Technology 9: 19-21, 1991).
  • one first determines the three-dimensional structure of a protein of interest (e.g. NF- ⁇ B or a receptor or a component of the Rel/NF- ⁇ B pathway) by x-ray crystallography, by computer modeling or most typically, by a combination of approaches.
  • Useful information regarding the structure of a polypeptide may also be gained by modeling based on the structure of homologous proteins.
  • An example of rational drug design is the development of HIV protease inhibitors (Erickson et al, Science 249: 527- 533, 1990).
  • target molecules may be analyzed by an alanine scan (Wells, Methods Enzymol. 202: 2699-2705, 1991).
  • an amino acid residue is replaced by Ala and its effect on the peptide 's activity is determined.
  • Each of the amino acid residues of the peptide is analyzed in this manner to determine the important regions of the peptide. It is also possible to isolate a target-specific antibody, selected by a functional assay and then to solve its crystal structure.
  • this approach yields a pharmacore upon which subsequent drug design can be based. It is possible to bypass protein crystallography altogether by generating anti-idiotypic antibodies (anti-ids) to a functional, pharmacologically active antibody. As a mirror image of a mirror image, the binding site of the anti-ids would be expected to be an analog of the original receptor. The anti-id could then be used to identify and isolate peptides from banks of chemically or biologically produced banks of peptides. Selected peptides would then act as the pharmacore.
  • anti-ids anti-idiotypic antibodies
  • Two-hybrid screening is also useful in identifying other members of a biochemical or genetic pathway associated with a target.
  • Two-hybrid screening conveniently uses Saccharomyces cerevisiae and Saccharomyces pombe.
  • Target interactions and screens for inhibitors can be carried out using the yeast two-hybrid system, which takes advantage of transcriptional factors that are composed of two physically separable, functional domains.
  • the most commonly used is the yeast GAL4 transcriptional activator consisting of a DNA binding domain and a transcriptional activation domain.
  • Two different cloning vectors are used to generate separate fusions of the GAL4 domains to genes encoding potential binding proteins.
  • the fusion proteins are co-expressed, targeted to the nucleus and if interactions occur, activation of a reporter gene (e.g. lacZ) produces a detectable phenotype.
  • a reporter gene e.g. lacZ
  • S. cerevisiae is co-transformed with a library or vector expressing a cDNA GAL4 activation domain fusion and a vector expressing a an Rel/NF- ⁇ B pathway component fused to GAL4.
  • lacZ is used as the reporter gene
  • co- expression of the fusion proteins will produce a blue color.
  • Small molecules or other candidate compounds which interact with a target will result in loss of color of the cells.
  • the present invention extends to a genetic approach to up-regulating expression of an Rel/NF- ⁇ B pathway component (including a receptor) or down-regulating an inhibitor of NF- ⁇ B.
  • nucleic acid molecules which encode a signaling component are used to elevate levels of the component.
  • nucleic acid molecules which induce temporary or permanent gene silencing of NF- ⁇ B inhibitors may also be used.
  • nucleic acids include RNA, cDNA, genomic DNA, synthetic forms and mixed polymers, both sense and antisense strands, and may be chemically or biochemically modified or may contain non-natural or derivatized nucleotide bases, as will be readily appreciated by those skilled in the art.
  • modifications include, for example, labels, methylation, substitution of one or more of the naturally occurring nucleotides with an analog (such as the morpholine ring) internucleotide modifications such as uncharged linkages (e.g.
  • synthetic molecules that mimic polynucleotides in their ability to bind to a designated sequence via hydrogen binding and other chemical interactions. Such molecules are known in the art and include, for example, those in which peptide linkages substitute for phosphate linkages in the backbone of the molecule.
  • Antisense polynucleotide sequences are useful in silencing transcripts of NF- KB inhibitors such as IKBS.
  • polynucleotide vectors containing all or a portion of a target NF- ⁇ B inhibitor gene locus may be placed under the control of a promoter in an antisense orientation and introduced into a cell. Expression of such an antisense construct within a cell will interfere with target transcription and/or translation.
  • co- suppression and mechanisms to induce RNAi or siRNA may also be employed.
  • antisense or sense molecules may be directly administered. In this latter embodiment, the antisense or sense molecules may be formulated in a composition and then administered by any number of means to target cells.
  • morpholinos are oligonucleotides composed of morpholine nucleotide derivatives and phosphorodiamidate linkages (for example, Summerton and Weller, Antisense and Nucleic Acid Drug Development 7: 187-195, 1997). Such compounds are injected into embryos and the effect of interference with mRNA is observed.
  • the present invention employs compounds such as oligonucleotides and similar species for use in modulating the function or effect of nucleic acid molecules encoding an NF- ⁇ B inhibitor, i.e. the oligonucleotides induce transcriptional or post- transcriptional gene silencing.
  • the oligonucleotides induce transcriptional or post- transcriptional gene silencing. This is accomplished by providing oligonucleotides which specifically hybridize with one or more nucleic acid molecules encoding the inhibitor.
  • the oligonucleotides may be provided directly to a cell or generated within the cell.
  • target nucleic acid and “nucleic acid molecule encoding an NF- ⁇ B inhibitor” have been used for convenience to encompass DNA encoding the inhibitor, RNA (including pre-mRNA and mRNA or portions thereof) transcribed from such DNA, and also cDNA derived from such RNA.
  • RNA including pre-mRNA and mRNA or portions thereof
  • cDNA derived from such RNA.
  • antisense The hybridization of a compound of the subject invention with its target nucleic acid is generally referred to as "antisense”.
  • antisense inhibition is typically based upon hydrogen bonding-based hybridization of oligonucleotide strands or segments such that at least one strand or segment is cleaved, degraded, or otherwise rendered inoperable. In this regard, it is presently preferred to target specific nucleic acid molecules and their functions for such antisense inhibition.
  • the functions of DNA to be interfered with can include replication and transcription.
  • Replication and transcription for example, can be from an endogenous cellular template, a vector, a plasmid construct or otherwise.
  • the functions of RNA to be interfered with can include functions such as franslocation of the RNA to a site of protein translation, franslocation of the RNA to sites within the cell which are distant from the site of RNA synthesis, translation of protein from the RNA, splicing of the RNA to yield one or more RNA species, and catalytic activity or complex formation involving the RNA which may be engaged in or facilitated by the RNA.
  • the result of such interference with target nucleic acid function is reduced levels of I ⁇ B.
  • modulation and modulation of expression mean either an increase (stimulation) or a decrease (inhibition) in the amount or levels of a nucleic acid molecule encoding the gene, e.g., DNA or RNA. Inhibition is often the preferred form of modulation of expression and mRNA is often a preferred target nucleic acid.
  • hybridization means the pairing of complementary strands of oligomeric compounds.
  • the preferred mechanism of pairing involves hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleoside or nucleotide bases (nucleobases) of the strands of oligomeric compounds.
  • nucleobases complementary nucleoside or nucleotide bases
  • adenine and thymine are complementary nucleobases which pair through the formation of hydrogen bonds.
  • Hybridization can occur under varying circumstances.
  • An antisense compound is specifically hybridizable when binding of the compound to the target nucleic acid interferes with the normal function of the target nucleic acid to cause a loss of activity, and there is a sufficient degree of complementarity to avoid non-specific binding of the antisense compound to non-target nucleic acid sequences under conditions in which specific binding is desired, i.e. under physiological conditions in the case of in vivo assays or therapeutic treatment, and under conditions in which assays are performed in the case of in vitro assays.
  • oligonucleotide and the further DNA, RNA, or oligonucleotide molecule are complementary to each other when a sufficient number of complementary positions in each molecule are occupied by nucleobases which can hydrogen bond with each other.
  • “specifically hybridizable” and “complementary” are terms which are used to indicate a sufficient degree of precise pairing or complementarity over a sufficient number of nucleobases such that stable and specific binding occurs between the oligonucleotide and a target nucleic acid.
  • compounds include antisense oligomeric compounds, antisense oligonucleotides, ribozymes, external guide sequence (EGS) oligonucleotides, alternate splicers, primers, probes, and other oligomeric compounds which hybridize to at least a portion of the target nucleic acid.
  • these compounds may be introduced in the form of single-stranded, double-stranded, circular or hairpin oligomeric compounds and may contain structural elements such as internal or terminal bulges or loops.
  • the compounds of the invention may elicit the action of one or more enzymes or structural proteins to effect modification of the target nucleic acid.
  • RNAse H a cellular endonuclease which cleaves the RNA strand of an RNA:DNA duplex. It is known in the art that single-stranded antisense compounds which are "DNA-like" elicit RNAse H. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of oligonucleotide-mediated inhibition of gene expression. Similar roles have been postulated for other ribonucleases such as those in the RNase III and ribonuclease L family of enzymes.
  • antisense compound is a single-stranded antisense oligonucleotide
  • dsRNA double-stranded RNA
  • oligomeric compound refers to a polymer or oligomer comprising a plurality of monomeric units.
  • oligonucleotide refers to an oligomer or polymer of ribonucleic acid
  • RNA deoxyribonucleic acid
  • DNA deoxyribonucleic acid
  • mimetics chimeras, analogs and homologs thereof.
  • This term includes oligonucleotides composed of naturally occurring nucleobases, sugars and covalent internucleoside (backbone) linkages as well as oligonucleotides having non-naturally occurring portions which function similarly.
  • modified or substituted oligonucleotides are often preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for a target nucleic acid and increased stability in the presence of nucleases.
  • oligonucleotides are a preferred form of the compounds of this invention, the present invention comprehends other families of compounds as well, including but not limited to oligonucleotide analogs and mimetics such as those herein described.
  • the open reading frame (ORF) or "coding region” which is known in the art to refer to the region between the translation initiation codon and the translation termination codon, is a region which may be effectively targeted. Within the context of the present invention, one region is the intragenic region encompassing the translation initiation or termination codon of the open reading frame (ORF) of a gene.
  • target regions include the 5' untranslated region (5'UTR), known in the art to refer to the portion of an mRNA in the 5' direction from the translation initiation codon, and thus including nucleotides between the 5' cap site and the translation initiation codon of an mRNA (or corresponding nucleotides on the gene), and the 3' untranslated region (3'UTR), known in the art to refer to the portion of an mRNA in the 3' direction from the translation termination codon, and thus including nucleotides between the translation termination codon and 3' end of an mRNA (or corresponding nucleotides on the gene).
  • 5'UTR 5' untranslated region
  • 3'UTR 3' untranslated region
  • the 5' cap site of an mRNA comprises an N7-methylated guanosine residue joined to the 5'-most residue of the mRNA via a 5'-5' triphosphate linkage.
  • the 5' cap region of an mRNA is considered to include the 5' cap structure itself as well as the first 50 nucleotides adjacent to the cap site. It is also preferred to target the 5' cap region.
  • eukaryotic mRNA transcripts are directly translated, many contain one or more regions, known as "introns", which are excised from a transcript before it is translated. The remaining (and, therefore, translated) regions are known as “exons” and are spliced together to form a continuous mRNA sequence.
  • Targeting splice sites i.e. intron- exon junctions or exon-intron junctions, may also be particularly useful in situations where aberrant splicing is implicated in disease, or where an overproduction of a particular splice product is implicated in disease. Aberrant fusion junctions due to rearrangements or deletions are also preferred target sites.
  • fusion transcripts mRNA transcripts produced via the process of splicing of two (or more) mRNAs from different gene sources are known as "fusion transcripts". It is also known that introns can be effectively targeted using antisense compounds targeted to, for example, DNA or pre-mRNA.
  • nucleoside is a base-sugar combination.
  • the base portion of the nucleoside is normally a heterocyclic base.
  • the two most common classes of such heterocyclic bases are the purines and the pyrimidines.
  • Nucleotides are nucleosides that further include a phosphate group covalently linked to the sugar portion of the nucleoside.
  • the phosphate group can be linked to either the 2', 3' or 5' hydroxyl moiety of the sugar.
  • the phosphate groups covalently link adjacent nucleosides to one another to form a linear polymeric compound.
  • linear compounds are generally preferred.
  • linear compounds may have internal nucleobase complementarity and may, therefore, fold in a manner as to produce a fully or partially double-stranded compound.
  • the phosphate groups are commonly referred to as forming the internucleoside backbone of the oligonucleotide.
  • the normal linkage or backbone of RNA and DNA is a 3' to 5' phosphodiester linkage.
  • oligonucleotides containing modified backbones or non-natural internucleoside linkages include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone.
  • modified oligonucleotides that do not have a phosphorus atom in their internucleoside backbone can also be considered to be oligonucleosides.
  • Preferred modified oligonucleotide backbones containing a phosphorus atom therein include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3 '-alkylene phosphonates, 5'-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3 '-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, selenophosphates and boranophosphates having normal 3 '-5' linkages, 2 '-5' linked analogs of these, and those having inverted polarity wherein one or more internucleotide linkages is a 3' to 3', 5' to 5' or 2'
  • Preferred oligonucleotides having inverted polarity comprise a single 3' to 3' linkage at the 3 '-most internucleotide linkage i.e. a single inverted nucleoside residue which may be abasic (the nucleobase is missing or has a hydroxyl group in place thereof).
  • Various salts, mixed salts and free acid forms are also included.
  • Antisense oligonucleotides are particularly useful in the treatment of inflammatory conditions of the skin.
  • the antisense oligonucleotides may target both the Rel/NF- ⁇ B pathway as well as the immune system. These can also be topically applied, generally in a cream-based composition.
  • genetic constructs including DNA vaccines are used to generate antisense molecules in vivo.
  • many of the preferred features described above are appropriate for sense nucleic acid molecules or for gene therapy applications to promote levels of Rel/NF- ⁇ B components.
  • Topical-based compositions are particularly useful in the treatment of inflammatory conditions of the skin.
  • an agent which potentiates the Rel/NF- ⁇ B signaling pathway may be manufactured and/or used in a preparation, i.e. in the manufacture or formulation or a composition such as a medicament, pharmaceutical composition or drug. These may be admimstered to individuals in a method of treatment or prophylaxis or regenerates therapy. Alternatively, they may be incorporated into a patch or slow release capsule or implant.
  • the present invention extends, therefore, to a pharmaceutical composition, medicament, drug or other composition including a patch or slow release formulation comprising an agonist or antagonist of target activity or gene expression.
  • Another aspect of the present invention contemplates a method comprising administration of such a composition to a patient such as for treatment or prophylaxis of an inflammatory condition.
  • the present invention contemplates a method of making a pharmaceutical composition comprising admixing a compound of the instant invention with a pharmaceutically acceptable excipient, vehicle or carrier, and optionally other ingredients. Where multiple compositions are provided, then such compositions may be given simultaneously or sequentially. Sequential administration includes administration within nanoseconds, seconds, minutes, hours or days. Preferably, within seconds or minutes.
  • Two- or multi-part pharmaceutical compositions or packs are also contemplated where multiple components such as comprising only those which potentiate the Rel/NF- ⁇ B pathway or both these latter components together with those which inhibit an immune response (locally or systemically).
  • Such multi-part pharmaceutical compositions or packs maintain different agents or groups of agents separately. These are either dispensed separately or admixed prior to being dispensed.
  • another aspect of the present invention contemplates a method for the treatment or prophylaxis of an inflammatory condition in an animal, said method comprising administering to said animal an effective amount of a compound as described herein or a composition comprising same.
  • administering to includes the topical application of a composition to target tissue such as skin.
  • the animal is a mammal such as a human or laboratory test animal such as a mouse, rat, rabbit, guinea pig, hamster, zebrafish or amphibian.
  • This method also includes providing a wild-type or mutant target gene function to a cell. This is particularly useful when generating an animal model. Alternatively, it may be part of a gene therapy approach. A target gene or a part of the gene may be introduced into the cell in a vector such that the gene remains extracl romosomal. In such a situation, the gene will be expressed by the cell from the extrachromosomal location. If a gene portion is introduced and expressed in a cell carrying a mutant target allele, the gene portion should encode a part of the target protein. Vectors for introduction of genes both for recombination and for extrachromosomal maintenance are known in the art and any suitable vector may be used. Methods for introducing DNA into cells such as elecfroporation calcium phosphate co-precipitation and viral transduction are known in the art.
  • Gene transfer systems known in the art may be useful in the practice of genetic manipulation. These include viral and non-viral transfer methods.
  • viruses have been used as gene transfer vectors or as the basis for preparing gene transfer vectors, including papovaviruses (e.g. SV40, Madzak et al, J. Gen. Virol. 73: 1533-1536, 1992), adenovirus (Berkner, Curr. Top. Microbiol. Immunol. 158: 39-66, 1992; Berkner et al, BioTechniques 6; 616-629, 1988; Gorziglia and Kapikian, J. Virol 66: 4407-4412, 1992; Quantin et al, Proc. Natl. Acad. Sci.
  • Non-viral gene transfer methods are known in the art such as chemical techniques including calcium phosphate co-precipitation, mechanical techniques, for example, microinjection, membrane fusion-mediated transfer via liposomes and direct DNA uptake and receptor-mediated DNA transfer.
  • Viral-mediated gene transfer can be combined with direct in vivo gene transfer using liposome delivery, allowing one to direct the viralvectors to particular cells.
  • the retroviral vector producer cell line can be injected into particular tissue. Injection of producer cells would then provide a continuous source of vector particles.
  • plasmid DNA of any size is combined with a polylysine-conjugated antibody specific to the adenovirus hexon protein and the resulting complex is bound to an adenovirus vector.
  • the trimolecular complex is then used to infect cells.
  • the adenovirus vector permits efficient binding, intemalization and degradation of the endosome before the coupled DNA is damaged.
  • Liposome/DNA complexes have been shown to be capable of mediating direct in vivo gene transfer. While in standard liposome preparations the gene transfer process is non-specific, localized in vivo uptake and expression have been reported in tumor deposits, for example, following direct in situ administration.
  • the polynucleotide encodes a sense or antisense polynucleotide or a ribozyme or DNAzyme
  • expression will produce the sense or antisense polynucleotide or ribozyme or DNAzyme.
  • expression does not require that a protein product be synthesized.
  • the vector also contains a promoter functional in eukaryotic cells.
  • the cloned polynucleotide sequence is under control of this promoter. Suitable eukaryotic promoters include those described above.
  • the expression vector may also include sequences, such as selectable markers and other sequences described herein.
  • Cells and animals which carry mutant target alleles (e.g. IKBS) or where one or both alleles are deleted can be used as model systems to study the effects of Rel/NF- ⁇ B signaling or inflammation.
  • Mice, rats, rabbits, guinea pigs, hamsters, zebrafish and amphibians are particularly useful as model systems.
  • a particularly useful insertion is a loxP sequence flanking a target gene which can be excised by ere.
  • the present invention provides, therefore, a mutation in or flanking a genetic locus encoding a target.
  • the mutation may be an insertion, deletion, substitution or addition to the target-coding sequence or its 5' or 3' untranslated region.
  • the animal model of the present invention is useful for screening for agents capable of ameliorating or mimicing the effects of a target.
  • the animal model produces low amounts of a target.
  • Another aspect of the present invention provides a genetically modified animal wherein said animal produces low amounts of a target relative to a non-genetically modified animal of the same species.
  • Reference to "low amounts” includes zero amounts or up to about 10% lower than normalized amounts.
  • Yet another aspect of the present invention provides multiple (i.e. two or more) genes which are modified.
  • the animal models of the present invention may be in the form of the animals including fish or may be, for example, in the form of embryos for transplantation.
  • the embryos are preferably maintained in a frozen state and may optionally be sold with instructions for use.
  • the genetically modified animals may also produce larger amounts of a target.
  • Another aspect of the present invention is directed to a genetically modified animal over-expressing genetic sequences encoding a target.
  • a genetically modified animal includes a transgenic animal, or a "knock-out” or “knock- in” animal as well as a conditional deletion mutant. Furthermore, co-suppression may be used to induce post-transcriptional gene silencing. Co-suppression includes induction of RNAi.
  • the compounds, agents, medicaments, nucleic acid molecules and other target antagonists or agonists of the present invention can be formulated in pharmaceutical compositions which are prepared according to conventional pharmaceutical compounding techniques. See, for example, Remington's Pharmaceutical Sciences, 18 Ed. (1990, Mack Publishing, Company, Easton, PA, U.S.A.).
  • the composition may contain the active agent or pharmaceutically acceptable salts of the active agent.
  • These compositions may comprise, in addition to one of the active substances, a pharmaceutically acceptable excipient, carrier, buffer, stabilizer or other materials well known in the art. Such materials should be non- toxic and should not interfere with the efficacy of the active ingredient.
  • the carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g.
  • the compounds can be formulated into solid or liquid preparations such as capsules, pills, tablets, lozenges, powders, suspensions or emulsions.
  • any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, suspending agents, and the like in the case of oral liquid preparations (such as, for example, suspensions, elixirs and solutions); or carriers such as starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations (such as, for example, powders, capsules and tablets). Because of their ease in administration, tablets and capsules represent the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are obviously employed.
  • tablets may be sugar-coated or enteric-coated by standard techniques.
  • the active agent can be encapsulated to make it stable to passage through the gastrointestinal tract while at the same time allowing for passage across the blood brain barrier. See for example, International Patent Publication No. WO 96/11698.
  • the compound may dissolved in a pharmaceutical carrier and administered as either a solution of a suspension.
  • suitable carriers are water, saline, dextrose solutions, fructose solutions, ethanol, or oils of animal, vegetative or synthetic origin.
  • the carrier may also contain other ingredients, for example, preservatives, suspending agents, solubilizing agents, buffers and the like.
  • the compounds When the compounds are being administered intrathecally, they may also be dissolved in cerebrospinal fluid.
  • the active agent is preferably administered in a therapeutically effective amount.
  • the actual amount administered and the rate and time-course of administration will depend on the nature and severity of the condition being treated. Prescription of treatment, e.g. decisions on dosage, timing, etc. is within the responsibility of general practitioners or specialists and typically takes account of the disorder to be treated, the condition of the individual patient, the site of delivery, the method of administration and other factors known to practitioners. Examples of techniques and protocols can be found in Remington's Pharmaceutical Sciences, supra.
  • targeting therapies may be used to deliver the active agent more specifically to certain types of cell, by the use of targeting systems such as antibodies or cell specific ligands or specific nucleic acid molecules. Targeting may be desirable for a variety of reasons, e.g. if the agent is unacceptably toxic or if it would otherwise require too high a dosage or if it would not otherwise be able to enter the target cells.
  • WO 92/19195 WO 94/25503, WO 95/01203, WO 95/05452, WO 96/02286, WO 96/02646, WO 96/40871, WO 96/40959 and WO 97/12635.
  • the vector could be targeted to the target cells.
  • the cell based delivery system is designed to be implanted in a patient's body at the desired target site and contains a coding sequence for the target agent.
  • the agent could be administered in a precursor form for conversion to the active form by an activating agent produced in, or targeted to, the cells to be treated. See, for example, European Patent Application No. 0 425 731 A and International Patent Publication No. WO 90/07936.
  • c-Rel generated as previously described were used (Kontgen et al, Genes Dev. 9: 1965- 1977, 1995) and maintained on a C57BL/6 background.
  • the RelA +/" and the NF- ⁇ Bl 7" mice are as previously described (Beg et al, Nature 376: 167-170, 1995; Sha et al, Cell 80: 321-330, 1995) and were on a mixed (C57BL/6 x 129) and C57BL/6 (> 9 generations) background, respectively. Due to the embryonic lethality of RelA mice, they were maintained at heterozygosity.
  • the TNF- ⁇ 7" mice (Korner et al, J. Exp. Med. 186: 1585- 1590, 1997) were on a C57BL/6 background. All mice were housed under specific pathogen free conditions.
  • the RelA "7" c-Rel “7” TNF- ⁇ "/_ triple mutant mice were derived by first mating RelA +/" c-Rel " _ TNF- ⁇ +/+ mice with the RelA +/+ c-Rel +/+ TNF- ⁇ _ " mice. Sibling matings were set up with mice that were heterozygous for all three genes. The resulting offspring, RelA +/" c-Rel 7" TNF- ⁇ "/_ , were viable and fertile. Subsequent sibling matings were set up to derive the triple mutant mice. This rationale was also employed to derive the RelA 7" NF- ⁇ Bl 7" TNF- ⁇ 7" triple mutant mice.
  • the primary antibodies used in this study were MoAb PCNA (clone PC10; Pharmingen); rabbit anti-mouse keratin-5, keratin-6, keratin- 14, keratin- 10, and filaggrin (BabCO).
  • MoAb PCNA clone PC10; Pharmingen
  • Paraffin tissue sections were deparaffinized and rehydrated through a graded ethanol series. The sections were blocked with a 1% BSA, 10% normal goat serum (NGS), 0.2%) Triton-X-100 solution.
  • Primary antibodies used were a mouse anti-human PCNA mAb (clone PC10/IgG2a; Pharmingen) and rabbit anti-keratin-6 antibody.
  • a goat anti-mouse secondary antibody was used to bind the anti-PCNA antibody (Santa Cruz) and all rabbit polyclonal antibodies were detected with the universal horse antirabbit secondary (Vector Labs). Tissues were then stained using the ABC peroxidase kit (Vector Labs) and counter stained with haematoxylin.
  • Pregnant mothers were injected intraperitoneally with 100 ⁇ g BrdU (Sigma) per gram body weight and killed after 1 h of pulsing.
  • Day 18 embryos were sacrificed and tissues taken for histological processing.
  • Paraffin embedded skin sections were first treated with 0.3%) v/v H 2 O 2 in 50% v/v methanol to block endogenous peroxidase.
  • the DNA was pretreated with 2 N HC1 at 37°C followed by washing in 0.1 M Na 2 B 4 O 7 pH 8.5 and PBS. The sections were then incubated in pepsin (0.4 mg/ml 0.1N HC1) for 20 mins at RT and then washed in PBS.
  • Sections were incubated with anti-BrdU (Company) for 1 hr in a humidified chamber, washed in PBS and then incubated for a further hour with the secondary universal horse anti-mouse biotinylated secondary antibody (Vector Labs). Staining was subsequently developed as described above.
  • a probe encoding part of the mouse c-rel cDNA (nucleotides 403-1621: Genebank accession X15842) was cloned into pBKS.
  • this plasmid was linearised with Hindlll and transcribed with T7 RNA polymerase in the presence of [33P] -labelled UTP (Amersham).
  • In situ hybridization was performed essentially as described (Thomas et al. Development 27:2537-2548, 2000).
  • TUNEL positive cells were subsequently detected by following the manufacturer's instructions to TUNEL staining (Promega and ApopTag TUNEL staining kit: Serologicals Corporation).
  • basal keratinocytes were stained with the rat anti-human ⁇ 6 integrin and anti-mouse CD71 (transferrin receptor). The stained cells were incubated on at 4°C for 30 mins and then washed. Cell culture and stains.
  • Isolated basal keratinocytes were seeded at a density of 106 cells in a 6-well plate (Costar) in serum-free keratinocyte media (Gibco-BRL) supplemented with hydrocortisone (0.5 ⁇ g/ml) and low levels of CaC12 (0.02 mM). Cultures were fixed in 2% v/v formaldehyde and subjected to immunoperoxidase staining for keratin 14 (LL001, IgG2a). Cells were then incubated with biotinylated secondary antibodies (Vector Laboratories), followed by streptavidin-HRP (ABC kit, Vector Laboratories) and enzyme substrate (AEC substrate kit, Vector Laboratories).
  • Basal keratinocytes were stained with FITC conjugated rat anti-human integrin-a6 antibody (BD Pharmingen) and PE conjugated anti-mouse CD71 antibody (BD Pharmingen) in a two-colour reaction or stained with a FITC conjugated anti-mouse CD29 antibody (integrin-bl) (Cymbus Biotechnology) in a single colour reaction.
  • Stained keratinocytes were either cell sorted or analysed immediately using a FACScan. Propidium iodide (PI) (20 ⁇ g/ml) was added to exclude dead cells during the analysis.
  • TA cells integratedin-a ⁇ hi CD71hi
  • Cell cycle profiles were analysed using the Modfit software program.
  • mice To follow the skin development of the RelA 7" c-Rel 7" TNF- ⁇ 7" null mice, the skin from day 18 embryos was surgically transplanted onto immunocompromised Rag 7" mice. About 1 cm 2 of dorsal skin was excised from genotyped day 18 embryos and kept in Mt.PBS. The B ⁇ Rag mice (11-15 weeks of age) were anaesthetized with Penthrane and the hair removed by plucking from the flank region above the ribcage. The graft bed was prepared by using a pair of curved scissors to remove a 1.5 cm 2 area of recipient skin. The donor skin was placed into the graft bed ensuring no contact between the donor and recipient skin.
  • the graft was dressed by adding a 1.5 cm 2 piece of Jelonet gauze (Smith & Nephew) over the entire graft bed and then bandaged with 3 M micropore surgical tape, followed by 3 M transpore surgical tape.
  • the mice were maintained in separate cages to avoid fighting between mice and, hence, possible graft damage.
  • the recipient mice were re-anaesthetized and bandages slowly cut away from the graft site.
  • the grafts were examined 2-3 times weekly up to four weeks post-transplant.
  • the donor graft was distinguished from the recipient coat by its agouti hair color.
  • Epidermal barrier function was determined as previously described (Hardman et al. Development 725:1541-1552, 1998).
  • RNA isolated from purified basal keratinocytes was subjected to Northern blotting essentially as described (Grumont ⁇ t al. Mol Cell 70:1283-1294,2002). Filters were sequentially probed with a 1.4 kb Xhol mouse c-myc cDNA, a rat 1.1 kb Pstl GAPDH cDNA and a 0.4 kb BamHl mouse K14 cDNA. nProtein blots were performed essentially as described (Nakamura Mol Cell Biol 22:5563-5574, 2002) using equivalent amounts of total cellular protein isolated from purifiedncontrol and mutant keratinocytes. Filters were probed with either mouse anti-cyclin DI monoclonal Ig (Santa Cruz Biotechnology), rabbit anti-cyclin D2 polyclonal Ig (Santa Cruz Biotechnology) or rat anti- HSP70 monoclonal antibody.
  • E19.5 pups was analysed. Images of control and mutant skin were taken at lOOOx magnification using a compound microscope and a digital camera (Zeiss). Images were analysed using an image analysis program (NIH Image 1.63 f). The areas of all basal cell profiles were measured per image. Seven to nine images representing 85 to 112 basal cells were analysed per animal. Results are given as mean ⁇ standard error of the mean. Data were analysed statistically using a two-factorial analysis of variance with the genotype and the developmental age as the factors and Scheffe's method for pair wise comparison using StatView 4.5 software.
  • EXAMPLE 2 Disruption of the TNF-signaling pathway rescues embryonic death associated with the combined losses ofNF- ⁇ Bl and RelA or RelA and c-Rel
  • nf kbl-/-rel ⁇ -/-tnf a-/- embryonic skin had a similar appearance to that of controls, a marginal 11 reduction in epidem al thickness was consistently noted.
  • the rel ⁇ -/-c-rel-/-tnf a-/- epidermis was distinctly thinner and hair follicles comprised only a few rudimentary buds (Fig. ID).
  • Fig. ID As the skin defects were most profound in the combined absence of RelA and c-Rel, all subsequent analysis focused on the rel ⁇ -/-c-rel-/-tnf a-/- mice.
  • EXAMPLE 3 c-Rel is expressed in the epidermis and in hair follicles.
  • c-rel was expressed in the sheath cells, the hair bulb and in the dermal papilla (Fig. 2E). c-rel was also expressed in a few scattered cells within the dermis (Fig. 2E). These findings confirm that c-rel is normally expressed in those skin structures that develop abnormally in rela-/-c-rel-/-tnf a-/- mice.
  • mice have four hair types (tylotrich, awl, suchene and zig zag) (Mann Anat Rec 144:135- 141, 1962).
  • the first wave (tylotrich or guard hair) commences around E14.
  • E17 a second wave of hair placodes appear, coinciding with the initiation of owl hair development.
  • the cells in the basal layer of rela-/-c-rel-/-tnf a-/- skin were organized differently from that of controls and the basal cell profiles appeared smaller in stained sections of rela-/-c-rel-/-tnf ' a-/- fetuses (Fig. 4A,B).
  • Measurement of basal cell size in rela-/-c-rel-/-tnf a-/- and rel ⁇ +/+c-rel-/-tnf a-/- mice confirmed that the mean basal cell profile area was reduced by -33% (0.397 ⁇ 0.014 vs.
  • K14 protein is normally expressed in basal cells and hair follicles (Fuchs and Byme Curr Opn Genet Dev 4:725-736, 1994). Although anti-K14 antibodies stained the inner most basal cells at the dermal/epidermal boundary of both control and mutant skin (Fig. 4C,D), staining which was usually two cell layers thick in the control was generally confined to one cell layer in the mutant, with little spinous staining.
  • the early differentiation marker K10 is expressed predominantly in the suprabasal spinous and granular layers (Fuchs and Byme 1994 supra).
  • Epidermal stem cell and transit-amplifying cell numbers are normal in the absence of c-Rel and RelA.
  • Cell surface marker expression has been used to delineate the stem cell, TA cell and maturing keratinocyte populations in the epidermis.
  • Stem cells and TA cells both express integrin bl with the highest levels expressed by stem cells. Integrin a6 expression is high in stem cells and TA cells, whereas keratinocytes committed to differentiate express low levels of this integrin.
  • Transferrin receptor (CD71) expression has also been used to distinguish between quiescent stem cells (integrin a ⁇ hi CD711o) and proliferating TA cells (integrin a ⁇ hi CD71hi).
  • Mitogen induced B cell growth is at least in part controlled by the Rel/NF-kB dependent induction of c-myc transcription, c-myc mRNA expression however was comparable in control and mutant epidermal TA cells (Fig. 6D).
  • the levels of cyclins DI and D2, important regulators of Gl that are also known targets of Rel/NF-kB were also similar in control and mutant TA cells (Fig. 6E), as were the cdk inhibitors p21 and p27. This indicates that the impaired expression of an unknown Rel/NF- kB target gene is responsible for the Gl growth defect observed in c-rel-/-rel ⁇ -/-tnf a-/- epidermal basal cells.
  • Rel/NF-kB regulates Gl/S phase progression and promotes cell survival.
  • TUNEL staining was performed on sections of control and mutant fetal skin (Fig. 6F). Mutant epidermis did not have abnormally increased numbers of TUNEL-positive cells. Dying cells were largely confined to the stratum granulosum of both control and mutant skin, a region of the epidermis in which keratinocytes undergo enucleation. Thus, the impaired cell cycle regulation of basal cells lacking RelA and c-Rel does not cause abnormally elevated cell death.
  • control grafts had an epidermis with a well-aligned basal cell layer, 1-2 cells thick (Fig. 8Bi).
  • the mutant graft exhibited extensive epidermal hyperplasia, hyperkeratosis, focal parakeratosis, comeal pustules with a granulocytic infiltrate and necrosis (Fig. 8Bii).
  • the mutant epidermis was abnormally thick (6-7 cell layers) with many hyperchromatic cells in the basal layer and a compacted stratum corneum (Fig. 8Ci,Cii).
  • the dermis in the mutant graft was extensively infiltrated with immune cells, eosinophils being most prominent (Fig. 8Ci,Cii).
  • the hyperproliferative state ofrela-/- c-rel-/- tnfa-/- embryonic skin grafts is dependent on TNF-a.

Abstract

The present invention relates generally to compositions and their use in the treatment and/or prophylaxis of inflammatory conditions in an animal such as a mammal including a human. More particularly, the compositions comprise agents which potentiate the function of the Rel/NF-kB signal transduction pathway. The compositions may also comprise agents which inhibit aspects of the immune system. Alternatively, multiple compositions may be employed which separately activate the NF-kB signaling pathway and inhibit systemically or locally an immune response. The present invention further contemplates methods of treatment and/or prophylaxis in an animal such as a mammal including a human by the administration of a potentiator of Rel/NF-kB signal transduction or a composition comprising such a potentiator, optionally together with an inhibitor of an immune response. Such potentiators are useful in the treatment or prophylaxis of inflammatory conditions such as but not limited to conditions affecting non-immune organs such as the skin, heart, kidney, liver, brain, pancreas, gall bladder, lung, adrenal gland, gastrointestinal tract, amongst other tissues.

Description

THERAPEUTIC AND PROPHYLACTIC COMPOSITIONS
AND USES THEREFOR
BACKGROUND OF THE INVENTION
FIELD OF THE INVENTION
The present invention relates generally to compositions and their use in the treatment and/or prophylaxis of inflammatory conditions in an animal such as a mammal including a human. More particularly, the compositions comprise agents which potentiate the function of the Rel/NF-κB signal transduction pathway. The compositions may also comprise agents which inhibit aspects of the immune system. Alternatively, multiple compositions may be employed which separately activate the NF-κB signaling pathway and inhibit systemically or locally an immune response. The present invention further contemplates methods of treatment and/or prophylaxis in an animal such as a mammal including a human by the administration of a potentiator of Rel/NF-κB signal transduction or a composition comprising such a potentiator, optionally together with an inhibitor of an immune response. Such potentiators are useful in the treatment or prophylaxis of inflammatory conditions such as but not limited to conditions affecting non-immune organs such as the skin, heart, kidney, liver, brain, pancreas, gall bladder, lung, adrenal gland, gastrointestinal tract, amongst other tissues.
DESCRIPTION OF THE PRIOR ART
Bibliographic details of the publications referred to by author in this specification are collected at the end of the description.
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. The Rel/NF-κB family of transcription factors are expressed as dimeric proteins comprising related subunits including c-Rel, RelA, RelB, F-κBl and NF-κB2. The Rel/NF-κB signaling pathways are conserved and have been implicated in the control of gene expression in many key physiological process including cell growth, division, survival and differentiation. In particular, Rel/NF-κB family members are thought to induce the transcription of genes involved in mediating stress responses as well as innate and adaptive immunity. Rel/NF-κB signaling is well suited to mediate such responses which optimally require rapid deployment because the Rel/NF-κB signaling pathway exists in a latent form within the cell and signaling can be rapidly induced. In essence, Rel/NF-κB transcription factors are retained in a latent state in the cytoplasm through association with inhibitory proteins (IκB proteins). A specific kinase complex (IKK complex) is activated in response to a wide range of signals and phosphorylates the IκB, targeting it for degradation. The released Rel/NF-κB transcription factors may then be transported to the nucleus where they recognize and bind to specific sequences in the regulatory region of target genes.
The Rel/NF-κB signaling pathways regulates the expression of a wide range of genes. In relation to regulating genes involved in immune function, target genes include regulators of immune and inflammatory response such as inflammatory cytokines (i.e. TNF, IL-2, IL- 6, IL-8), cytokine receptors (i.e. IL-2Rα), chemokines, chemokine receptors, adhesion molecules, free radical regulatory molecules, innate immune regulatory proteins such as defensins and the like.
Furthermore, activation of Rel/NFκB signaling pathways has been implicated in most if not all immune-mediated pathogenesis including diseases such as asthma, multiple sclerosis, inflammatory bowel disease and diabetes.
To date, the focus has been the role of Rel/NF-κB signaling pathways in regulating the immune response in these types of diseases. It has been generally accepted that activation of Rel/NF-κB signaling pathways leads to activation of the immune and inflammatory responses seen in these conditions and accordingly, it has been proposed to treat these conditions with antagonists of the Rel/NF-κB signaling pathway.
It has now been surprisingly and unexpectedly determined in accordance with the present invention that activation of the Rel/NF-κB signal transduction pathway is required to prevent or at least reduce inflammatory conditions. A summary of the two distinct pathways for Rel/NK-kB transcription factor activation is provided in Figure 9.
SUMMARY OF THE INVENTION
Throughout this specification, unless the context requires otherwise, the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element or integer or group of elements or integers but not the exclusion of any other element or integer or group of elements or integers.
In accordance with the present invention, it is determined that suppression of the Rel/NF- KB signaling pathways leads to, or at least contributes to, an inflammatory response, especially in non-immune organs. It is proposed, therefore, that the treatment and/or prophylaxis of inflammatory conditions in certain organs and tissues requires activation of the Rel/NF-κB signaling pathway. This is in direct contrast to prior teachings that inhibition of Rel/NF-κB signaling is required to prevent inflammatory responses.
The present invention provides, therefore, potentiators of the Rel/NF-κB signal transduction pathway for use in the treatment and prophylaxis of inflammation or inflammatory conditions. Generally, but not necessarily, the inflammatory condition is in a non-immune organ such as the skin, heart, kidney, liver, brain, pancreas, gall bladder, lung, adrenal gland, gastrointestinal tract, amongst other tissues.
The potentiators are conveniently in a composition comprising the potentiator and one or more pharmaceutically acceptable carriers, diluents and/or excipients.
The potentiators may also be used in conjunction with inhibitors of an immune response. Consequently, the present invention provides compositions or two- or multi-part pharmaceutical compositions comprising at least one potentiator of the Rel/NF-κB pathway and at least one inhibitor of an immune response or an immune component or portion of an inhibitor of an immune response pathway.
A potentiator of the Rel/NF-κB pathway may be a chemical agent such as a chemical molecule or peptide, polypeptide or protein or chemical analogs thereof or may be a genetic agent such as a sense or antisense molecule, ribozyme, DNAzyme or ribonuclease- type complex.
The potentiators of Rel/NF-κB signaling and compositions comprising same of the present invention may be used systemically or locally such as topically.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 is a photographic representation showing embryonic death resulting from the combined loss of NF-kB 1 and RelA or c-Rel and RelA is overcome by blocking TNF-a signalling. (A) rela-/-nfkbl-/-tnfa-/- or rela-/-c-rel-/-tnf a-/- fetuses were isolated at E18 from the respective inter-crosses of rela+/-nfkbl-/-tnf&-/- and rela+/-crel-/- tnfa-/- mice. (B-D) H&E stained sections of dorsal skin from El 8 fetuses. (B) relα+/+c-rel-/- tnf a.-/-, (C) relα-/-nf kbl-Atnf a-/- and (D) relα-/-c-rel-/-tnf a-/- genotypes (scale bar: 425μm). Color versions of this Figure are available from the patentee.
Figure 2 is a photographic representation showing c-rel mRNA is expressed in fetal skin. A c-rel cRNA probe was incubated with sections of El 8.5 skin. Panels A and C are dark- field images of the corresponding bright-field images in panels B and D. Sense probe controls are shown in panels C and D. Note the c-rel mRNA expression in the epidermis and hair follicles (A). At higher magnification (E, F), silver grains overlay the sheath cells (arrow in E) and the dermal papilla (arrowhead in E) of hair follicles, plus basal cells (arrow in F) and differentiating keratinocytes in the epidermis (bracket in E). HF, hair follicle; SC, sheath cells; DP, dermal papilla; BL, basal layer; K, keratinocytes. Scale bars: 43 μm in A, B, C, D and 22 μm in E, F. Color versions of this Figure are available from the patentee.
Figure 3 is a photographic representation showing Hair follicle morphogenesis and tooth development is impaired in the absence of c-Rel and RelA. (A-C) Hair follicle development in control (relα+/+c-rel-/-tnf a-/-) and mutant (relα-Z-crel-/- tnf a-/-) fetal sections stained with H&E (scale bar: 425μm). (A) E16: Hair placodes (tylotrich) are evident in control (black arrows) but not mutant skin. (B) El 7: In controls, continuing development of tylotrich hair follicles is evident, along with a new wave (Awl type, arrowhead) of hair placodes that is also evident in mutant skin (Awl type; arrow). (C) E18: In control skin, tylotrich and awl hair follicles have developed further. Only premature buds are evident in the mutant skin. (D, E). Serial sections of El 8 control and mutant embryos stained with H&E. (D) Mystacial vibrissae follicles develop normally in mutant embryos (scale bar: 35 1600μm). Note the paucity of hair follicles (arrow) between the whiskers. (E) The outgrowth of incisor teeth (arrow) was impaired in the mutant embryo. Note the lack of hair follicles (asterisks) adjacent to the vibrissae follicles (scale bar: 875μm). Color versions of this Figure are available from the patentee.
Figure 4 is a photographic representation showing epidermal differentiation is impaired in rela-/-c-rel-/-tnf a-/- embryos. Biochemical markers were used to analyse keratinocyte differentiation in control (rela+/+c-rel-/-tnf a-/-) and mutant (rela-/-c-rel-/-tnf a-/-) El 8 dorsal skin. H&E stained sections of (A) control and (B) mutant dorsal skin, depict the basal layer (B), the stratum spinosum (S), the stratum granulosum (G) and stratum corneum (C) (scale bar: 225μm). Frozen sections of control (C, E, G, I) and mutant (D, F, H, J) dorsal skin were subjected to indirect immunofluorescent staining for K14, K10, involucrin (Inv) and loricrin (Lor) expression. Two-colour immunofluorescent staining was performed on control (K, M, O, Q) and mutant (L, N, P, R) dorsal skin. Stains are colour coded, with yellow representing co-localization of Alexa and FITC fluorochromes. (S, T) X-gal staining of El 8 fetuses was performed to assess epidermal barrier function. Except where the tail was excised (arrow), the dye did not penetrate control or mutant fetuses. Color versions of this Figure are available from the patentee.
Figure 5 is a graphical representation showing Flow cytometric analysis of epidermal basal cells lacking c-Rel and RelA. Basal keratinocytes from El 8 control (rela+/-c-rel-/- tnf a-/-) and mutant (rela-/ -c-rel-/ -tnf a-/-) fetuses were stained with a FITC conjugated anti-integrin-bl (shaded histogram) or control anti-CD4 (open histogram) antibody (A). (B) Two-colour FACS analysis was performed by staining with FITC conjugated anti-integrin- a6 antibody and PE-conjugated anti-CD71 antibody. Three phenotypically distinct populations were identified by flow cytometry: quiescent stem cells, a6hi CD711o (gate 1); transit amplifying cells, aόhi CD71hi (gate 2) and post-mitotic differentiating cells, aόlo CD71hi (gate 3). Cells with high CD71 expression in the control profile (asterisk) were markedly reduced in the mutant. Figure 6 is a graphical and photographic represenation showing cell cycle defect in TA cells lacking RelA and c-Rel. Immunohistochemical staining of control (rela+/+c-rel-/-tnf a-/-) and mutant (rela-/-c-rel-/-tnf a-/-) skin sections for (A) PCNA expression and (B) BrdU incorporation (scale bar: 425μm). (C) Control and mutant TA cells (aόhi CD71hi) were purified by FACS sorting based on the gates shown in Figure 5B (gate 2) and the DNA content determined by PI staining of viable cells. Profiles are representative of 6 control and 6 mutant fetuses. Mod-Fit software was used to determine the proportion of TA in G0/G1, S and G2/M. (D) c-myc mRNA expression is normal in rela-/-c-rel-/-tnf a-/- keratinocytes. Total RNA isolated from rela+/+c-rel-/-tnfa-/- (lane 1) and mutant relα-/-c- rel-Z-tnf a-/- (lane 2) keratinocytes was subjected to Northern blot analysis using murine c- myc, K14 and GAPDH radiolabeled cDNA probes. (E) Cyclin DI and D2 expression in relα-/ -c-rel-/ -tnf 'a-/- keratinocytes. Total protein extracts from rel +/+c-rel-/-tnfa-/- (lane 1) and mutant relα-/ -c-rel-/ -tnf a-/- (lane 2) keratinocytes was subjected to Western blotting. Filters were sequentially probed with antibodies for cyclin DI, cyclin D2 and HSP70. (F) Control (relα+/+c-rel-/-tnfa-/-) (left panel) and mutant (relα-/-c-rel-/-tnf a-/- ) (right panel) dorsal skin was analysed by TUNEL staining. Note the apoptotic bodies (arrow) in the stratum granulosum (scale bar: 225 μm). Color versions of this Figure are available from the patentee.
Figure 7 is a photographic representation showing keratinocytes lacking RelA and c-Rel fail to form colonies in culture. Equivalent numbers of viable keratinocytes (106) were grown for 7 days. (A) Control (relα+/+c-rel-/-tnf a-/-) and mutant (relα-/-c-rel-/-tnf a-/-) keratinocyte cultures were stained with anti-keratin- 14 (+) or isotype matched negative control (-) antibodies. (B) Keratinocytes of differing genotypes (relα+/+c-rel+/+tnf a-/-, relα+/+c-rel-/-tnf a-/-, relα-/-c-rel+/+tnf a-/-, relα-/-c-rel-/-tnf a-/-) axe, shown by phase contrast. Color versions of this Figure are available from the patentee.
Figure 8 is a graphical representation showing relA/c-rel deficient skin grafts develop
TNF-a dependent epidermal hyperplasia. Skin from El 8 control (relα+/+c-rel-/-tnf a-/-) and mutant (relα-/-c-rel-/-tnf a-/-) fetuses was grafted 37 onto B6rαg-1-/- mice (A, Bi-iv,
Ci-iv) or B6rαg-l-/-tnfa-/- mice (Di-v). (A) Appearance of representative donor skin grafts over a 4 week period. (B, C) Histological analysis of skin grafts on βrag-1-/- recipients. H&E stained sections from control (Bi) and mutant (Bii) skin grafts were analysed under low power at 4 weeks (scale bar: lόOOμm). Staining for K6, was confined to hair follicles in the control graft (Biii) and the epidermis in the mutant graft (Biv) (Scale bar: 425μm). High power analysis of H&E stained sections reveals pockets of dermal infiltrate (arrow) in the mutant graft (Ci) that is mostly eosinophilic (asterisks) and granulocytic (arrowhead) (Cii) (scale bar: 225 μm). Note red cytoplasmic staining of eosinophils. Pearl staining for melanin on paraffin sections of control (Ciii) and mutant (Civ) grafts. (D) Skin grafts on B6rag-l-/-tnf a-/- recipients at 4 weeks. H&E stained sections from control (Di) and mutant (Dii) grafts were analysed at low power (scale bar: lόOOμm). Some dermal infiltrate (fibroblasts and macrophages: asterisks) was observed in the mutant graft (Diii). Three major hair types (tyrlotrich, awl and zig-zag) were observed in the control graft (Div) while only two (awl and zig-zag) were evident in the mutant graft (Dv). Note the mutant hairs are thinner. Color versions of this Figure are available from the patentee.
Figure 9 is a schematic representation of how Rel/NF-κB transcription factors are activated by two distinct pathways engaged by distinct extracellular signals. The "classical" pathway involves the activation of cytoplasmic c-Rel and RelA complexes via the IKKβ/IKKγ-dependent phosphorylation and degradation of IκB- -like proteins. The "alternative" pathway involves the activation of NF-κB2/RelB complexes via the IKKα dependent phosphorylation and degradation of NF-κB2 pi 00.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
The present invention is predicated in part on the surprising elucidation that the Rel/NF-κB signal transduction pathway needs to be up-regulated in certain organs or tissues in order to prevent or at least reduce inflammatory responses.
The terms "inflammation", "inflammatory response" and "inflammatory condition" are used interchangeably throughout the specification.
Generally, although not exclusively, the inflammatory response being prevented or treated affects a non-immune organ such as but not limited to the skin, heart, kidney, liver, brain, pancreas, gall bladder, lung, adrenal gland, gastrointestinal tract, amongst other tissues. Reference to the "gastrointestinal tract" includes the oesophagus, stomach and small and large bowel.
The present invention provides, therefore, potentiators of the Rel/NF-κB signal transduction pathway and compositions comprising the potentiators. The potentiators may elevate levels of NF-κB or various components of the Rel/NF-κB pathway directly or via inhibition of components which antagonize NF-κB function such as IKBS.
Before describing the present invention detail, it is to be understood that unless otherwise indicated, the subject invention is not limited to specific formulations of components, manufacturing methods, dosage regimens, 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 clearly dictates otherwise. Thus, for example, reference to a "solvent" includes a single solvent, as well as two or more solvents; reference to "an active agent" includes a single active agent, as well as two or more active agents; and so forth. In describing and claiming the present invention, the following terminology are used in accordance with the definitions set forth below.
The terms "compound", "active agent", "pharmacologically active agent", "medicament", "active" and "drug" are used interchangeably herein to refer to a chemical compound 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", "active 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 term "compound" is not to be construed as a chemical compound only but extends to peptides, polypeptides and proteins as well as genetic molecules such as RNA, DNA and chemical analogs thereof. The term "potentiator" is an example of a compound, active agent, pharmacologically active agent, medicament, active and drug which up-regulates the Rel/NF-κB signal transduction pathway. The term "up- regulates" encompasses inducing function of the pathway to correspondingly reduce an inflammatory response or the risk of an inflammatory response being elicited.
The present invention contemplates, therefore, compounds useful in up-regulating the function or activity of the Rel/NF-κB signaling pathway. The compounds have an effect on reducing or preventing or treating inflammatory conditions such as in non-immune organs including skin, heart, kidney, liver, brain, pancreas, gall bladder, lung, adrenal gland, gastrointestinal tract, amongst other tissues. Reference to a "compound", "active agent", "pharmacologically active agent", "medicament", "active" and "drug" includes combinations of two or more actives such as a potentiator of Rel/NF-κB signaling and an inhibitor of an immune response or immune response pathway. A "combination" also includes a two- or multi-part pharmaceutical composition where the agents are provided separately and given or dispensed separately or admixed together prior to dispensation. The terms "effective amount" and "therapeutically effective amount" of an agent as used herein mean a sufficient amount of the agent to provide the desired therapeutic or physiological effect. Furthermore, an "effective Rel/NF-κB potentiating amount" of an agent is a sufficient amount of the agent to directly or indirectly up-regulate the function of the Rel/NF-κB signaling pathway. This may be accomplished by the agents acting as an agonist of signaling pathway components, by the agents which are or mimic components of the signaling pathway, by agents which induce the pathway via cellular receptors or by the agents antagonizing inhibitors of signaling componets. 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.
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 emusifying agents, pH buffering agents, preservatives, and the like.
Similarly, a "pharmacologically acceptable" salt, ester, emide, 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, elimination of symptoms and/or underlying cause, prevention of the occurrence of symptoms and/or their underlying cause, and improvement or remediation of damage. Thus, for example, "treating" a patient involves prevention of a particular disorder or adverse physiological event in a susceptible individual as well as treatment of a clinically symptomatic individual by inhibiting or causing regression of a disorder or disease. Generally, such a condition or disorder is an inflammatory response or mediates or facilitates an inflammatory response or is a downstream product of an inflammatory response. Thus, for example, the present method of "treating" a patient with an inflammatory condition or with a propensity for one to develop encompasses both prevention of the condition, disease or disorder as well as treating the condition, disease or disorder. In any event, the present invention contemplates the treatment or prophylaxis of any inflammatory-type condition. Inflammatory conditions contemplated herein include but are not limited to skin inflammatory conditions such as psoriasis, ichthyosis, pityriasis, rubra pilaris, seborrhoea, keloids, keratoses, neoplasias and scleroderma, warts, benign growths and cancers of the skin. Other inflammatory conditions contemplated herein are those induced by microorganism and viruses such as enteropathogenic E. coli., Gardnerella vaginalis, Helicobacter plylori, Lactobacilli, Listeria monocytogenes, Micoplasma fermentans, Mycobacteria tuberculosis, Neisseria gonorrhoeae, Rickettsia ricettsii, Salmonella dublin, Salmonella typhimurium, Shigella flexneri, Staphyllococcus aureus, Rhodobacter sphaeroides, adenovirus, cytomegalovirus, Εpstein-Barr virus (ΕBV), hepatitis B virus, herpes virus saimiri, human herpesvirus 6, HIV-1, herpex simplex virus- 1, HTLV-1, influenza virus, measles virus, molony murine leukemia virus, Newcastle disease virus, respiratory syncytial virus, Sendai paramyxo virus and sindbis virus. Εukaryotic organisms causing inflammation include Theileria pai-va. Other inflammatory conditions include those caused or exacerbated by drug abuse or use of ethical drugs and physical stress including radiation exposure. Inflammatory conditions exacerbated by immunological mediators are also contemplated such as multiple sclerosis, asthma, systemic lupus, nephritis, diabetes, Chrohn's disease and other inflammatory bowel diseaess, arthritis, atherosclerosis and autoimmune myocarditis.
"Patient" as used herein refers to an animal, preferably a mammal and more preferably 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 patient regardless of whether a human or non-human animal may be referred to as an individual, subject, 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. For convenience, an "animal" includes an avian species such as a poultry bird, an aviary bird or game bird.
The compounds of the present invention may be large or small molecules, nucleic acid molecules (including antisense or sense molecules), peptides, polypeptides or proteins or hybrid molecules such as RNAi- or siRNA-complexes, ribozymes or DNAzymes. The compounds may need to be modified so as to facilitate entry into a cell. This is not a requirement if the compound interacts with an extracellular receptor.
The preferred animals are humans or other primates, livestock animals, laboratory test animals, companion animals or captive wild animals.
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 and donkeys. Non-mammalian animals such as avian species, zebrafish, amphibians (including cane toads) and Drosophila species such as Drosophila melanogaster are also contemplated.
The present invention provides, therefore, drugs which potentiate, activate or otherwise up- regulate the Rel/NF-κB signaling pathway including agents which antagonize inhibitors of NF-κB such as IκBs.
The present invention contemplates, therefore, methods of screening for drugs comprising, for example, contacting a candidate drug with an Rel/NF-κB pathway component including an extracellular receptor. Such a molecule is referred to herein as a "target" or "target molecule". The screening procedure includes assaying (i) for the presence of a complex between the drug and the target, or (ii) an alteration in the expression levels of nucleic acid molecules encoding the target. One form of assay involves competitive binding assays. In such competitive binding assays, the target is typically labeled. Free target is separated from any putative complex and the amount of free (i.e. uncomplexed) label is a measure of the binding of the agent being tested to target molecule. One may also measure the amount of bound, rather than free, target. It is also possible to label the compound rather than the target and to measure the amount of compound binding to target in the presence and in the absence of the drug being tested. Such compounds may inhibit the target which is useful, for example, in finding inhibitors of IKBS or may protect NF-κB or other components from being inhibited in complexes comprising inter alia IKBS. In addition, the compounds may be capable of interacting with extracellular receptors which in turn stimulate the Rel/NF-κB pathway.
Another technique for drug screening provides high throughput screening for compounds having suitable binding affinity to a target and is described in detail in Geysen (International Patent Publication No. WO 84/03564). Briefly stated, large numbers of different small peptide test compounds are synthesized on a solid substrate, such as plastic pins or some other surface. The peptide test compounds are reacted with a target and washed. Bound target molecule is then detected by methods well known in the art. This method may be adapted for screening for non-peptide, chemical entities. This aspect, therefore, extends to combinatorial approaches to screening for target antagonists or agonists.
Purified target can be coated directly onto plates for use in the aforementioned drug screening techniques. However, non-neutralizing antibodies to the target may also be used to immobilize the target on the solid phase.
The present invention also contemplates the use of competitive drug screening assays in which neutralizing antibodies capable of specifically binding the target compete with a test compound for binding to the target or fragments thereof. In this manner, the antibodies can be used to detect the presence of any peptide which shares one or more antigenic determinants of the target. Yet another useful source of compounds is a chemically modified cytokine or other activator of an extracellular receptor which then in turn activates the Rel/NF-κB.
Analogs contemplated herein include but are not limited to modification to side chains, incorporating of unnatural amino acids and/or their derivatives during peptide, polypeptide or protein synthesis and the use of crosslinkers and other methods which impose conformational constraints on the proteinaceous molecule or their analogs.
Examples of side chain modifications contemplated by the present invention include modifications of amino groups such as by reductive alkylation by reaction with an aldehyde followed by reduction with NaBH4; amidination with mefhylacetimidate; acylation with acetic anhydride; carbamoylation of amino groups with cyanate; trinitrobenzylation of amino groups with 2, 4, 6-trinitrobenzene sulphonic acid (TNBS); acylation of amino groups with succinic anhydride and tetrahydrophthalic anhydride; and pyridoxylation of lysine with pyridoxal-5-phosphate followed by reduction with NaBH4.
The guanidine group of arginine residues may be modified by the formation of heterocyclic condensation products with reagents such as 2,3-butanedione, phenylglyoxal and glyoxal.
The carboxyl group may be modified by carbodiimide activation via O-acylisourea formation followed by subsequent derivitization, for example, to a corresponding amide.
Sulphydryl groups may be modified by methods such as carboxymethylation with iodoacetic acid or iodoacetamide; performic acid oxidation to cysteic acid; formation of a mixed disulphides with other thiol compounds; reaction with maleimide, maleic anhydride or other substituted maleimide; formation of mercurial derivatives using 4- chloromercuribenzoate, 4-chloromercuriphenylsulphonic acid, phenylmercury chloride, 2- chloromercuri-4-nitrophenol and other mercurials; carbamoylation with cyanate at alkaline pH. Tryptophan residues may be modified by, for example, oxidation with N- bromosuccinimide or alkylation of the indole ring with 2-hydroxy-5-nitrobenzyl bromide or sulphenyl halides. Tyrosine residues on the other hand, may be altered by nitration with tetranitromethane to form a 3-nitrotyrosine derivative.
Modification of the imidazole ring of a histidine residue may be accomplished by alkylation with iodoacetic acid derivatives or N-carbethoxylation with diethylpyrocarbonate .
Examples of incorporating unnatural amino acids and derivatives during peptide synthesis include, but are not limited to, use of norleucine, 4-amino butyric acid, 4-amino-3- hydroxy-5-phenylpentanoic acid, 6-aminohexanoic acid, t-butylglycine, norvaline, phenylglycine, ornithine, sarcosine, 4-amino-3-hydroxy-6-methylheptanoic acid, 2-thienyl alanine and/or D-isomers of amino acids. A list of unnatural amino acid, contemplated herein is shown in Table 1.
TABLE 1
Codes for non-conventional amino acids
Non-conventional Code Non-conventional Code amino acid amino acid
α-aminobutyric acid Abu L-N-methylalanine Nmala α-amino-α-methylbutyrate Mgabu L-N-methylarginine Nmarg aminocyclopropane- Cpro L-N-methylasparagine Nmasn carboxylate L-N-methylaspartic acid Nmasp aminoisobutyric acid Aib L-N-methylcysteine Nmcys aminonorbornyl- Norb L-N-methylglutamine Nmgln carboxylate L-N-methylglutamic acid Nmglu cyclohexylalanine Chexa L-Nmethylhistidine Nmhis cyclopentylalanine Cpen L-N-methylisolleucine Nmile
D-alanine Dal L-N-methylleucine Nmleu
D-arginine Darg L-N-methyllysine Nmlys
D-aspartic acid Dasp L-N-methylmethionine Nmmet
D-cysteine Dcys L-N-methylnorleucine Nmnle
D-glutamine Dgln L-N-methylnorvaline Nmnva
D-glutamic acid Dglu L-N-methylornithine Nmorn
D-histidine Dhis L-N-methylphenylalanine Nmphe
D-isoleucine Dile L-N-methylproline Nmpro
D-leucine Dleu L-N-methylserine Nmser
D-lysine Dlys L-N-methylthreonine Nmthr
D-methionine Dmet L-N-methyltryptophan Nmtrp
D-ornithine Dorn L-N-methyltyrosine Nmtyr
D-phenylalanine Dphe L-N-methylvaline Nmval
D-proline Dpro L-N-methylethylglycine Nmetg
D-serine Dser L-N-methyl-t-butylglycine Nmtbug D-threonine Dthr L-norleucine Nle
D-tryptophan Dtrp L-norvaline Nva
D-tyrosine Dtyr α-methyl-aminoisobutyrate Maib
D-valine Dval α-methyl-γ-aminobutyrate Mgabu D-α-methylalanine Dmala α-methylcyclohexylalanine Mchexa
D-α-methylarginine Dmarg α-methylcylcopentylalanine Mcpen
D-α-methylasparagine Dmasn α-methyl-α-napthylalanine Manap
D-α-methylaspartate Dmasp α-methylpenicillamine Mpen
D-α-methylcysteine Dmcys N-(4-aminobutyl)glycine Nglu D-α-methylglutamine Dmgln N-(2-aminoethyl)glycine Naeg
D-α-methylhistidine Dmhis N-(3 -aminopropyl)glycine Norn
D-α-methylisoleucine Dmile N-amino- -methylbutyrate Nmaabu
D-α-methylleucine Dmleu α-napthylalanine Anap
D-α-methyllysine Dmlys N-benzylglycine Nphe D-α-methylmethionine Dmmet N-(2-carbamylethyl)glycine Ngln
D-α-methylornithine Dmorn N-(carbamylmethyl)glycine Nasn
D-α-methylphenylalanine Dmphe N-(2-carboxyethyl)glycine Nglu
D-α-methylproline Dmpro N-(carboxymethyl)glycine Nasp
D-α-methylserine Dmser N-cyclobutylglycine Ncbut D-α-methylthreonine Dmthr N-cycloheptylglycine Nchep
D-α-methyltryptophan Dmtrp N-cyclohexylglycine Nchex
D-α-methyltyrosine Dmty N-cyclodecylglycine Ncdec
D-α-methylvaline Dmval N-cylcododecylglycine Ncdod
D-N-methylalanine Dnmala N-cyclooctylglycine Ncoct D-N-methylarginine Dnmarg N-cyclopropylglycine Ncpro
D-N-methylasparagine Dnmasn N-cycloundecylglycine Ncund
D-N-methylaspartate Dnmasp N-(2,2-diphenylethyl)glycine Nbhm
D-N-methylcysteine Dnmcys N-(3,3-diphenylpropyl)glycine Nbhe
D-N-methylglutamine Dnmgln N-(3-guanidinopropyl)glycine Narg D-N-methylglutamate Dnmglu N-( 1 -hydroxyethyl)glycine Nthr D-N-methylhistidine Dnmhis N-(hydroxyethyl))glycine Nser
D-N-methylisoleucine Dnmile N-(imidazolylethyl))glycine Nhis
D-N-methylleucine Dnmleu N-(3-indolylyethyl)glycine Nhtrp
D-N-methyllysine Dnmlys N-methyl-γ-aminobutyrate Nmgabu N-methylcyclohexylalanine Nmchexa D-N-methylmethionine Dnmmet
D-N-methylornithine Dnmorn N-methylcyclopentylalanine Nmcpen
N-methylglycine Nala D-N-methylphenylalanine Dnmphe
N-methylaminoisobutyrate Nmaib D-N-methylproline Dnmpro
N-(l-methylpropyl)glycine Nile D-N-methylserine Dnmser N-(2-methylpropyl)glycine Nleu D-N-methylthreonine Dnmthr
D-N-methyltryptophan Dnmtrp N-(l-methylethyl) glycine Nval
D-N-methyltyrosine Dnmtyr N-methyla-napthylalanine Nmanap
D-N-methylvaline Dnmval N-methylpenicillamine Nmpen γ-aminobutyric acid Gabu N-(p-hydroxyphenyl)glycine Nhtyr L-t-butylglycine Tbug N-(thiomethyl)glycine Ncys
L-ethylglycine Etg penicillamine Pen
L-homophenylalanine Hphe L-α-methylalanine Mala
L-α-methylarginine Marg L-α-methylasparagine Masn
L-α-methylaspartate Masp L-α-methyl-t-butylglycine Mtbug L-α-methylcysteine Mcys L-methylethylglycine Metg
L-α-methylglutamine Mgln L-α-methylglutamate Mglu
L-α-methylhistidine Mhis L-α-methylhomophenylalanine Mhphe
L-α-methylisoleucine Mile N-(2-methylthioethyl)glycine Nmet
L-α-methylleucine Mleu L-α-methyllysine Mlys L-α-methylmethionine Mmet L-α-methylnorleucine Male
L-α-methylnorvaline Mnva L-a-methylornithine Morn
L-α-methylphenylalanine Mphe L-a-methylproline Mpro
L-α-methylserine Mser L-a-methylthreonine Mthr
L-α-methyltryptophan Mtrp L-a-methyltyrosine Mtyr L-α-methylvaline Mval L-N-methylhomophenylalanine Nmhphe N-(N-(2,2-diphenylethyl) Nnbhm N-(N-(3,3-diphenylpropyl) Nnbhe carbamylmethyl)glycine carbamylmethyl)glycine l-carboxy-l-(2,2-diphenyl- Nmbc ethylamino)cyclopropane
Crosslinkers can be used, for example, to stabilize 3D conformations, using homo- bifunctional crosslinkers such as the bifunctional imido esters having (CH2)n spacer groups with n=l to n=6, glutaraldehyde, N-hydroxysuccinimide esters and hetero-bifunctional reagents which usually contain an amino-reactive moiety such as N-hydroxysuccinimide and another group specific-reactive moiety such as maleimido or dithio moiety (SH) or carbodiimide (COOH). In addition, peptides can be conformationally constrained by, for example, incorporation of Cα and N α-methylamino acids, introduction of double bonds between Cα and Cp atoms of amino acids and the formation of cyclic peptides or analogs by introducing covalent bonds such as forming an amide bond between the N and C termini, between two side chains or between a side chain and the N or C terminus.
Accordingly, one aspect of the present invention contemplates any compound which binds or otherwise interacts with a component of the Rel/NF-κB pathway resulting in potentiation, activation or general up-regulation of the Rel/NF-κB signaling transduction pathway.
The present invention is also useful for screening for other compounds which reduce expression of a gene encoding an NF-κB inhibitor or which up-regulates expression of genes encoding Rel/NF-κB components such as NF-κB itself or a receptor linked to the Rel/NF-κB pathway. Such targets may be used in any of a variety of drug screening techniques, such as those described herein and in International Publication No. WO 97/02048.
A target antagonist or agonist includes a variant of the target molecule. In one embodiment, the target is a polypeptide. The term "polypeptide" refers to a polymer of amino acids and its equivalent and does not refer to a specific length of the product, thus, peptides, oligopeptides and proteins are included within the definition of a polypeptide. This term also does not exclude modifications of the polypeptide, for example, glycosylations, aceylations, phosphorylations and the like. Included within the definition are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids such as those given in Table 1) or polypeptides with substituted linkages. Such polypeptides may need to be able to enter the cell. Alternatively, the NF-κB pathway is potentiated via an external receptor such as BCR, IL-IR, TNFR, LTBR, CDl lb/CD18, CD28, CD2, CD35, CD3, CD40, CD4, fc-2a-receptor (IgG2a), Flt- 1 , Ly6A/E, N-CAM, trail receptor- 1 , trail-receptor-2 or trail-receptor-4.
Examples of Rel/NF-κB signal transduction potentiators include IL-1, IL-2, IL-12, IL-15, IL-17, IL-18, LIF, THANK, TNF-α, TNF-β as well as any of the TNF-receptor superfamily ligands, 1-β-D-arabinofuranosyl-cytosine (ara-C), anthralin, azidothymidine (AZT), camptothecin, ciprofibrate, cisplatin, daunomycin, daunorubicin, doxorubicin, etoposide, haloperidol, methamphetamine, phenobarbital, tamoxifen, taxol (paclitaxel), vinblastine, vincristine, advanced glycated end products (AGEs), amyloid protein fragment (βA4), maleylated BSA, modified (oxidized) LDL, bone morphogenic protein 2, bone morphogenic protein 4, folicle stimulating hormone, human growth hormone, insulin, M- CSF, nerve growth factor, platelet-derived growth factor, serum, TGF-α, 12(R)- hydroxyeicosatrienoic acid, amino acid analogs, anaphylatoxin C3a, anaphylatoxin C5a, angiotensin II, basic calcium phosphate crystals, bradikinin, C2-ceramide (N-acetyl- sphingosine), cerulein, collagen lattice, collagen type I, Des-ArglO-kallidin (BI receptor agonist), double-stranded polynucleotides, f-Met-Leu-Phe, heat shock protein 60 (HSP 60), hemoglobin, hyaluronan, kaianic acid (kainate), leukotriene B4, L-glutamate, lysophosphatidylcholine (LysoPC), platelet activating factor (PAF), potassium, thrombin, 2-deoxyglucose, anisomycin, Brefeldin A, calchicine, calcium ionophores, calyculin A, cobalt chloride, Con A, cycloheximide, cyclopiazonic acid, forskolin, glass fibres, linoleic acid, L-NMA, lysophosphatidic acid, monensin, N-methyl-D-aspartate, nocodazol, okadaic acid, PHA, phorbol ester, podophyllotoxin, pyrogallol, guinolinic acid, thapsigargin or tunicamycin. Another useful group of compounds is a mimetic. The terms "peptide mimetic", "target mimetic" or "mimetic" are intended to refer to a substance which has some chemical similarity to the target but which antagonizes or agonizes or mimics the target. A peptide mimetic may be a peptide-containing molecule that mimics elements of protein secondary structure (Johnson et al, "Peptide Turn Mimetics" in Biotechnology and Pharmacy, Pezzuto et al, Eds., Chapman and Hall, New York, 1993). The underlying rationale behind the use of peptide mimetics is that the peptide backbone of proteins exists chiefly to orient amino acid side chains in such a way as to facilitate molecular interactions such as those of antibody and antigen, enzyme and substrate or scaffolding proteins. A peptide mimetic is designed to permit molecular interactions similar to the natural molecule. Peptide or non- peptide mimetics may be useful, for example, to activate the NF-κB signaling pathway via an external receptor or enter the cell and agonize or antagonize a particular component.
Again, the compounds of the present invention may be selected to interact with a target alone or single or multiple compounds may be used to affect multiple targets. For example, multiple Rel/NF-κB components may be targeted with or without certain immune response components being inhibited.
The target polypeptide or fragment employed in such a test may either be free in solution, affixed to a solid support, or borne on a cell surface. One method of drug screening utilizes eukaryotic or prokaryotic host cells which are stably transformed with recombinant polynucleotides expressing the polypeptide or fragment, preferably in competitive binding assays. Such cells, either in viable or fixed form, can be used for standard binding assays. One may measure, for example, the formation of complexes between a target or fragment and the agent being tested, or examine the degree to which the formation of a complex between a target or fragment and a known ligand is aided or interfered with by the agent being tested.
A substance identified as a modulator of target function or gene activity may be a peptide or non-peptide in nature. Non-peptide "small molecules" are often preferred for many in vivo pharmaceutical uses. Accordingly, a mimetic or mimic of the substance (particularly if a peptide) may be designed for pharmaceutical use.
The designing of mimetics to a pharmaceutically active compound is a known approach to the development of pharmaceuticals based on a "lead" compound. This might be desirable where the active compound is difficult or expensive to synthesize or where it is unsuitable for a particular method of administration, e.g. peptides are unsuitable active agents for oral compositions as they tend to he quickly degraded by proteases in the alimentary canal. Mimetic design, synthesis and testing is generally used to avoid randomly screening large numbers of molecules for a target property. Conveniently, and in one example, the mimetic is of a cytokine or other molecule which interacts with a cell surface receptor and stimulates the Rel/NF-κB pathway.
There are several steps commonly taken in the design of a mimetic from a compound having a given target property. First, the particular parts of the compound that are critical and/or important in determining the target property are determined. In the case of a peptide, this can be done by systematically varying the amino acid residues in the peptide, e.g. by substituting each residue in turn. Alanine scans of peptides are commonly used to refine such peptide motifs. These parts or residues constituting the active region of the compound are known as its "pharmacophore".
Once the pharmacophore has been found, its structure is modeled according to its physical properties, e.g. stereochemistry, bonding, size and/or charge, using data from a range of sources, e.g. spectroscopic techniques, x-ray diffraction data and NMR. Computational analysis, similarity mapping (which models the charge and/or volume of a pharmacophore, rather than the bonding between atoms) and other techniques can be used in this modeling process.
In a variant of this approach, the three-dimensional structure of the ligand and its binding partner are modeled. This can be especially useful where the ligand and/or binding partner change conformation on binding, allowing the model to take account of this in the design of the mimetic. Modeling can be used to generate inhibitors which interact with the linear sequence or a three-dimensional configuration.
A template molecule is then selected onto which chemical groups which mimic the pharmacophore can be grafted. The template molecule and the chemical groups grafted onto it can conveniently be selected so that the mimetic is easy to synthesize, is likely to be pharmacologically acceptable, and does not degrade in vivo, while retaining the biological activity of the lead compound. Alternatively, where the mimetic is peptide-based, further stability can be achieved by cyclizing the peptide, increasing its rigidity. The mimetic or mimetics found by this approach can then be screened to see whether they have the target property, or to what extent they exhibit it. Further optimization or modification can then be carried out to arrive at one or more final mimetics for in vivo or clinical testing.
The goal of rational drug design is to produce structural analogs of biologically active polypeptides of interest or of small molecules with which they interact (e.g. agonists, antagonists, inhibitors or enhancers) in order to fashion drugs which are, for example, more active or stable forms of the polypeptide, or which, e.g. enhance or interfere with the function of a polypeptide in vivo. See, e.g. Hodgson (Bio/Technology 9: 19-21, 1991). In one approach, one first determines the three-dimensional structure of a protein of interest (e.g. NF-κB or a receptor or a component of the Rel/NF-κB pathway) by x-ray crystallography, by computer modeling or most typically, by a combination of approaches. Useful information regarding the structure of a polypeptide may also be gained by modeling based on the structure of homologous proteins. An example of rational drug design is the development of HIV protease inhibitors (Erickson et al, Science 249: 527- 533, 1990). In addition, target molecules may be analyzed by an alanine scan (Wells, Methods Enzymol. 202: 2699-2705, 1991). In this technique, an amino acid residue is replaced by Ala and its effect on the peptide 's activity is determined. Each of the amino acid residues of the peptide is analyzed in this manner to determine the important regions of the peptide. It is also possible to isolate a target-specific antibody, selected by a functional assay and then to solve its crystal structure. In principle, this approach yields a pharmacore upon which subsequent drug design can be based. It is possible to bypass protein crystallography altogether by generating anti-idiotypic antibodies (anti-ids) to a functional, pharmacologically active antibody. As a mirror image of a mirror image, the binding site of the anti-ids would be expected to be an analog of the original receptor. The anti-id could then be used to identify and isolate peptides from banks of chemically or biologically produced banks of peptides. Selected peptides would then act as the pharmacore.
Two-hybrid screening is also useful in identifying other members of a biochemical or genetic pathway associated with a target. Two-hybrid screening conveniently uses Saccharomyces cerevisiae and Saccharomyces pombe. Target interactions and screens for inhibitors can be carried out using the yeast two-hybrid system, which takes advantage of transcriptional factors that are composed of two physically separable, functional domains. The most commonly used is the yeast GAL4 transcriptional activator consisting of a DNA binding domain and a transcriptional activation domain. Two different cloning vectors are used to generate separate fusions of the GAL4 domains to genes encoding potential binding proteins. The fusion proteins are co-expressed, targeted to the nucleus and if interactions occur, activation of a reporter gene (e.g. lacZ) produces a detectable phenotype. In the present case, for example, S. cerevisiae is co-transformed with a library or vector expressing a cDNA GAL4 activation domain fusion and a vector expressing a an Rel/NF-κB pathway component fused to GAL4. If lacZ is used as the reporter gene, co- expression of the fusion proteins will produce a blue color. Small molecules or other candidate compounds which interact with a target will result in loss of color of the cells. Reference may be made to the yeast two-hybrid systems as disclosed by Munder et al. (Appl. Microbiol. Biotechnol. 52(3): 311-320, 1999) and Young et al., Nat. Biotechnol. 16(10): 946-950, 1998). Molecules thus identified by this system are then re-tested in animal cells.
The present invention extends to a genetic approach to up-regulating expression of an Rel/NF-κB pathway component (including a receptor) or down-regulating an inhibitor of NF-κB. In one example, nucleic acid molecules which encode a signaling component are used to elevate levels of the component. Alternatively, nucleic acid molecules which induce temporary or permanent gene silencing of NF-κB inhibitors may also be used.
The terms "nucleic acids", "nucleotide" and "polynucleotide" include RNA, cDNA, genomic DNA, synthetic forms and mixed polymers, both sense and antisense strands, and may be chemically or biochemically modified or may contain non-natural or derivatized nucleotide bases, as will be readily appreciated by those skilled in the art. Such modifications include, for example, labels, methylation, substitution of one or more of the naturally occurring nucleotides with an analog (such as the morpholine ring) internucleotide modifications such as uncharged linkages (e.g. methyl phosphonates phosphotriesters, phosphoamidates, carbamates, etc.), charged linkages (e.g phosphorothioates, phosphorodithioates, etc.), pendent moieties (e.g. polypeptides) intercalators (e.g. acridine, psoralen, etc.), chelators, alkylators and modified linkages (e.g α-anomeric nucleic acids, etc.). Also included are synthetic molecules that mimic polynucleotides in their ability to bind to a designated sequence via hydrogen binding and other chemical interactions. Such molecules are known in the art and include, for example, those in which peptide linkages substitute for phosphate linkages in the backbone of the molecule.
Antisense polynucleotide sequences, for example, are useful in silencing transcripts of NF- KB inhibitors such as IKBS. Furthermore, polynucleotide vectors containing all or a portion of a target NF-κB inhibitor gene locus may be placed under the control of a promoter in an antisense orientation and introduced into a cell. Expression of such an antisense construct within a cell will interfere with target transcription and/or translation. Furthermore, co- suppression and mechanisms to induce RNAi or siRNA may also be employed. Alternatively, antisense or sense molecules may be directly administered. In this latter embodiment, the antisense or sense molecules may be formulated in a composition and then administered by any number of means to target cells.
A variation on antisense and sense molecules involves the use of morpholinos, which are oligonucleotides composed of morpholine nucleotide derivatives and phosphorodiamidate linkages (for example, Summerton and Weller, Antisense and Nucleic Acid Drug Development 7: 187-195, 1997). Such compounds are injected into embryos and the effect of interference with mRNA is observed.
In one embodiment, the present invention employs compounds such as oligonucleotides and similar species for use in modulating the function or effect of nucleic acid molecules encoding an NF-κB inhibitor, i.e. the oligonucleotides induce transcriptional or post- transcriptional gene silencing. This is accomplished by providing oligonucleotides which specifically hybridize with one or more nucleic acid molecules encoding the inhibitor. The oligonucleotides may be provided directly to a cell or generated within the cell. As used herein, the terms "target nucleic acid" and "nucleic acid molecule encoding an NF-κB inhibitor" have been used for convenience to encompass DNA encoding the inhibitor, RNA (including pre-mRNA and mRNA or portions thereof) transcribed from such DNA, and also cDNA derived from such RNA. The hybridization of a compound of the subject invention with its target nucleic acid is generally referred to as "antisense". Consequently, the preferred mechanism believed to be included in the practice of some preferred embodiments of the invention is referred to herein as "antisense inhibition." Such antisense inhibition is typically based upon hydrogen bonding-based hybridization of oligonucleotide strands or segments such that at least one strand or segment is cleaved, degraded, or otherwise rendered inoperable. In this regard, it is presently preferred to target specific nucleic acid molecules and their functions for such antisense inhibition.
The functions of DNA to be interfered with can include replication and transcription. Replication and transcription, for example, can be from an endogenous cellular template, a vector, a plasmid construct or otherwise. The functions of RNA to be interfered with can include functions such as franslocation of the RNA to a site of protein translation, franslocation of the RNA to sites within the cell which are distant from the site of RNA synthesis, translation of protein from the RNA, splicing of the RNA to yield one or more RNA species, and catalytic activity or complex formation involving the RNA which may be engaged in or facilitated by the RNA. In one example, the result of such interference with target nucleic acid function is reduced levels of IκB. In the context of the present invention, "modulation" and "modulation of expression" mean either an increase (stimulation) or a decrease (inhibition) in the amount or levels of a nucleic acid molecule encoding the gene, e.g., DNA or RNA. Inhibition is often the preferred form of modulation of expression and mRNA is often a preferred target nucleic acid.
In the context of this invention, "hybridization" means the pairing of complementary strands of oligomeric compounds. In the present invention, the preferred mechanism of pairing involves hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleoside or nucleotide bases (nucleobases) of the strands of oligomeric compounds. For example, adenine and thymine are complementary nucleobases which pair through the formation of hydrogen bonds. Hybridization can occur under varying circumstances.
An antisense compound is specifically hybridizable when binding of the compound to the target nucleic acid interferes with the normal function of the target nucleic acid to cause a loss of activity, and there is a sufficient degree of complementarity to avoid non-specific binding of the antisense compound to non-target nucleic acid sequences under conditions in which specific binding is desired, i.e. under physiological conditions in the case of in vivo assays or therapeutic treatment, and under conditions in which assays are performed in the case of in vitro assays.
"Complementary" as used herein, refers to the capacity for precise pairing between two nucleobases of an oligomeric compound. For example, if a nucleobase at a certain position of an oligonucleotide (an oligomeric compound), is capable of hydrogen bonding with a nucleobase at a certain position of a target nucleic acid, said target nucleic acid being a DNA, RNA, or oligonucleotide molecule, then the position of hydrogen bonding between the oligonucleotide and the target nucleic acid is considered to be a complementary position. The oligonucleotide and the further DNA, RNA, or oligonucleotide molecule are complementary to each other when a sufficient number of complementary positions in each molecule are occupied by nucleobases which can hydrogen bond with each other. Thus, "specifically hybridizable" and "complementary" are terms which are used to indicate a sufficient degree of precise pairing or complementarity over a sufficient number of nucleobases such that stable and specific binding occurs between the oligonucleotide and a target nucleic acid.
According to the present invention, compounds include antisense oligomeric compounds, antisense oligonucleotides, ribozymes, external guide sequence (EGS) oligonucleotides, alternate splicers, primers, probes, and other oligomeric compounds which hybridize to at least a portion of the target nucleic acid. As such, these compounds may be introduced in the form of single-stranded, double-stranded, circular or hairpin oligomeric compounds and may contain structural elements such as internal or terminal bulges or loops. Once introduced to a system, the compounds of the invention may elicit the action of one or more enzymes or structural proteins to effect modification of the target nucleic acid. One non-limiting example of such an enzyme is RNAse H, a cellular endonuclease which cleaves the RNA strand of an RNA:DNA duplex. It is known in the art that single-stranded antisense compounds which are "DNA-like" elicit RNAse H. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of oligonucleotide-mediated inhibition of gene expression. Similar roles have been postulated for other ribonucleases such as those in the RNase III and ribonuclease L family of enzymes.
While the preferred form of antisense compound is a single-stranded antisense oligonucleotide, in many species the introduction of double-stranded structures, such as double-stranded RNA (dsRNA) molecules, has been shown to induce potent and specific antisense-mediated reduction of the function of a gene or its associated gene products. This phenomenon occurs in both plants and animals.
In the context of the subject invention, the term "oligomeric compound" refers to a polymer or oligomer comprising a plurality of monomeric units. In the context of this invention, the term "oligonucleotide" refers to an oligomer or polymer of ribonucleic acid
(RNA) or deoxyribonucleic acid (DNA) or mimetics, chimeras, analogs and homologs thereof. This term includes oligonucleotides composed of naturally occurring nucleobases, sugars and covalent internucleoside (backbone) linkages as well as oligonucleotides having non-naturally occurring portions which function similarly. Such modified or substituted oligonucleotides are often preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for a target nucleic acid and increased stability in the presence of nucleases.
While oligonucleotides are a preferred form of the compounds of this invention, the present invention comprehends other families of compounds as well, including but not limited to oligonucleotide analogs and mimetics such as those herein described.
The open reading frame (ORF) or "coding region" which is known in the art to refer to the region between the translation initiation codon and the translation termination codon, is a region which may be effectively targeted. Within the context of the present invention, one region is the intragenic region encompassing the translation initiation or termination codon of the open reading frame (ORF) of a gene.
Other target regions include the 5' untranslated region (5'UTR), known in the art to refer to the portion of an mRNA in the 5' direction from the translation initiation codon, and thus including nucleotides between the 5' cap site and the translation initiation codon of an mRNA (or corresponding nucleotides on the gene), and the 3' untranslated region (3'UTR), known in the art to refer to the portion of an mRNA in the 3' direction from the translation termination codon, and thus including nucleotides between the translation termination codon and 3' end of an mRNA (or corresponding nucleotides on the gene). The 5' cap site of an mRNA comprises an N7-methylated guanosine residue joined to the 5'-most residue of the mRNA via a 5'-5' triphosphate linkage. The 5' cap region of an mRNA is considered to include the 5' cap structure itself as well as the first 50 nucleotides adjacent to the cap site. It is also preferred to target the 5' cap region.
Although some eukaryotic mRNA transcripts are directly translated, many contain one or more regions, known as "introns", which are excised from a transcript before it is translated. The remaining (and, therefore, translated) regions are known as "exons" and are spliced together to form a continuous mRNA sequence. Targeting splice sites, i.e. intron- exon junctions or exon-intron junctions, may also be particularly useful in situations where aberrant splicing is implicated in disease, or where an overproduction of a particular splice product is implicated in disease. Aberrant fusion junctions due to rearrangements or deletions are also preferred target sites. mRNA transcripts produced via the process of splicing of two (or more) mRNAs from different gene sources are known as "fusion transcripts". It is also known that introns can be effectively targeted using antisense compounds targeted to, for example, DNA or pre-mRNA.
As is known in the art, a nucleoside is a base-sugar combination. The base portion of the nucleoside is normally a heterocyclic base. The two most common classes of such heterocyclic bases are the purines and the pyrimidines. Nucleotides are nucleosides that further include a phosphate group covalently linked to the sugar portion of the nucleoside. For those nucleosides that include a pentofuranosyl sugar, the phosphate group can be linked to either the 2', 3' or 5' hydroxyl moiety of the sugar. In forming oligonucleotides, the phosphate groups covalently link adjacent nucleosides to one another to form a linear polymeric compound. In turn, the respective ends of this linear polymeric compound can be further joined to form a circular compound, however, linear compounds are generally preferred. In addition, linear compounds may have internal nucleobase complementarity and may, therefore, fold in a manner as to produce a fully or partially double-stranded compound. Within oligonucleotides, the phosphate groups are commonly referred to as forming the internucleoside backbone of the oligonucleotide. The normal linkage or backbone of RNA and DNA is a 3' to 5' phosphodiester linkage.
Specific examples of preferred antisense compounds useful in this invention include oligonucleotides containing modified backbones or non-natural internucleoside linkages. As defined in this specification, oligonucleotides having modified backbones include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone. For the purposes of this specification, and as sometimes referenced in the art, modified oligonucleotides that do not have a phosphorus atom in their internucleoside backbone can also be considered to be oligonucleosides.
Preferred modified oligonucleotide backbones containing a phosphorus atom therein include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3 '-alkylene phosphonates, 5'-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3 '-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, selenophosphates and boranophosphates having normal 3 '-5' linkages, 2 '-5' linked analogs of these, and those having inverted polarity wherein one or more internucleotide linkages is a 3' to 3', 5' to 5' or 2' to 2' linkage. Preferred oligonucleotides having inverted polarity comprise a single 3' to 3' linkage at the 3 '-most internucleotide linkage i.e. a single inverted nucleoside residue which may be abasic (the nucleobase is missing or has a hydroxyl group in place thereof). Various salts, mixed salts and free acid forms are also included.
Antisense oligonucleotides are particularly useful in the treatment of inflammatory conditions of the skin. The antisense oligonucleotides may target both the Rel/NF-κB pathway as well as the immune system. These can also be topically applied, generally in a cream-based composition.
In an alternative embodiment, genetic constructs including DNA vaccines are used to generate antisense molecules in vivo. Furthermore, many of the preferred features described above are appropriate for sense nucleic acid molecules or for gene therapy applications to promote levels of Rel/NF-κB components. Topical-based compositions are particularly useful in the treatment of inflammatory conditions of the skin.
Following identification of an agent which potentiates the Rel/NF-κB signaling pathway, it may be manufactured and/or used in a preparation, i.e. in the manufacture or formulation or a composition such as a medicament, pharmaceutical composition or drug. These may be admimstered to individuals in a method of treatment or prophylaxis or regenerates therapy. Alternatively, they may be incorporated into a patch or slow release capsule or implant.
Thus, the present invention extends, therefore, to a pharmaceutical composition, medicament, drug or other composition including a patch or slow release formulation comprising an agonist or antagonist of target activity or gene expression. Another aspect of the present invention contemplates a method comprising administration of such a composition to a patient such as for treatment or prophylaxis of an inflammatory condition. Furthermore, the present invention contemplates a method of making a pharmaceutical composition comprising admixing a compound of the instant invention with a pharmaceutically acceptable excipient, vehicle or carrier, and optionally other ingredients. Where multiple compositions are provided, then such compositions may be given simultaneously or sequentially. Sequential administration includes administration within nanoseconds, seconds, minutes, hours or days. Preferably, within seconds or minutes.
Two- or multi-part pharmaceutical compositions or packs are also contemplated where multiple components such as comprising only those which potentiate the Rel/NF-κB pathway or both these latter components together with those which inhibit an immune response (locally or systemically). Such multi-part pharmaceutical compositions or packs maintain different agents or groups of agents separately. These are either dispensed separately or admixed prior to being dispensed.
Accordingly, another aspect of the present invention contemplates a method for the treatment or prophylaxis of an inflammatory condition in an animal, said method comprising administering to said animal an effective amount of a compound as described herein or a composition comprising same.
The term "administering to" includes the topical application of a composition to target tissue such as skin. Preferably, the animal is a mammal such as a human or laboratory test animal such as a mouse, rat, rabbit, guinea pig, hamster, zebrafish or amphibian.
This method also includes providing a wild-type or mutant target gene function to a cell. This is particularly useful when generating an animal model. Alternatively, it may be part of a gene therapy approach. A target gene or a part of the gene may be introduced into the cell in a vector such that the gene remains extracl romosomal. In such a situation, the gene will be expressed by the cell from the extrachromosomal location. If a gene portion is introduced and expressed in a cell carrying a mutant target allele, the gene portion should encode a part of the target protein. Vectors for introduction of genes both for recombination and for extrachromosomal maintenance are known in the art and any suitable vector may be used. Methods for introducing DNA into cells such as elecfroporation calcium phosphate co-precipitation and viral transduction are known in the art.
Gene transfer systems known in the art may be useful in the practice of genetic manipulation. These include viral and non-viral transfer methods. A number of viruses have been used as gene transfer vectors or as the basis for preparing gene transfer vectors, including papovaviruses (e.g. SV40, Madzak et al, J. Gen. Virol. 73: 1533-1536, 1992), adenovirus (Berkner, Curr. Top. Microbiol. Immunol. 158: 39-66, 1992; Berkner et al, BioTechniques 6; 616-629, 1988; Gorziglia and Kapikian, J. Virol 66: 4407-4412, 1992; Quantin et al, Proc. Natl. Acad. Sci. USA 89: 2581-2584, 1992; Rosenfeld et al, Cell 68: 143-155, 1992; Wilkinson et al, Nucleic Acids Res. 20: 2233-2239, 1992; Stratford- Perricaudet et al, Hum. Gene Ther. 1: 241-256, 1990; Schneider et al, Nature Genetics 18: 180-183, 1998), vaccinia virus (Moss, Curr. Top. Microbiol. Immunol. 158: 25-38, 1992; Moss, Proc. Natl. Acad. Sci. USA 93: 11341-11348, 1996), adeno-associaied virus (Muzyczka, Curr. Top. Microbiol. Immunol. 158: 97-129, 1992; Ohi et al, Gene 89: 279- 282, 1990; Russell and Hirata, Nature Genetics 18: 323-328, 1998), herpesviruses including HSV and EBV (Margolskee, Curr. Top., Microbiol Immunol. 158: 67-95, 1992; Johnson et al, J. Virol. 66: 2952-2965, 1992; Fink et al, Hum. Gene Ther. 3: 11-19, 1992; Breakefield and Geller, Mol. Neurobiol 1: 339-371, 1987; Freese et al, Biochem. Pharmacol. 40: 2189-2199, 1990; Fink et al, Ann. Rev. Neurosci. 19: 265-287, 1996), lentiviruses (Naldini et al, Science 272: 263-267, 1996), Sindbis and Semliki Forest virus (Berglund et al, Biotechnology 11: 916-920, 1993) and refroviruses of avian (Bandyopadhyay and Temin, Mol. Cell. Biol. 4: 749-754, 1984; Pefropoulos et al, J. Viol. 66: 3391-3397, 1992], murine [Miller, Curr. Top. Microbiol. Immunol. 158: 1-24, 1992; Miller et al, Mol. Cell. Biol. 5: 431-437, 1985; Sorge et al, Mol. Cell. Biol. 4: 1730-1737, 1984; and Baltimore, J. Virol. 54: 401-407, 1985; Miller et al, J. Virol. 62: 4337-4345, 1988] and human [Shimada et al, J. Clin. Invest. 88: 1043-1047, 1991; Helseth et al, J. Virol 64: 2416-2420, 1990; Page et al, J. Virol. 64: 5270-5276, 1990; Buchschacher and Panganiban, J. Virol. 66: 2731-2739, 1982] origin.
Non-viral gene transfer methods are known in the art such as chemical techniques including calcium phosphate co-precipitation, mechanical techniques, for example, microinjection, membrane fusion-mediated transfer via liposomes and direct DNA uptake and receptor-mediated DNA transfer. Viral-mediated gene transfer can be combined with direct in vivo gene transfer using liposome delivery, allowing one to direct the viralvectors to particular cells. Alternatively, the retroviral vector producer cell line can be injected into particular tissue. Injection of producer cells would then provide a continuous source of vector particles.
In an approach which combines biological and physical gene transfer methods, plasmid DNA of any size is combined with a polylysine-conjugated antibody specific to the adenovirus hexon protein and the resulting complex is bound to an adenovirus vector. The trimolecular complex is then used to infect cells. The adenovirus vector permits efficient binding, intemalization and degradation of the endosome before the coupled DNA is damaged. For other techniques for the delivery of adenovirus based vectors, see U.S. Patent No. 5,691,198.
Liposome/DNA complexes have been shown to be capable of mediating direct in vivo gene transfer. While in standard liposome preparations the gene transfer process is non-specific, localized in vivo uptake and expression have been reported in tumor deposits, for example, following direct in situ administration.
If the polynucleotide encodes a sense or antisense polynucleotide or a ribozyme or DNAzyme, expression will produce the sense or antisense polynucleotide or ribozyme or DNAzyme. Thus, in this context, expression does not require that a protein product be synthesized. In addition to the polynucleotide cloned into the expression vector, the vector also contains a promoter functional in eukaryotic cells. The cloned polynucleotide sequence is under control of this promoter. Suitable eukaryotic promoters include those described above. The expression vector may also include sequences, such as selectable markers and other sequences described herein.
Cells and animals which carry mutant target alleles (e.g. IKBS) or where one or both alleles are deleted can be used as model systems to study the effects of Rel/NF-κB signaling or inflammation. Mice, rats, rabbits, guinea pigs, hamsters, zebrafish and amphibians are particularly useful as model systems. A particularly useful insertion is a loxP sequence flanking a target gene which can be excised by ere.
The present invention provides, therefore, a mutation in or flanking a genetic locus encoding a target. The mutation may be an insertion, deletion, substitution or addition to the target-coding sequence or its 5' or 3' untranslated region.
The animal model of the present invention is useful for screening for agents capable of ameliorating or mimicing the effects of a target. In one embodiment, the animal model produces low amounts of a target.
Another aspect of the present invention provides a genetically modified animal wherein said animal produces low amounts of a target relative to a non-genetically modified animal of the same species. Reference to "low amounts" includes zero amounts or up to about 10% lower than normalized amounts. Yet another aspect of the present invention provides multiple (i.e. two or more) genes which are modified.
The animal models of the present invention may be in the form of the animals including fish or may be, for example, in the form of embryos for transplantation. The embryos are preferably maintained in a frozen state and may optionally be sold with instructions for use.
The genetically modified animals may also produce larger amounts of a target.
Accordingly, another aspect of the present invention is directed to a genetically modified animal over-expressing genetic sequences encoding a target.
A genetically modified animal includes a transgenic animal, or a "knock-out" or "knock- in" animal as well as a conditional deletion mutant. Furthermore, co-suppression may be used to induce post-transcriptional gene silencing. Co-suppression includes induction of RNAi.
The compounds, agents, medicaments, nucleic acid molecules and other target antagonists or agonists of the present invention can be formulated in pharmaceutical compositions which are prepared according to conventional pharmaceutical compounding techniques. See, for example, Remington's Pharmaceutical Sciences, 18 Ed. (1990, Mack Publishing, Company, Easton, PA, U.S.A.). The composition may contain the active agent or pharmaceutically acceptable salts of the active agent. These compositions may comprise, in addition to one of the active substances, a pharmaceutically acceptable excipient, carrier, buffer, stabilizer or other materials well known in the art. Such materials should be non- toxic and should not interfere with the efficacy of the active ingredient. The carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g. topical, intravenous, oral, intrathecal, epineural or parenteral. For oral administration, the compounds can be formulated into solid or liquid preparations such as capsules, pills, tablets, lozenges, powders, suspensions or emulsions. In preparing the compositions in oral dosage form, any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, suspending agents, and the like in the case of oral liquid preparations (such as, for example, suspensions, elixirs and solutions); or carriers such as starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations (such as, for example, powders, capsules and tablets). Because of their ease in administration, tablets and capsules represent the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be sugar-coated or enteric-coated by standard techniques. The active agent can be encapsulated to make it stable to passage through the gastrointestinal tract while at the same time allowing for passage across the blood brain barrier. See for example, International Patent Publication No. WO 96/11698.
For parenteral administration, the compound may dissolved in a pharmaceutical carrier and administered as either a solution of a suspension. Illustrative of suitable carriers are water, saline, dextrose solutions, fructose solutions, ethanol, or oils of animal, vegetative or synthetic origin. The carrier may also contain other ingredients, for example, preservatives, suspending agents, solubilizing agents, buffers and the like. When the compounds are being administered intrathecally, they may also be dissolved in cerebrospinal fluid.
The active agent is preferably administered in a therapeutically effective amount. The actual amount administered and the rate and time-course of administration will depend on the nature and severity of the condition being treated. Prescription of treatment, e.g. decisions on dosage, timing, etc. is within the responsibility of general practitioners or specialists and typically takes account of the disorder to be treated, the condition of the individual patient, the site of delivery, the method of administration and other factors known to practitioners. Examples of techniques and protocols can be found in Remington's Pharmaceutical Sciences, supra. Alternatively, targeting therapies may be used to deliver the active agent more specifically to certain types of cell, by the use of targeting systems such as antibodies or cell specific ligands or specific nucleic acid molecules. Targeting may be desirable for a variety of reasons, e.g. if the agent is unacceptably toxic or if it would otherwise require too high a dosage or if it would not otherwise be able to enter the target cells.
Instead of administering these agents directly, they could be produced in the target cell, e.g. in a viral vector such as described above or in a cell based delivery system such as described in U.S. Patent No. 5,550,050 and International Patent Publication Nos. WO 92/19195, WO 94/25503, WO 95/01203, WO 95/05452, WO 96/02286, WO 96/02646, WO 96/40871, WO 96/40959 and WO 97/12635. The vector could be targeted to the target cells. The cell based delivery system is designed to be implanted in a patient's body at the desired target site and contains a coding sequence for the target agent. Alternatively, the agent could be administered in a precursor form for conversion to the active form by an activating agent produced in, or targeted to, the cells to be treated. See, for example, European Patent Application No. 0 425 731 A and International Patent Publication No. WO 90/07936.
The present invention is further described by the following non-limiting Examples.
EXAMPLE 1
Materials and methods
Mice
c-Rel generated as previously described were used (Kontgen et al, Genes Dev. 9: 1965- 1977, 1995) and maintained on a C57BL/6 background. The RelA+/" and the NF-κBl7" mice are as previously described (Beg et al, Nature 376: 167-170, 1995; Sha et al, Cell 80: 321-330, 1995) and were on a mixed (C57BL/6 x 129) and C57BL/6 (> 9 generations) background, respectively. Due to the embryonic lethality of RelA mice, they were maintained at heterozygosity. The TNF-α7" mice (Korner et al, J. Exp. Med. 186: 1585- 1590, 1997) were on a C57BL/6 background. All mice were housed under specific pathogen free conditions.
The RelA"7" c-Rel"7" TNF-α"/_ triple mutant mice were derived by first mating RelA+/" c-Rel" _ TNF-α+/+ mice with the RelA+/+ c-Rel+/+ TNF-α_ " mice. Sibling matings were set up with mice that were heterozygous for all three genes. The resulting offspring, RelA+/" c-Rel7" TNF-α"/_, were viable and fertile. Subsequent sibling matings were set up to derive the triple mutant mice. This rationale was also employed to derive the RelA7" NF-κBl7" TNF- α7" triple mutant mice. The RelA c-Rel7" TNF-α7" triple mutants did not survive to weaning age. Timed mating revealed that these mice died within 12 hours after birth. Consequently, triple mutants were taken at embryonic day 18 from timed matings where day 0 was defined upon observation of a vaginal plug.
Immunohistochemistry and histolosv
Skin, embryo and paraffin section samples were taken from day 18 embryos and fixed overnight in 4% v/v paraformaldehyde. Paraffin embedded skin was cut into 5 μm sections and mounted onto AES coated slides. Sections were incubated for 1 hr in a humidified chamber with primary antibodies, washed extensively in PBS and then incubated for a further hour with the universal horse anti-mouse biotinylated secondary antibody (Vector Labs). After washing in PBS 0.2% v/v Triton-x-100 sections were incubated for 30 mins with the A/B kit (Vector Labs) and the staining was developed with DAB for 5 mins. The primary antibodies used in this study were MoAb PCNA (clone PC10; Pharmingen); rabbit anti-mouse keratin-5, keratin-6, keratin- 14, keratin- 10, and filaggrin (BabCO). For frozen sections, dorsal skin was embedded in OCT (Tissue-Tek), snap forzen on dry ice and sectioned at a thickness of 5 μm. Again, tissues were mounted onto AES coated slides and fixed in acetone.
Paraffin tissue sections were deparaffinized and rehydrated through a graded ethanol series. The sections were blocked with a 1% BSA, 10% normal goat serum (NGS), 0.2%) Triton-X-100 solution. Primary antibodies used were a mouse anti-human PCNA mAb (clone PC10/IgG2a; Pharmingen) and rabbit anti-keratin-6 antibody. A goat anti-mouse secondary antibody was used to bind the anti-PCNA antibody (Santa Cruz) and all rabbit polyclonal antibodies were detected with the universal horse antirabbit secondary (Vector Labs). Tissues were then stained using the ABC peroxidase kit (Vector Labs) and counter stained with haematoxylin.
For indirect immunofluorescence, frozen sections were treated with a blocking solution (2% gelatin, 1% triton-XlOO, 5% FCS and 5% NGS in PBS). Three incubation steps were conducted to achieve double staining of tissue sections.
BrdU Stainins
Pregnant mothers were injected intraperitoneally with 100 μg BrdU (Sigma) per gram body weight and killed after 1 h of pulsing. Day 18 embryos were sacrificed and tissues taken for histological processing. Paraffin embedded skin sections were first treated with 0.3%) v/v H2O2 in 50% v/v methanol to block endogenous peroxidase. Inturn the DNA was pretreated with 2 N HC1 at 37°C followed by washing in 0.1 M Na2B4O7 pH 8.5 and PBS. The sections were then incubated in pepsin (0.4 mg/ml 0.1N HC1) for 20 mins at RT and then washed in PBS. Sections were incubated with anti-BrdU (Company) for 1 hr in a humidified chamber, washed in PBS and then incubated for a further hour with the secondary universal horse anti-mouse biotinylated secondary antibody (Vector Labs). Staining was subsequently developed as described above.
In situ hybridization
A probe encoding part of the mouse c-rel cDNA (nucleotides 403-1621: Genebank accession X15842) was cloned into pBKS. To produce a radiolabeled antisense riboprobe, this plasmid was linearised with Hindlll and transcribed with T7 RNA polymerase in the presence of [33P] -labelled UTP (Amersham). In situ hybridization was performed essentially as described (Thomas et al. Development 27:2537-2548, 2000).
TUNEL Staining
Skin sections were incubated with proteinase K (20 μg/ml) for 15 mins at RT, washed in PBS and the endogenous peroxidase was blocked with 3%> v/v H2O2 in methanol. TUNEL positive cells were subsequently detected by following the manufacturer's instructions to TUNEL staining (Promega and ApopTag TUNEL staining kit: Serologicals Corporation).
Isolation of Basal Keratinocytes and Flow Cvtometry
Skin flanks excised from day 18 embryos were washed in chilled mouse phosphate buffered saline (Mt.PBS) and then incubated overnight at 4°C in Dispase II (2 mg/mL). The epidermis was gently teased away from the dermis and then subjected to 3 mins of pipetting in the presence of tissue culture grade trypsin to release basal keratinocytes. The reaction was inhibited by the addition of Soybean Inliibitor and then the cells were either cultured or prepared for Fluorescence Activated Cell Sorting using the Mo-Flo (Bectin Dickenson). For cells sorting basal keratinocytes were stained with the rat anti-human α6 integrin and anti-mouse CD71 (transferrin receptor). The stained cells were incubated on at 4°C for 30 mins and then washed. Cell culture and stains.
Isolated basal keratinocytes were seeded at a density of 106 cells in a 6-well plate (Costar) in serum-free keratinocyte media (Gibco-BRL) supplemented with hydrocortisone (0.5μg/ml) and low levels of CaC12 (0.02 mM). Cultures were fixed in 2% v/v formaldehyde and subjected to immunoperoxidase staining for keratin 14 (LL001, IgG2a). Cells were then incubated with biotinylated secondary antibodies (Vector Laboratories), followed by streptavidin-HRP (ABC kit, Vector Laboratories) and enzyme substrate (AEC substrate kit, Vector Laboratories).
Flow cvtometry and cell cycle analysis.
Basal keratinocytes were stained with FITC conjugated rat anti-human integrin-a6 antibody (BD Pharmingen) and PE conjugated anti-mouse CD71 antibody (BD Pharmingen) in a two-colour reaction or stained with a FITC conjugated anti-mouse CD29 antibody (integrin-bl) (Cymbus Biotechnology) in a single colour reaction. Stained keratinocytes were either cell sorted or analysed immediately using a FACScan. Propidium iodide (PI) (20μg/ml) was added to exclude dead cells during the analysis. For cell cycle analysis on a FACScan, TA cells (integrin-aόhi CD71hi) were fixed with chilled 70% ethanol and treated with PI and ribonuclease. Cell cycle profiles were analysed using the Modfit software program.
Skin Grafting
To follow the skin development of the RelA7" c-Rel7" TNF-α7" null mice, the skin from day 18 embryos was surgically transplanted onto immunocompromised Rag7" mice. About 1 cm2 of dorsal skin was excised from genotyped day 18 embryos and kept in Mt.PBS. The BόRag mice (11-15 weeks of age) were anaesthetized with Penthrane and the hair removed by plucking from the flank region above the ribcage. The graft bed was prepared by using a pair of curved scissors to remove a 1.5 cm2 area of recipient skin. The donor skin was placed into the graft bed ensuring no contact between the donor and recipient skin. The graft was dressed by adding a 1.5 cm2 piece of Jelonet gauze (Smith & Nephew) over the entire graft bed and then bandaged with 3 M micropore surgical tape, followed by 3 M transpore surgical tape. The mice were maintained in separate cages to avoid fighting between mice and, hence, possible graft damage. After eight days, the recipient mice were re-anaesthetized and bandages slowly cut away from the graft site. The grafts were examined 2-3 times weekly up to four weeks post-transplant. The donor graft was distinguished from the recipient coat by its agouti hair color.
Skin permeability assay.
Epidermal barrier function was determined as previously described (Hardman et al. Development 725:1541-1552, 1998).
Northern and western blotting.
5 μg samples of total RNA isolated from purified basal keratinocytes was subjected to Northern blotting essentially as described (Grumont βt al. Mol Cell 70:1283-1294,2002). Filters were sequentially probed with a 1.4 kb Xhol mouse c-myc cDNA, a rat 1.1 kb Pstl GAPDH cDNA and a 0.4 kb BamHl mouse K14 cDNA. nProtein blots were performed essentially as described (Nakamura Mol Cell Biol 22:5563-5574, 2002) using equivalent amounts of total cellular protein isolated from purifiedncontrol and mutant keratinocytes. Filters were probed with either mouse anti-cyclin DI monoclonal Ig (Santa Cruz Biotechnology), rabbit anti-cyclin D2 polyclonal Ig (Santa Cruz Biotechnology) or rat anti- HSP70 monoclonal antibody.
Cell size measurements.
Each of 4 control (rela+/+c-rel-/-tnf a-/-) and mutant (rela-/-c-rel-/-tnf a-/-) fetuses were used for epidermal basal keratinocyte size measurements. To avoid stretching and contraction artefacts, skin overlying the nuchal fat pad attached to the body of El 8.5 and
E19.5 pups was analysed. Images of control and mutant skin were taken at lOOOx magnification using a compound microscope and a digital camera (Zeiss). Images were analysed using an image analysis program (NIH Image 1.63 f). The areas of all basal cell profiles were measured per image. Seven to nine images representing 85 to 112 basal cells were analysed per animal. Results are given as mean ± standard error of the mean. Data were analysed statistically using a two-factorial analysis of variance with the genotype and the developmental age as the factors and Scheffe's method for pair wise comparison using StatView 4.5 software.
EXAMPLE 2 Disruption of the TNF-signaling pathway rescues embryonic death associated with the combined losses ofNF-κBl and RelA or RelA and c-Rel
In order to examine what overlapping roles NF-kB 1 and RelA or c-Rel and RelA might serve during skin development, these compound Rel/NF-kB mutants were generated on a TNF-a deficient background to overcome the death at -E14.5. The birth of nfkbl-Z-rela-/- tnf -/- and relα-/-c-rel-/-tnf a-/- mice from the respective relα+/-c-rel-/-tnf -/- or nfkbl-/- relα+/-tnf a-/- intercrosses established that if TNF-a mediated hepatocyte apoptosis was prevented, the absence of NF-kB 1 or c-Rel in combination with a lack of RelA did not prevent murine embryogenesis from proceeding beyond E14.5. Whilst relα-/-c-rel-/-tnf a-/- mice were born at a frequency of ~25%, the number of newborn nf kbl-/-relα-/-tnf a-/- mice was lower than expected, suggesting that some embryonic death at a developmental stage yet to be determined occurred in the absence of NF-kB 1 and RelA. The gross morphology of El 8 (Fig. 1A) and newborn relα-/-c-rel-/-tnf a-/- and nfkbl-/-relα-/-tnfa-/- mice appeared relatively normal. The most notable difference was a reduced body weight of -30% for relα-/-c-rel-/-tnf a-/- El 8 fetuses and newborn mice when compared with littermate controls. Despite the relatively normal appearance of relα-/-c-rel-/-tnf a-/- and nf kbl-/-relα-/-tnf a-/- newborns, both mutants died within 12 hrs of birth from as yet unknown causes.
Concerns that stress associated with the neonatal death of these mutants might indirectly affect skin architecture prompted us to perform all subsequent studies on fetal skin. The dorsal skin was focused upon as it represents a location in which epidermal and hair follicle morphogenesis is well advanced in a fetus. Microscopic examination of stained histological sections revealed that at El 8, control fetuses (wt, tnfa-/-, c-rel-/-, nfkbl-/-, nf kbl-/-tnfa-/-, c-r l-/ -tnf a-/- and relα-/ -tnf 'a-/-, of which the c-r l-/ -tnf 'a-/- skin shown in Fig. IB is a typical example) displayed a well-defined epidermis with numerous hair follicles at different stages of differentiation. Although nf kbl-/-relα-/-tnf a-/- embryonic skin (Fig. IC) had a similar appearance to that of controls, a marginal 11 reduction in epidem al thickness was consistently noted. In contrast, the relα-/-c-rel-/-tnf a-/- epidermis was distinctly thinner and hair follicles comprised only a few rudimentary buds (Fig. ID). As the skin defects were most profound in the combined absence of RelA and c-Rel, all subsequent analysis focused on the relα-/-c-rel-/-tnf a-/- mice.
EXAMPLE 3 c-Rel is expressed in the epidermis and in hair follicles.
Epidermal and hair follicle defects in the combined absence of RelA and c-Rel indicated that both transcription factors probably shared overlapping patterns of expression within the skin. Whilst a role for RelA in the epidermis was consistent with its ubiquitous expression, c-Rel function was thought to be hemopoietic restricted. To assess the relationship between the skin defects observed in the rela-/-c-rel-/-tnf a-/- mice and c-rel expression in the skin, in situ hybridization was performed on sagittal sections of C57BL/6 embryos at E15.5 and back skin sections from a C57BL/6 E18.5 fetus. In E15.5 embryos, no c-rel expression above background could be detected in the skin (results not shown). At El 8.5, low level expression of c-rel mRNA was detected in the epidermis and in hair follicles (Fig. 2A,B). In the epidermis, c-rel mRNA was present in basal cells, as well as in the spinous and granular layers (Fig. 2E,F). Differentiating keratinocytes appeared to maintain c-rel expression whilst these cells contained a nucleus, suggesting that its expression continued until the terminal stage of keratinocyte differentiation. In hair follicles, c-rel was expressed in the sheath cells, the hair bulb and in the dermal papilla (Fig. 2E). c-rel was also expressed in a few scattered cells within the dermis (Fig. 2E). These findings confirm that c-rel is normally expressed in those skin structures that develop abnormally in rela-/-c-rel-/-tnf a-/- mice.
EXAMPLE 4 Impaired hair follicle morphogenesis in rela-A c-rel-/- tnfa-/- mice.
Mice have four hair types (tylotrich, awl, auchene and zig zag) (Mann Anat Rec 144:135- 141, 1962). During embryogenesis two distinct waves of hair follicle morphogenesis occur. The first wave (tylotrich or guard hair) commences around E14. By E17, a second wave of hair placodes appear, coinciding with the initiation of owl hair development. To determine at which point during embryogenesis follicle development is impaired in the absence of RelA and c-Rel, histological sections of El 6, 17 and 18 control and mutant embryos were examined. In control embryos (relα+/+c-rel-/-tnf a-/-) hair placodes were present in El 6 dorsal skin, consistent with the first wave of placode development having been initiated (Fig. 3 A). At El 7, new hair placodes were present, and follicles associated with the first wave of hair development had begun to develop hair shaft cells (Fig. 3B). By El 8, the development of both groups of follicles had progressed further (Fig. 3C). By contrast, mutant skin (relα-/-c-rel-/-tnf a-/-) showed no evidence of placode formation at El 6 (Fig. 3 A). However, by El 7, the first hair placodes were observed in mutant fetuses, albeit in markedly reduced numbers (Fig. 3B). In the skin of El 8 mutant mice, only a few premature follicles were present and these had failed to develop hair shafts (Fig. 3C). Whiskers were also examined in serial sections of control and mutant fetuses. No obvious defects were observed in the maturation or number of developing mystacial and supernumerary vibrissae follicles of relα-/-c-rel-/-tnf a-/- fetuses (Fig. 3D), indicating that c-Rel and RelA are dispensible for whisker development. Interestingly, an examination of the embryonic teeth, which are epidermal appendages, revealed that the sizes of the molars and incisors were abnormally small in the relα-/ -c-rel-/ -tnf a-/- fetuses (Fig 3E). As the developmental status of the surrounding structures appeared normal for an El 8 fetus, the smaller teeth in the mutants was unlikely to reflect a delay in development. Collectively, these findings indicate that RelA and c-Rel are required for the normal development of certain hair types and epithelial appendages, such as teeth. EXAMPLE 5 Abnormal epidermal development in the combined absence of RelA and c-Rel.
Epidermal development was investigated in rela-/-c-rel-/-tnf a-/- mice. A comparison of H&E stained sections from El 8 control and mutant mice revealed the basal (B), spinous (S), granular (G) and cornified (C) layers of the skin were present in fetuses of both genotypes (Fig. 4A,B). However, the mutant epidermis was noticeably thinner, with fewer differentiating keratinocytes in the suprabasal region compared with age matched controls (Fig. 3C and 4A,B). The cells in the basal layer of rela-/-c-rel-/-tnf a-/- skin were organized differently from that of controls and the basal cell profiles appeared smaller in stained sections of rela-/-c-rel-/-tnf 'a-/- fetuses (Fig. 4A,B). Measurement of basal cell size in rela-/-c-rel-/-tnf a-/- and relα+/+c-rel-/-tnf a-/- mice confirmed that the mean basal cell profile area was reduced by -33% (0.397 ± 0.014 vs. 0.592 ± 0.019, PO.0001) in the relα- /-c-rel-/-tnf a-/- mutant. The age of the fetus (E18.5 or E19.5) did not significantly influence the cell profile size ( =0.67).
To determine if keratinocyte differentiation was impaired in the absence of c-Rel and RelA, the expression of proteins that characterize specific epidermal layers was examined. K14 protein is normally expressed in basal cells and hair follicles (Fuchs and Byme Curr Opn Genet Dev 4:725-736, 1994). Although anti-K14 antibodies stained the inner most basal cells at the dermal/epidermal boundary of both control and mutant skin (Fig. 4C,D), staining which was usually two cell layers thick in the control was generally confined to one cell layer in the mutant, with little spinous staining. The early differentiation marker K10 is expressed predominantly in the suprabasal spinous and granular layers (Fuchs and Byme 1994 supra). In the control epidermis, anti-KlO antibody staining of the suprabasal layers was at least three cell layers thick. In the mutant epidermis, K10 expression was confined to two cell layers (Fig. 4E,F). The late differentiation markers involucrin and loricrin, are major components of the cornified envelope, with involucrin expressed in the upper spinous and granular layers, whilst loricrin is restricted to the granular layer. Involucrin and loricrin staining was confined to these layers in the mutant epidermis, the region in which these proteins were expressed being somewhat compacted compared with control skin (Fig. 4G,H,I,J).
The stratification of the mutant skin was next examined using double immunofluorescence staining. In control epidermis, co-staining with antibodies to K14 and K10 revealed a significant region of co-localization, indicative of keratinocytes leaving the basal layer and entering the spinous layer (Fig. 4K). Co-localization of K14 and K10 was reduced in the mutant epidermis (Fig. 4L). Staining for K14 and loricrin revealed that basal and spinosal expression of these proteins in the granular layer was distinct in both control and mutant epidermis (Fig. 4M,N). Whilst co-staining for K10 and involucrin revealed the expected pattern of overlapping expression in the upper spinous and granular layers, the region of co-expression was markedly reduced in mutant fetuses compared with controls (Fig. 4 O,P). The region of K10 and loricrin co-expression was only marginally reduced in the mutant epidermis (Fig. 4 Q,R) and the expression of filaggrin, a marker for terminally differentiated enucleated squames was equivalent in control and mutant epidermis. Taken together, these data show that the suprabasal layers were reduced in rela-/-c-rel-/-tnf a-/- fetal skin.
To determine if the differences in the mutant epidermis resulted in barrier dysfunction, dye penetration assays were performed by immersing control and mutant El 8 fetuses overnight in an X-gal solution. These experiments demonstrated that dye did not penetrate the skin of either control or mutant fetuses (Fig. 4 S,T), except at the site of the tail biopsy wound. Hence barrier function of the mutant epidermis is intact. Collectively, these findings establish that in the combined absence of c-Rel and RelA, epidermal development is abnormal, but keratinocytes still appear able to undergo terminal differentiation. EXAMPLE 6
Epidermal stem cell and transit-amplifying cell numbers are normal in the absence of c-Rel and RelA.
Cell surface marker expression has been used to delineate the stem cell, TA cell and maturing keratinocyte populations in the epidermis. Stem cells and TA cells both express integrin bl with the highest levels expressed by stem cells. Integrin a6 expression is high in stem cells and TA cells, whereas keratinocytes committed to differentiate express low levels of this integrin. Transferrin receptor (CD71) expression has also been used to distinguish between quiescent stem cells (integrin aόhi CD711o) and proliferating TA cells (integrin aόhi CD71hi). To assess if the impaired epidermal development in the absence of c-Rel and RelA might be due to reduced numbers of epidermal stem cells and TA cells, equivalent areas of skin (0.5 cm2) were removed from the flank of El 8 control and mutant fetuses and epidermal basal cells isolated by protease treatment. Comparable numbers of basal cells were isolated from control [rela+/-c-rel-/-tnf a-/-; 7.3x105 ± 1.3 (n=4)] and mutant [rela-/-c-rel-/-tnf a-/-; 6.3x105 ± 1.8 (n=3)] embryonic skin. Flow cytometric analysis of these cells stained with anti-bl- integrin antibody (Fig. 5 A) demonstrated equivalent patterns of integrin-bl expression in control and mutant epidermal populations, indicating stem and TA cell numbers were comparable. This conclusion was generally reinforced by the staining profiles for integrin a6 and CD71 (Fig. 5B). However, in the mutant, a minor population of integrin aόhi CD71hi TA cells was markedly reduced (highlighted by the symbol *). This population probably represents cells in transition from TA cells to differentiating keratinocytes. Reduced numbers of this population in rela-/-c- r el-/ -tnf a-/- skin is consistent with fewer cells leaving the basal layer, a notion supported by the presence of fewer post-mitotic differentiating cells in the rela-/-c-rel-/-tnf a-/- epidermis (gate 3) (Fig. 5B). EXAMPLE 7 Epidermal basal cells lacking c-Rel and RelA exhibit a cell cycle defect.
Although normal numbers of keratinocyte progenitors are present in the epidermis of El 8 rela-/-c-rel-/-tnf a-/- mice and are able to undergo terminal differentiation, a thinner suprabasal layer in the mutant fetus indicated that epidermal differentiation was abnormal. Since Rel/NF-kB is required for lymphocyte and fibroblast proliferation, one plausible explanation for the reduced thickness of the epidermis was that a defect in cell division might influence the rate at which TA cells undergo terminal differentiation. Cell division within the mutant epidermis was initially examined in fetal skin sections by immunostaining for proliferating nuclear cell antigen (PCNA) expression, a marker of cells in the late Gl and S-phases of the cell cycle (Fig. 6A). In control skin, only some basal cells were PCNA positive, a finding consistent with a non-synchronous population of cells at different stages of the cell cycle. PCNA+ cells were also detected in developing hair follicles and the stratum spmosum. In contrast, the majority of cells in the mutant basal layer were PCNA+, but few positive cells were observed in the suprabasal region. To distinguish between basal keratinocytes in Gl and S-phase, BrdU incorporation studies were performed using day 18 fetuses (Fig. 6B). In control skin, BrdU was incorporated by cells within hair follicles and by basal keratinocytes, with -20% of basal cells being BrdU positive. However, in rela-/-c-rel-/-tnf a-/- fetal skin, only -9% of basal cells were BrdU positive.
The combined PCNA expression and BrdU incorporation studies indicated that the transition from Gl to S-phase may be impaired in mutant epidermal basal cells. This was confirmed by measuring the DNA content of FACS-sorted TA cells (integrin aόhi CD71hi) isolated from El 8 control and mutant fetuses. A typical set of data is shown in Fig. 6C. Consistent with the findings for BrdU incorporation, only 12-14% of mutant TA cells were in S-phase compared with 20-22%) of control TA cells. Moreover, whereas a comparable proportion of control and mutant TA cells were in G2/M (10% vs. 9% respectively), the reduced frequency of mutant TA cells in S-phase was accompanied by a corresponding increase of cells in G0/G1 (70 vs. 78 % for control and mutant TA cells respectively). These results indicated that TA cells lacking c-Rel and RelA exhibit a delay in Gl/S phase progression. This finding coupled with the reduced size of mutant c-rel-/-rela-/-tnf a-/- TA cells indicated that impaired cell growth may underly the cell cycle defect. In eukaryotic cells, growth is thought to proceed throughout Gl until a threshold is reached at which point levels of certain cellular components that function as indicators of cell size are sufficient to trigger entry into S-phase. Mitogen induced B cell growth is at least in part controlled by the Rel/NF-kB dependent induction of c-myc transcription, c-myc mRNA expression however was comparable in control and mutant epidermal TA cells (Fig. 6D). The levels of cyclins DI and D2, important regulators of Gl that are also known targets of Rel/NF-kB were also similar in control and mutant TA cells (Fig. 6E), as were the cdk inhibitors p21 and p27. This indicates that the impaired expression of an unknown Rel/NF- kB target gene is responsible for the Gl growth defect observed in c-rel-/-relα-/-tnf a-/- epidermal basal cells.
During B cell proliferation, Rel/NF-kB regulates Gl/S phase progression and promotes cell survival. To assess whether increased apoptosis accompanied impaired c-rel-/-relα-/- tnfa-/- TA cell division, TUNEL staining was performed on sections of control and mutant fetal skin (Fig. 6F). Mutant epidermis did not have abnormally increased numbers of TUNEL-positive cells. Dying cells were largely confined to the stratum granulosum of both control and mutant skin, a region of the epidermis in which keratinocytes undergo enucleation. Thus, the impaired cell cycle regulation of basal cells lacking RelA and c-Rel does not cause abnormally elevated cell death.
EXAMPLE 8 Keratinocytes lacking c-Rel and RelA fail to form colonies in culture.
To examine if the c-rel-/-rela-/-tnf a-/- TA cell cycle defect observed in vivo influenced the capacity of these cells to form keratinocyte colonies in culture, colony formation analysis was performed with El 8 control and mutant keratinocytes. Whereas numerous keratin- 14 positive colonies were evident in 7 day cultures of control cells, c-rel-/-rela-/-tnf a-/- keratinocytes generated only a few colonies that comprised 3-4 cells (Fig. 7 A). The defect in keratinocyte colony formation was only observed in the combined absence pf RelA and c-Rel (Fig. 7B) and coincided with high levels of cell death.
EXAMPLE 9 Hyperproliferation ofrela-/-c-rel-/-tnf-a-/- embryonic skin grafted onto rag-1-/- mice.
In order to examine the consequences of the loss of RelA and c-Rel for adult murine skin, El 8 control and mutant skin was transplanted onto immunocompromised rag-1-/- mice. The clinical appearance of a typical set of skin grafts over a 4-week period is shown in Figure 8A. Two weeks post-transplantation, the appearance of control and mutant donor skin was comparable. After 3 weeks there was abundant hair growth in control grafts. Only sparse hair growth was present in the mutant skin, which by 4 weeks had disappeared and was accompanied by scaling of the skin. Histology revealed a striking difference in the architecture of the control and mutant skin grafts. After 4 weeks, control grafts had an epidermis with a well-aligned basal cell layer, 1-2 cells thick (Fig. 8Bi). The mutant graft exhibited extensive epidermal hyperplasia, hyperkeratosis, focal parakeratosis, comeal pustules with a granulocytic infiltrate and necrosis (Fig. 8Bii). The mutant epidermis was abnormally thick (6-7 cell layers) with many hyperchromatic cells in the basal layer and a compacted stratum corneum (Fig. 8Ci,Cii). The dermis in the mutant graft was extensively infiltrated with immune cells, eosinophils being most prominent (Fig. 8Ci,Cii). Interestingly, numerous abnormal hair follicles were present in the mutant skin graft that consisted of enlarged sebaceous glands at the base of the follicles (Fig. 8Bii). Staining for melanin granules revealed extensive hyperpigmentation at the dermal/epidermal junction of the mutant graft (Fig. 8Civ), whereas in control grafts, melanin deposits were confined to the reticular dermis and hair bulbs (Fig. 8Ciii). Antibody staining for K6, a marker of proliferative skin conditions, confirmed that the increased thickness of the epidermis in the mutant skin graft was the result of keratinocyte proliferation (Fig. 8Biv), whereas K6 staining was confined to hair follicles in control skin grafts (Fig. 8Biii). EXAMPLE 10
The hyperproliferative state ofrela-/- c-rel-/- tnfa-/- embryonic skin grafts is dependent on TNF-a.
Hyperproliferation of rela-/-c-rel-/-tnf 'a-/- epidermal cells in the skin grafts was associated with an immune infiltration, suggesting that this condition may be due in part to these cells secreting inflammatory mediators. Consistent with this possibility was the recent finding that skin inflammation in mice arising from the conditional deletion of IKKb in epidermal basal cells could be prevented by loss of TNFR1. However, as TNFR1 binds the inflammatory mediators TNF-a and lymphotoxin (LTa), it was unclear if this inflammation was caused by one or both cytokines. Consequently, rela-/-c-rel-/-tnf a-/- El 8 fetal skin was grafted onto rag-Z-tnf a-/- recipients to determine if the hyperproliferative state was TNF-a dependent. Four weeks post-transplantation, mutant grafts showed no evidence of the hyperproliferative condition (Fig. 8Di,Dii). In mutant grafts, the epidermis was 2 to 3 cell layers thick and hair follicles were of relatively normal appearance. The extensive infiltration of leukocytes was markedly reduced, although some macrophages and granulocytic foci were still seen in the dermis (Fig. 8Diii). Despite the hyper-proliferative condition being eliminated in the absence of TNF-a, a higher frequency of PCNA+ basal cells was still detected in the mutant graft. This suggests that the cell cycle defect seen in mutant fetal skin may still afflict mutant epidermal cells in an adult enviroment.
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
Bandyopadhyay and Temin, Mol. Cell. Biol. 4: 749-754, 1984;
Beg et al, Nature 376: 167-170, 1995;
Berglund et al, Biotechnology 11: 916-920, 1993;
Berkner et al, BioTechniques 6; 616-629, 1988;
Berkner, Curr. Top. Microbiol Immunol. 158: 39-66, 1992;
Breakefield and Geller, Mol Neurobiol 1: 339-371, 1987;
Buchschacher and Panganiban, J. Virol. 66: 2731-2739, 1982;
Erickson et al, Science 249: 527-533, 1990;
Fink et al, Ann. Rev. Neurosci. 19: 265-287, 1996;
Ym et al, Hum. Gene Ther. 3: 11-19, 1992;
Freese et al, Biochem. Pharmacol. 40: 2189-2199, 1990;
Fuchs and Byrne Curr Opn Genet Dev 4:725-736, 1994
Gorziglia and Kapikian, J. Virol. 66: 4407-4412, 1992;
Grumont et al. Mol Cell 70:1283-1294,2002
Hardman et al. Development 725:1541-1552, 1998 Helseth et /., J. Virol. 64: 2416-2420, 1990;
Hodgson, Bio/Technology 9: 19-21, 1991;
Johnson et al, "Peptide Turn Mimetics" in Biotechnology and Pharmacy, Pezzuto et al. Eds., Chapman and Hall, New York, 1993;
Johnson et al. . Virol. 66: 2952-2965, 1992;
Kontgen et al, Genes Dev. 9: 1965-1977, 1995;
Komer et al, J. Exp. Med. 186: 1585-1590, 1997;
Madzak et α/., J. Gen. Virol. 73: 1533-1536, 1992;
Mann. Anat Rec 144:135-141, 1962
Mann and Baltimore, J. Virol. 54: 401-407, 1985;
Margolskee, Curr. Top., Microbiol. Immunol 158: 67-95, 1992;
Miller et al, J. Virol 62: 4337-4345, 1988;
Miller et al, Mol. Cell. Biol. 5: 431-437, 1985;
Miller, Curr. Top. Microbiol. Immunol. 158: 1-24, 1992;
Moss, Curr. Top. Microbiol. Immunol. 158: 25-38, 1992;
Moss, Proc. Natl. Acad. Sci. USA 93: 11341-11348, 1996; Munder et al, Appl. Microbiol. Biotechnol. 52(3): 311-320, 1999;
Muzyczka, Curr. Top. Microbiol. Immunol. 158: 97-129, 1992;
( :
Nakamura Mol Cell Biol 22:5563-5574, 2002
Naldini et al, Science 272: 263-267, 1996;
Ohi et al, Gene 89: 279-282, 1990;
Page et al, J. Virol. 64: 5270-5276, 1990;
Petropoulos et al, J. Viol 66: 3391-3397, 1992;
Quantin et al, Proc. Natl. Acad. Sci. USA 89: 2581-2584, 1992;
Remington's Pharmaceutical Sciences, 18th Ed. (1990, Mack Publishing, Company, Easton, PA, U.S.A.
Rosenfeld et al, Cell 68: 143-155, 1992;
Russell and Hirata, Nature Genetics 18: 323-328, 1998;
Sha et al, Cell 80: 321-330, 1995;
Schneider et al, Nature Genetics 18: 180-183, 1998;
Shimada et al, J. Clin. Invest. 88: 1043-1047, 1991;
Sorge et al, Mol. Cell. Biol. 4: 1730-1737, 1984; Stratford-Perricaudet et al, Hum. Gene Ther. 1: 241-256, 1990;
Summerton and Weller, Antisense and Nucleic Acid Drug Development 7: 187-195, 1997;
Tani et al, Proc. Natl. Acad. Sci. USA 97: 10960-1965, 2000;
Thomas et al. Development 727:2537-2548, 2000
Watt, EMBOJ. 21: 3919-3926, 2002;
Wells, Methods Enzymol 202: 2699-2705, 1991;
Wilkinson et al, Nucleic Acids Res. 20: 2233-2239, 1992;
Young et al, Nat. Biotechnol. 16(10): 946-950, 1998.

Claims

1. A composition when used for the treatment or prophylaxis of an inflammatory condition, said composition comprising a compound capable of up-regulating the Rel/NF- KB pathway in tissue or an organ being treated and one or more pharmaceutically acceptable carriers and/or diluents.
2. The composition of Claim 1 wherein the compound elevates the levels of NF- KB.
3. The composition of Claim 1 wherein the compound inhibits levels of IκB.
4. The composition of Claim 1 wherein the compound interacts with a cell surface receptor which is linked to the NF-κB pathway.
5. The composition of Claim 4 wherein the cell surface receptor is selected from BCR, IL-IR, TNFR, LTBR, CDl lb/CD185 CD28, CD2, CD35, CD3, CD40, CD4, fc-2a- receptor (IgG2a), Flt-1, LyόA/E, N-CAM, trail receptor- 1, trail-receptor-2 or trail- receptor-4.
6. The composition of Claim 1 wherein the compound is selected from IL-1, IL-2, IL-12, IL-15, IL-17, IL-18, LIF, THANK, TNF-α, TNF-β as well as any of the TNF- receptor superfamily ligands, 1-β-D-arabinofuranosyl-cytosine (ara-C), anthralin, azidothymidine (AZT), camptothecin, ciprofibrate, cisplatin, daunomycin, daunorubicin, doxorubicin, etoposide, haloperidol, methamphetamine, phenobarbital, tamoxifen, taxol (paclitaxel), vinblastine, vincristine, advanced glycated end products (AGEs), amyloid protein fragment (βA4), maleylated BSA, modified (oxidized) LDL, bone morphogenic protein 2, bone morphogenic protein 4, folicle stimulating hormone, human growth hormone, insulin, M-CSF, nerve growth factor, platelet-derived growth factor, serum, TGF-α, 12(R)-hydroxyeicosatrienoic acid, amino acid analogs, anaphylatoxin C3a, anaphylatoxin C5a, angiotensin II, basic calcium phosphate crystals, bradikinin, C2- ceramide (N-acetyl-sphingosine), cerulein, collagen lattice, collagen type I, Des-ArglO- kallidin (BI receptor agonist), double-stranded polynucleotides, f-Met-Leu-Phe, heat shock protein 60 (HSP 60), hemoglobin, hyaluronan, kaianic acid (kainate), leukotriene B4, L- glutamate, lysophosphatidylcholine (LysoPC), platelet activating factor (PAF), potassium, thrombin, 2-deoxyglucose, anisomycin, Brefeldin A, calchicine, calcium ionophores, calyculin A, cobalt chloride, Con A, cycloheximide, cyclopiazonic acid, forskolin, glass fibres, linoleic acid, L-NMA, lysophosphatidic acid, monensin, N-methyl-D-aspartate, nocodazol, okadaic acid, PHA, phorbol ester, podophyllotoxin, pyrogallol, guinolinic acid, thapsigargin or tunicamycin.
7. The composition of any one of Claims 1 to 6 further comprising an inhibitor of the immune system, systemically or locally.
8. A multi-part pharmaceutical pack comprising a first part or group of parts comprising a compound which up-regulates the Rel/NF-κB pathway and a second part or group of parts comprising an inhibitor of an immune response.
9. The multi-part pharmaceutical pack of Claim 8 wherein a compound in the first part elevates levels of NF-κB.
10. The multi-part pharmaceutical pack of Claim 8 wherein the compound inhibits levels of IκB.
11. The multi-part pharmaceutical pack of Claim 8 wherein a compound of the first part interacts with a cell surface receptor.
12. The multi-part pharmaceutical pack of Claim 11 wherein the cell surface receptor is selected from BCR, IL-IR, TNFR, LTBR, CDl lb/CD18, CD28, CD2, CD35, CD3, CD40, CD4, fc-2a-receptor (IgG2a), Flt-1, Ly6A/E, N-CAM, trail receptor-1, trail- receptor-2 or trail-receptor-4.
13. The multi-part pharmaceutical pack of Claim 8 wherein a compound of the first part is selected from IL-1, IL-2, IL-12, IL-15, IL-17, IL-18, LIF, THANK, TNF-α, TNF-β as well as any of the TNF -receptor superfamily ligands, 1-β-D-arabinofuranosyl- cytosine (ara-C), anthralin, azidothymidine (AZT), camptothecin, ciprofibrate, cisplatin, daunomycin, daunorubicin, doxorubicin, etoposide, haloperidol, methamphetamine, phenobarbital, tamoxifen, taxol (paclitaxel), vinblastine, vincristine, advanced glycated end products (AGEs), amyloid protein fragment (βA4), maleylated BSA, modified (oxidized) LDL, bone morphogenic protein 2, bone morphogenic protein 4, folicle stimulating hormone, human growth hormone, insulin, M-CSF, nerve growth factor, platelet-derived growth factor, serum, TGF-α, 12(R)-hydroxyeicosatrienoic acid, amino acid analogs, anaphylatoxin C3a, anaphylatoxin C5a, angiotensin II, basic calcium phosphate crystals, bradikinin, C2-ceramide (N-acetyl-sphingosine), cerulein, collagen lattice, collagen type I, Des-ArglO-kallidin (BI receptor agonist), double-stranded polynucleotides, f-Met-Leu- Phe, heat shock protein 60 (HSP 60), hemoglobin, hyaluronan, kaianic acid (kainate), leukotriene B4, L-glutamate, lysophosphatidylcholine (LysoPC), platelet activating factor (PAF), potassium, thrombin, 2-deoxyglucose, anisomycin, Brefeldin A, calchicine, calcium ionophores, calyculin A, cobalt chloride, Con A, cycloheximide, cyclopiazonic acid, forskolin, glass fibres, linoleic acid, L-NMA, lysophosphatidic acid, monensin, N- methyl-D-aspartate, nocodazol, okadaic acid, PHA, phorbol ester, podophyllotoxin, pyrogallol, guinolinic acid, thapsigargin or tunicamycin.
14. A method for the treatment or prophylaxis of an inflammatory condition in an animal including avian species, said method comprising administering to said animal an effective amount of a compound which up-regulates the Rel/NF-κB pathway.
15. The method of Claim 14 wherein the compound elevates the levels of NF-κB.
16. The method of Claim 14 wherein the compound inhibits levels of IκB.
17. The method of Claim 14 wherein the compound interacts with a cell surface receptor which is linked to the NF-κB pathway.
18. The method of Claim 17 wherein the cell surface receptor is selected from BCR, IL-IR, TNFR, LTBR, CDl lb/CD18, CD28, CD2, CD35, CD3, CD40, CD4, fc-2a- receptor (IgG2a), Flt-1, Ly6A/E, N-CAM, trail receptor- 1, trail-receptor-2 or trail- receptor-4.
19. The method of Claim 14 wherein the compound is selected from IL-1, IL-2, IL-12, IL-15, IL-17, IL-18, LIF, THANK, TNF-α, TNF-β as well as any of the TNF- receptor superfamily ligands, 1-β-D-arabinofuranosyl-cytosine (ara-C), anthralin, azidothymidine (AZT), camptothecin, ciprofibrate, cisplatin, daunomycin, daunorubicin, doxorubicin, etoposide, haloperidol, methamphetamine, phenobarbital, tamoxifen, taxol (paclitaxel), vinblastine, vincristine, advanced glycated end products (AGEs), amyloid protein fragment (βA4), maleylated BSA, modified (oxidized) LDL, bone morphogenic protein 2, bone morphogenic protein 4, folicle stimulating hormone, human growth hormone, insulin, M-CSF, nerve growth factor, platelet-derived growth factor, serum, TGF-α, 12(R)-hydroxyeicosatrienoic acid, amino acid analogs, anaphylatoxin C3a, anaphylatoxin C5a, angiotensin II, basic calcium phosphate crystals, bradikinin, C2- ceramide (N-acetyl-sphingosine), cerulein, collagen lattice, collagen type I, Des-ArglO- kallidin (B 1 receptor agonist), double-stranded polynucleotides, f-Met-Leu-Phe, heat shock protein 60 (HSP 60), hemoglobin, hyaluronan, kaianic acid (kainate), leukotriene B4, L- glutamate, lysophosphatidylcholine (LysoPC), platelet activating factor (PAF), potassium, thrombin, 2-deoxyglucose, anisomycin, Brefeldin A, calchicine, calcium ionophores, calyculin A, cobalt chloride, Con A, cycloheximide, cyclopiazonic acid, forskolin, glass fibres, linoleic acid, L-NMA, lysophosphatidic acid, monensin, N-methyl-D-aspartate, nocodazol, okadaic acid, PHA, phorbol ester, podophyllotoxin, pyrogallol, guinolinic acid, thapsigargin or tunicamycin.
20. The method of any one of Claims 14 to 19 further comprising the administration of an inhibitor of the immune system.
21. The method of Claim 14 wherein the animal is a mammal.
22. The method of Claim 21 wherein the mammal is a human.
PCT/AU2004/000300 2003-03-14 2004-03-12 Therapeutic and prophylactic compositions and uses therefor WO2004080478A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
AU2003901205 2003-03-14
AU2003901205A AU2003901205A0 (en) 2003-03-14 2003-03-14 Therapeutic and prophylactic compositions and uses therefor

Publications (1)

Publication Number Publication Date
WO2004080478A1 true WO2004080478A1 (en) 2004-09-23

Family

ID=31500249

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2004/000300 WO2004080478A1 (en) 2003-03-14 2004-03-12 Therapeutic and prophylactic compositions and uses therefor

Country Status (2)

Country Link
AU (1) AU2003901205A0 (en)
WO (1) WO2004080478A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009052454A3 (en) * 2007-10-19 2009-10-15 University Of California Compositions and methods for ameliorating cns inflammation, psychosis, delirium, ptsd or ptss
US20100075923A1 (en) * 2008-09-16 2010-03-25 Jung San Huang Method of enhancing tgf-beta signalling
US8946201B2 (en) 2007-08-27 2015-02-03 Saint Louis University Methods for inhibiting TGF-β
US9549914B2 (en) 2014-04-03 2017-01-24 The Johns Hopkins University Treatment of human cytomegalovirus by modulating Wnt
WO2019241663A1 (en) * 2018-06-15 2019-12-19 Afecta Pharmaceuticals, Inc. Ccl5 inhibitors
WO2023212841A1 (en) * 2022-05-05 2023-11-09 Wenzhou Medical University A kind of polypeptide analog with hair growth effect

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000018894A1 (en) * 1998-09-25 2000-04-06 The Research Foundation Of State University Of New York A BIOLOGICALLY ACTIVE ALTERNATIVE FORM OF THE IKKα IλB KINASE
US20040009463A1 (en) * 1999-03-26 2004-01-15 Michael Simons Method for PR-39 peptide mediated selective inhibition of IkappaBalpha

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000018894A1 (en) * 1998-09-25 2000-04-06 The Research Foundation Of State University Of New York A BIOLOGICALLY ACTIVE ALTERNATIVE FORM OF THE IKKα IλB KINASE
US20040009463A1 (en) * 1999-03-26 2004-01-15 Michael Simons Method for PR-39 peptide mediated selective inhibition of IkappaBalpha

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
GVERIC D. ET AL.: "Transcription factor NF-kappa B and inhibitor I kappa B alpha are localised in macrophages in active multiple sclerosis lesions", J. NEUROPATHOLOGY AND EXPERIMENTAL NEUROLOGY, vol. 57, no. 2, 1998, pages 168 - 178 *
HOGAN S.P. ET AL.: "Mucosal IL-12 gene delivery inhibits allergic airways disease and restores local antiviral immunity", EUR. J. IMMUNOL., vol. 28, 1998, pages 413 - 423 *
NICOLA N.A.: "Guidebook to cytokines and their receptors", 1997, OXFORD UNIVERSITY PRESS, pages: 17-21, - 103-109 *
WINDHOLZ M.: "The merck index", 1983, pages: 2810, - 5112 *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8946201B2 (en) 2007-08-27 2015-02-03 Saint Louis University Methods for inhibiting TGF-β
US9944583B2 (en) 2007-10-19 2018-04-17 The Regents Of The University Of California Methods for ameliorating and preventing central nervous system inflammation
US9550827B2 (en) 2007-10-19 2017-01-24 The Regents Of The University Of California Methods for ameliorating and preventing central nervous system inflammation
WO2009052454A3 (en) * 2007-10-19 2009-10-15 University Of California Compositions and methods for ameliorating cns inflammation, psychosis, delirium, ptsd or ptss
US10407374B2 (en) 2007-10-19 2019-09-10 The Regents Of The University Of California Compositions and methods for ameliorating CNS inflammation, psychosis, delirium, PTSD or PTSS
US8487006B2 (en) * 2008-09-16 2013-07-16 Auxagen, Inc. Method of enhancing TGF-β signalling
US20100075923A1 (en) * 2008-09-16 2010-03-25 Jung San Huang Method of enhancing tgf-beta signalling
US9549914B2 (en) 2014-04-03 2017-01-24 The Johns Hopkins University Treatment of human cytomegalovirus by modulating Wnt
WO2019241663A1 (en) * 2018-06-15 2019-12-19 Afecta Pharmaceuticals, Inc. Ccl5 inhibitors
US10940132B2 (en) 2018-06-15 2021-03-09 Afecta Pharmaceuticals, Inc. CCL5 inhibitors
US11318111B2 (en) 2018-06-15 2022-05-03 Lapko Inc CCL5 inhibitors
US11497724B2 (en) 2018-06-15 2022-11-15 Lapko Inc CCL5 inhibitors
US11628154B2 (en) 2018-06-15 2023-04-18 Lapko Inc CCL5 inhibitors
WO2023212841A1 (en) * 2022-05-05 2023-11-09 Wenzhou Medical University A kind of polypeptide analog with hair growth effect

Also Published As

Publication number Publication date
AU2003901205A0 (en) 2003-04-03

Similar Documents

Publication Publication Date Title
US20070037883A1 (en) Therapeutic compositions
US20070135336A1 (en) Follistatin isoforms and uses thereof
WO2006099667A1 (en) Prophylactic and therapeutic agents and uses therefor
US20070281986A1 (en) Methods and Compositions for Modulating Satiety
US20080254023A1 (en) Treating Gliosis, Glial Scarring, Inflammation or Inhibition of Axonal Growth in the Nervous System by Modulating Eph Receptor
WO2008025069A1 (en) Methods of modulating cellular activity and compositions therefor
US20070043512A1 (en) Therapeutic and prophylactic compositions and uses therefor
WO2004080478A1 (en) Therapeutic and prophylactic compositions and uses therefor
US20090077677A1 (en) Mammalian grainyhead transcription factors
US20070148129A1 (en) Therapeutic agents and uses therefor
EP1542528B1 (en) A method of treatment and prophylaxis
US20070275916A1 (en) Methods of Identifying Compounds Which Modulate Granulocite-Colony Stimulating Factor (G-Csf) Dependent Processes by Modulation of the Levels of Suppressor of Cytokine Signaling (Socs)
WO2008086573A1 (en) Use of galanin in a method of treating neurodegenerative diseases or conditions
US20090054307A1 (en) Prophylactic and therapeutic agents and uses therefor
US20070270352A1 (en) Method of Treatment
AU2003222681B8 (en) A method of treatment
US20060205688A1 (en) Method of modulating epithelial cell activity by modulating the functional levels of sphingosine kinase
WO2004099412A1 (en) Nucleic acid molecules differentially expressed in animals exhibiting behavioural disorders
WO2011038469A1 (en) Suppressive agents
WO2006029462A1 (en) A nucleic acid molecule differentially expressed in a mouse behavioural model system and uses thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase