WO2009047791A2 - Toll like receptor (tlr) signaling antagonist - Google Patents

Toll like receptor (tlr) signaling antagonist Download PDF

Info

Publication number
WO2009047791A2
WO2009047791A2 PCT/IN2008/000608 IN2008000608W WO2009047791A2 WO 2009047791 A2 WO2009047791 A2 WO 2009047791A2 IN 2008000608 W IN2008000608 W IN 2008000608W WO 2009047791 A2 WO2009047791 A2 WO 2009047791A2
Authority
WO
WIPO (PCT)
Prior art keywords
rscl
tlr
compound
disease
pharmaceutical composition
Prior art date
Application number
PCT/IN2008/000608
Other languages
French (fr)
Other versions
WO2009047791A3 (en
Inventor
Upadhyay Shakti
Kanekar Yogesh
Kshirsagar Rajendra
Rajagopal Vikram
Datla Praneel
Bellary Akshaya
Prasa Singh Shiva
Original Assignee
Reliance Life Sciences Pvt. Ltd.
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 Reliance Life Sciences Pvt. Ltd. filed Critical Reliance Life Sciences Pvt. Ltd.
Publication of WO2009047791A2 publication Critical patent/WO2009047791A2/en
Publication of WO2009047791A3 publication Critical patent/WO2009047791A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C43/00Ethers; Compounds having groups, groups or groups
    • C07C43/02Ethers
    • C07C43/20Ethers having an ether-oxygen atom bound to a carbon atom of a six-membered aromatic ring
    • C07C43/23Ethers having an ether-oxygen atom bound to a carbon atom of a six-membered aromatic ring containing hydroxy or O-metal groups
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/10Anti-acne agents
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C43/00Ethers; Compounds having groups, groups or groups
    • C07C43/02Ethers
    • C07C43/20Ethers having an ether-oxygen atom bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/02Esters of acyclic saturated monocarboxylic acids having the carboxyl group bound to an acyclic carbon atom or to hydrogen
    • C07C69/12Acetic acid esters
    • C07C69/14Acetic acid esters of monohydroxylic compounds
    • C07C69/145Acetic acid esters of monohydroxylic compounds of unsaturated alcohols
    • C07C69/157Acetic acid esters of monohydroxylic compounds of unsaturated alcohols containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/66Esters of carboxylic acids having esterified carboxylic groups bound to acyclic carbon atoms and having any of the groups OH, O—metal, —CHO, keto, ether, acyloxy, groups, groups, or in the acid moiety
    • C07C69/73Esters of carboxylic acids having esterified carboxylic groups bound to acyclic carbon atoms and having any of the groups OH, O—metal, —CHO, keto, ether, acyloxy, groups, groups, or in the acid moiety of unsaturated acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2603/00Systems containing at least three condensed rings
    • C07C2603/02Ortho- or ortho- and peri-condensed systems
    • C07C2603/04Ortho- or ortho- and peri-condensed systems containing three rings
    • C07C2603/22Ortho- or ortho- and peri-condensed systems containing three rings containing only six-membered rings
    • C07C2603/26Phenanthrenes; Hydrogenated phenanthrenes

Definitions

  • the present invention relates to novel molecules, compositions and methods for preparation and modulation of immune functions mediated through Toll-like receptor (TLR) signaling.
  • TLR Toll-like receptor
  • the innate or natural immune system recognizes a wide spectrum of pathogens without a need for prior exposure.
  • Cells of the innate immune system effectively prevent free growth of bacteria within the body; however, many pathogens have evolved mechanisms allowing them to evade the innate immune system, but unlike the adaptive immune system, it does not confer long-lasting or protective immunity to the host.
  • Innate immune systems provide immediate defense against infection, and are found in all classes of plant and animal life.
  • the main cells responsible for innate immunity are monocytes /macrophages and neutrophils, which phagocytose microbial pathogens and are responsible for triggering the innate, inflammatory, and specific immune responses.
  • Toll-like receptor are a family of receptors involved in the recognition of a wide range of microbial molecules e.g. Lipopolysaccharides (LPS) from Gram- negative bacteria and peptidoglycan from Gram- positive bacteria.
  • LPS Lipopolysaccharides
  • the prototype receptor Toll was first identified in the fruit fly Drosophila but several TLR was found in mammals, particularly on mononuclear phagocytes.
  • Toll-like receptors TLRs
  • TLR2 TLR 4
  • TLR5 TLR5 and each receptor recognizes a small range of structurally conserved molecules once they have breached physical barriers such as the skin or intestinal tract mucosa, and activate immune cell responses.
  • TLRs are a type of pattern recognition receptors (PRRs) and recognize molecules that are broadly shared by pathogens but distinguishable from host molecules, collectively referred to as pathogen-associated molecular patterns (PAMPs).
  • PRRs pattern recognition receptors
  • PAMPs pathogen-associated molecular patterns
  • TLRs seem only to be involved in the cytokine production and cellular activation in response to microbes, and do not play a significant role in the adhesion and phagocytosis of microorganisms. Binding of TLR leads to the production of inflammatory cytokines, including TNF-alpha and IL- 12 and enhances the cells' antimicrobial killing mechanisms and antigen presenting capacity. The function of the TLRs was discovered by Beutler and colleagues.
  • TLR2 and TLR4' The significance of the toll like receptors in the immune response to LPS has further demonstrated specifically two receptors TLR2 and TLR4' (Yang RB, Mark MR, Gray A, Huang A, Xie MH, Zhang M, Goddard A, Wood WI, Gurney AL, Godowski PJ. Toll-like receptor-2 mediates lipopolysaccharide-induced cellular signalling. Nature. 1998; 395: 284-88; Kirschning CJ, Wesche H, Merrill Ayres T, Rothe M. Human toll-like receptor 2 confers responsiveness to bacterial lipopolysaccharide. J Exp Med. 1998; 188: 2091-97.). Further reports concluded that TLR4 was required for a response to LPS.
  • LPS is an imm ⁇ nomodulatroty agent
  • its medicinal use is limited due to its extreme toxicity including the induction of systemic inflammatory response syndrome.
  • the biologically active endotoxio endotoxic sub-structural moeity of LPS is a lipid-A, a phosphorylated, multiple fatty acid acylated glucosamine disaccharide that serves to anchor the entire structure in the outer membrane of the gram-negative bacteria.
  • the toxic effects of the lipid A was addressed by selective chemical modification of the lipid A to produce monophosphoryl lipid A compounds (MPL ® : vaccine adjuvant and immunostimulant from Corixa (Seattle, Wash., US) and structurally like MPL ® compounds) which is described in U.S.
  • Johnson DA Keegan DS, Sowell CG, Livesay MT, Johnson CL, Taubner LM, Harris A, Myers KR, Thompson JD, Gustafson GL, Rhodes MJ, Ulrich JT, Ward JR, Yorgensen YM, Cantrell JL, Brookshire VG. 3-O- Desacyl monophosphoryl lipid A derivatives: synthesis and immunostimulant activities. J Med Chem. 1999; 42: 4640-49). US Pub. patent App.
  • 20070167409 is based on discovery that animals that do not express Toll-like receptor 2 (TLR2) are protected from dextran sulfate sodium (DSS) induction of colitis, a model for inflammatory bowel disease (IBD).
  • TLR2 Toll-like receptor 2
  • DSS dextran sulfate sodium
  • IBD inflammatory bowel disease
  • the European patent EP 1635846 has methods and compositions useful for modulating signaling through Toll-like receptors that involve contacting a TLR-expressing cell with a small molecule having a core structure including at least two rings. Certain of the compounds are 4-primary amino quinolines.
  • US Pub. patent App. US20060058365 relates to the treatment of inflammatory bowel disease (IBD) and related gastrointestinal pathologies that are cytokine-mediated ,or associated with Toll-like receptor 4 using methimazole derivatives and tautomeric cyclic thiones.
  • IBD inflammatory bowel disease
  • US Pub. patent App. 20050004144 provides a broad-spectrum, long- lasting, and non-toxic combination of synthetic immunostimulatory agents, which are useful for activating the immune system of a mammal and treating diseases such as cancer and autoimmune disease involving 7-substituted, 8-substituted and 7,8-di substituted 7-deazaguanosines.
  • US Pub. patent App. 20030139364 involves administration of an imidazoquinoline agent in combination with another therapeutic agent in synergistic amounts to enhance ADCC, stimulate immune responses and/or patient and treat certain disorders.
  • the PCT patent WO 2006027345 discloses novel 3-Thia-10-aza-phenanthrene derivatives as novel effective PDE4 inhibitors.
  • the PCT WO2006089881 has described novel phenanthrene derivatives as antiinflammatory agents and WO 9113855 has proivided a new phenanthrene derivative having IL-I inhibiting activity and useful for the treatment of chronic inflammatory diseases.
  • US patent 3683091 specifically provides di-7-hydroxy or methyl-2,3,4,4a,9,10- hexahydrophenanthren-2-one and 4a-alkyl derivatives, useful as specific anti-acne agents.
  • UK patent GB 1069067 provides novel phenanthrene derivatives having analgesic and morphine antagonistic activity.
  • US patent 476678 discloses phenathrene derivatives possessing valuable fungicidal properties useful in agriculture, horticulture and other antifungal compositions.
  • US patent 4808625 discloses aminoalkanol derivatives of containing a polycarbocyclic aromatic ring system such as phenanthrene, as biocidal agents, particularly antitumor agents.
  • Japanese patent JP 8067626 discloses hydrogenated condensed ring hydrocarbons such as hydrogenated phenanthrene (e.g. 1,2,3,4,5,6,7,8-octahydrophenanthrene), a hydrogenated anthracene (e.g. 1,2-dihydroanthracene), a hydrogenated naphthalene (e.g. 1,2- dihydronaphthalene), etc as capable of inhibiting the carcinogenesis induced by a carcinogenic organic compound without accompanied by side effects
  • hydrogenated phenanthrene e.g. 1,2,3,4,5,6,7,8-octahydrophenanthrene
  • anthracene e.g. 1,2-dihydroanthracene
  • naphthalene e.g. 1,2- dihydronaphthalene
  • UK patent GB2186570 provides 9, 10-dihydrophenanthrene derivatives which are useful in treating diseases characterized by an immunological imbalance and bacterial and viral infections in mammals.
  • Certain phenanthrene derivatives from plants have also been used in the prior art for immune system disorders such as 9, 10- dihydrophenanthrene called eulophiol isolated from the tubers of Eulophia nuda' (Bhandari SR and Kapadi AH.
  • eulophiol isolated from the tubers of Eulophia nuda
  • Tuchinda P Udchachon J
  • Khumtaveeporn K Taylor WC, Engelhardt LM
  • White A.H. Phenanthrenes of Eulophia nuda. Phytochemistry. 1988; 27: 3267-71
  • Majumder PL Sen S, Majumder S. Phenanthrene derivatives from the orchid Coelogyne cristata. Phytochemistry. 2001; 58: 581-586).
  • Japanese patent JP 7267895 has provided plant extracts containing phenanthrene derivatives from Raikoutou (a root or leaf of Tripterygium wilfordii Hook. F.) a Chinese herbal drug. These compounds have been found to be useful as a therapeutic agent for diseases owing to leukotriene of pollinosis, bronchial asthma, arthritis etc.
  • PCT application WO 2006089881 has focused on methoxy phenanthrene derivatives from Tamus communis as anti-inflammatory agents.
  • compositions of the phenanthrene derivatives and its analogs are provided.
  • compositions useful for the prevention or treatment of inflammation, wounds, autoimmunity, allergy, asthma, graft rejection, graft versus host disease, infection, sepsis, cancer and immunodeficiency are provided.
  • the inventors of the present invention have designed novel phenanthrene derivatives that act as antagonists of Toll-like receptors and methods and compositions for modulating the immune functions through Toll-like receptor.
  • the novel phenanthrene derivatives of the present invention have found their potential in inhibiting signaling of Toll-like receptors.
  • the present invention focuses on novel derivatives of phenanthrenes for potential use in inhibition of immune stimulation involving toll like receptor ligands. These molecules have been developed for potential use in treatment of inflammation, autoimmunity, allergy, asthma, graft rejection, graft versus host disease, infection, sepsis, cancer and immunodeficiency. More specifically, whereas the agents described herein have been discovered to affect TLRs directly and thus directly affect TLR-bearing cells, e.g., antigen-presenting cells (APCs), such agents can be used in conjunction with additional agents which affect non-APC immune cells, e.g., T lymphocytes (T cells). Such an approach effectively introduces an immunomodulatory intervention at two levels: innate immunity and acquired immunity.
  • APCs antigen-presenting cells
  • the present invention relates to phenanthrene derivatives, methods of their preparation, and compositions for use in TLR mediated immune conditions.
  • the present invention also relates to compositions and methods for modulating immune functions mediated through Toll-like receptor (TLR) molecules.
  • TLR Toll-like receptor
  • the present invention provides compositions that are useful for the prevention or treatment of inflammation, wounds, autoimmunity, allergy, asthma, graft rejection, graft versus host disease, infection, sepsis, cancer, and immunodeficiency.
  • the compositions as described in the present invention or the compositions are useful as for inhibition of TLR signaling in response to TLR ligands.
  • compositions for inhibition of TLR signaling as described in the present invention in a therapeutically effective amount and pharmaceutically inert adjuvants, diluents or carriers.
  • compositions as described in the present invention or composition and the method of manufacture comprising the same is believed to have the ability to inhibit TLR signaling under physiological conditions, and thereby would have corresponding effectiveness for prevention or treatment of inflammation, wounds, autoimmunity, allergy, asthma, graft rejection, graft versus host disease, infection, sepsis, cancer, and immunodeficiency.
  • the compositions as described in the present invention can be used to prevent or treat clinical manifestations and diseases caused by microbial pathogens.
  • the composition can be used in veterinary medicine to prevent or treat clinical manifestations and diseases caused by microbial pathogens.
  • the present invention also provides pharmaceutical formulations either by itself or in a suitable pharmaceutically acceptable adjuvant useful for inhibition of TLR mediated clinical manifestations.
  • the methods of the invention can be combined with administration of additional agents to achieve synergistic effect on TLR-mediated immunostimulation.
  • agents described herein have been discovered to affect TLRs directly and thus directly affect TLR-bearing cells, e.g., antigen-presenting cells (APCs)
  • such agents can be used in conjunction with additional agents which affect non-APC immune cells, e.g., T lymphocytes (T cells).
  • T cells T lymphocytes
  • a method of affecting TLR-mediated signaling in response to a TLR ligand is provided.
  • a method of inhibiting TLR-mediated immunostimulatory signaling is provided.
  • the invention provides a method of affecting TLR-mediated immunostimulation in a subject.
  • Methods of treatment for variety of conditions involving autoimmunity, inflammation, allergy, asthma, graft rejection, graft-versus-host disease (GvHD), infection, sepsis, cancer, and immunodeficiency will employ small molecules that augment TLR-mediated signaling in response to a suitable TLR ligand.
  • the methods can be used to inhibit or promote TLR-mediated signaling in response to a TLR ligand or TLR signaling agonist.
  • the methods can be used to inhibit TLR-mediated immunostimulatory signaling in response to a TLR ligand or TLR signaling agonist.
  • the methods can be used to inhibit or promote TLR- mediated immunostimulation in a subject. In some instances the methods can be used to inhibit TLR-mediated immunostimulation in a subject. In some instances the methods can be used to inhibit an immunostimulatory nucleic acid-associated response in a subject.
  • the present invention provides molecules and methods useful for modulating TLR-mediated signaling.
  • the molecules of the present invention are applicable to alter any TLR-mediated signaling in response to a suitable ligand or signaling agonist.
  • the present invention also provides methods for identifying agents that decrease or inhibit activation of Toll-like receptor 2. These methods involve (i) contacting a cell expressing the receptor with a candidate agent in the presence of an activator of the receptor ( in vitro or in vivo) and (ii) determining the effect of the agent on activation of the receptor. Detection of a decrease in activation of the receptor by the activator in the presence of the agent indicates the identification of agent that can be used to decrease or inhibit activation of the receptor.
  • the effect of the agent on the activation of the receptor can be determined by analysis of the expression of a reporter gene that is under the control of a promoter that is induced in a signaling pathway triggered by activation of the receptor.
  • the present invention provides compounds which can be isolated from plant species such as Eulophia.
  • this base compound Eulophiol RSCL-0520
  • further derivatives can be prepared by synthetic routes.
  • the present invention also provides compounds, which can be prepared by synthetic routes.
  • a method of affecting TLR-mediated signaling in response to a TLR ligand involves contacting a cell expressing a TLR with an effective amount of a compound of Formula I
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 and R 10 are identical or different and may each be hydrogen, hydroxy, C 1-6 alkoxy, C 1-6 alkyl, halogen, haloalkyl, acyloxy, hydroxyalkyl, alkenyl, alkenyloxy, carboxyl, carbalkoxy, carbamido, a conjugated group, substituted or unsubstituted phenyl, substituted or unsubstituted heterocyclic group, nitro, amino, acylamino, dialkylamino, nitric oxide (NO) - releasing moiety, pharmaceutically acceptable salts, amides and esters thereof.
  • Ring A, Ring B, and Ring C may be aliphatic or aromatic.
  • FIG. Ia shows that RSCL-0520 inhibits TLR4 induced TNF- ⁇ secretion in human monocytic (THP-I) cells.
  • FIG. Ib shows that RSCL-0520 inhibits TLR2 and TLR4 induced TNF- ⁇ secretion in and peripheral blood monocytes (PBMCs).
  • PBMCs peripheral blood monocytes
  • FIG. 2a shows inhibition of TNF- ⁇ secretion in THP-I cells by RSCL-0520 is dose-dependent
  • FIG. 2b indicates that RSCL-0520 is not toxic to THP-I cells
  • FIG. 3 shows RSCL-0520 inhibits LPS induced TNF- ⁇ release in PBMC
  • FIG. 4 shows the dose dependent effect of RSCL-0520 on different concentrations of LPS in THP-I cells
  • FIG. 5a shows effect of RSCL-0520 on TNF- ⁇ mRNA expression in THP-I cells in real time
  • FIG. 5b shows effect of RSCL-0520 on mRNA expression of pro-inflammatory genes in THP-I Cells
  • FIG. 5c Demonstrates ability of RSCL-0520 to inhibit Arachidonic acid induced PGE2 release in A549 cells
  • FIG. 6 shows RSCL-0520 suppresses LPS induced Nitric oxide (NO) release in RAW 264.7 cells
  • FIG. 7a represents effect of RSCL-0520 on activation of NEMO and degradation of I kappa B-alpha (I ⁇ B- ⁇ ).
  • FIG. 7b represents effect of RSCL-0520 activation of NF- ⁇ B.
  • FIG. 7c represents effect of RSCL-0520 on translocation of NF- ⁇ B to the nucleus
  • FIG. 8a represents effect of RSCL-0520 on TLR related genes
  • FIG. 8b represents RSCL-0520 inhibits MyD88 dependent TLR signaling by LPS.
  • FIG. 9a shows pre-treatment of RSCL-0520 suppresses LPS induced TNF- ⁇ release in Balb/c mice
  • FIG. 9b Treatment with RSCL-0520 post LPS induction suppresses the induced TNF- ⁇ release in Balb/c mice
  • LPS Lipopolysaccharide
  • TLR Toll like receptor
  • salts refers to those salts which are, within the scope of sound medical judgement, suitable for use in contact with the tissues of humans and animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al. describes pharmaceutically acceptable salts in detail. (Berge SM, Bighley LD, Monkhouse DC. Pharmaceutical salts. J Pharm Sci. 1977; 66:1-19).
  • the salts can be prepared in situ during the final isolation and purification of a compound of the invention or separately by reacting the free base group with a suitable organic acid.
  • Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphersulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pe
  • alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like.
  • pharmaceutically acceptable ester represents esters that hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof.
  • Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic, and alkanedioic acids, in which each alkyl or alkenyl group preferably has not more than 6 carbon atoms.
  • Examples of particular esters include formates, acetates, propionates, butyates, acrylates, and ethylsuccinates.
  • Amides and esters could also be prepared by coupling the compounds of the present invention with phenocli acids such as Non-steroidal anti-inflammatory drugs (NSAIDs) etc.
  • NSAIDs Non-steroidal anti-inflammatory drugs
  • pro-drugs means pro-drugs of the compounds of the present invention which are, within the scope of sound medical judgement, suitable for use in contact with the tissues of humans and animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention.
  • prodrug represents compounds that are transformed in vivo into a parent compound of the above formula, for example, by hydrolysis in blood.
  • prodrugs as used herein, represents compounds that are transformed in vivo into a parent compound of the above formula, for example, by hydrolysis in blood.
  • a thorough discussion of prodrugs is provided in T. Higuchi and V. Stella (Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, Edward B. Roche, ed., “Bioreversible Carriers in Drug Design,” American Pharmaceutical Association and Pergamon Press, 1987), and Judkins et al” (Synthetic Communications. 1996; 26: 4351- 67), each of which is incorporated herein by reference.
  • Asymmetric or chiral centers may exist in the compounds of the present invention.
  • the present invention includes the various stereoisomers and mixtures thereof.
  • Individual stereoisomers of compounds or the present invention may be prepared synthetically from commercially available starting materials that contain asymmetric or chiral centers or by preparation of mixtures of enantiomeric compounds followed by resolution well-known to those of ordinary skill in the art. These methods of resolution are exemplified by (1) attachment of a racemic mixture of enantiomers, designated (+/-), to a chiral auxiliary, separation of the resulting diastereomers by recrystallization or chromatography and liberation of the optically pure product from the auxiliary or (2) direct separation of the mixture of optical enantiomers on chiral chromatographic columns.
  • Enantiomers are designated herein by the symbols "R” or “S,” depending on the configuration of substituents around the chiral carbon atom, or are drawn by conventional means with a bolded line defining a substituent above the plane of the page in three-dimensional space and a hashed or dashed line defining a substituent beneath the plane of the printed page in three-dimensional space. If no stereochemical designation is made, it is to be assumed that the structure definition includes both stereochemical possibilities.
  • the present invention relates to the compounds of formula (I) and derivatives thereof including but not limited to polymorphs, isomers and prodrugs thereof, geometric or optical isomers thereof, and pharmaceutically acceptable esters, ethers, carbamates of such compounds, all solvates and hydrates thereof and all salts thereof.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 and R 10 are identical or different and may each be hydrogen, hydroxy, C 1-6 alkoxy, C 1-6 alkyl, halogen, haloalkyl, acyloxy, hydroxyalkyl, alkenyl, alkenyloxy, carboxyl, carbalkoxy, carbamido, a conjugated group, substituted or unsubstituted phenyl, substituted or unsubstituted heterocyclic group, nitro, amino, acylamino, dialkylamino, nitric oxide (NO) — releasing moiety, pharmaceutically acceptable salts, amides and esters thereof.
  • Ring A, Ring B, and Ring C may be aliphatic or aromatic.
  • the compounds of the present invention may contain asymmetric or chiral centers, and therefore may exist in different stereoisomeric forms. All suitable optical isomers and stereoisomeric forms of the compounds of the present invention as well as mixtures thereof, including racemic mixtures, form part of the present invention. In addition, the present invention embraces all geometric and positional isomers. Moreover, some compounds of the present invention may exhibit polymorphism. The present invention includes all polymorphic forms of the compounds according to the invention, which forms the further aspect of the invention. It is to be understood that the present invention encompasses any and all racemic, optically-active, polymorphic and stereoisomeric forms, or mixtures thereof, which form or forms possess properties useful in the treatment of the conditions indicated herein.
  • the present invention also include isotopically-labeled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • reaction products are isolated and purified by conventional methods, typically by solvent extraction into a compatible solvent.
  • the products may be further purified by column chromatography or other appropriate methods, including medium pressure or high pressure liquid chromatography.
  • a general method of synthesis includes the preparation of phenanthrene molecules by any of the following approaches:
  • a mixture of substituted benzaldehyde, 3, 4, 5-trimethoxyphenyl acetic acid, acetic anhydride and triethylamine is refluxed and then acidified to obtain acid (A) which is crystallized from suitable solvent.
  • the acid (A) obtained is mixed with quinoline and copper chromite and refluxed under inert atmosphere and then precipitated with ethyl acetate. After filtration, the filtrate is washed with dilute hydrochloric acid, water, and brine, dried (sodium sulphate), and evaporated under reduced pressure to isolate the product.
  • B) A stirred solution of the product obtained (B) and iodine in ethanol is irradiated at 254 run. The solution is then evaporated under reduced pressure and the product (C) is isolated by silica gel column chromatography. (Pet-ether-ethyl acetate gradient)
  • the cooled filtrate is acidified with glacial acetic acid and the precipitate is filtered off and vacuum dried (B).
  • B a mixture of amine (B), ethanol and 5N-hydrochloric acid at O 0 C is added a 50% ethanolic solution of isopentyl nitrite. The mixture stirred and then diluted with water, further copper powder is added and stirred at 50 0 C.
  • the cooled mixture is then extracted with ethyl acetate, which on concentration under reduced pressure gives acid (C).
  • a mixture of acid (C) quinoline and basic copper carbonate is refluxed, and extracted with ethyl acetate. The ethyl acetate layer is washed with acid, alkali, and evaporated.
  • the product (P) is purified by column chromatography on silica (with pet-ether-ethyl acetate gradient).
  • the present invention provides agents that can be used to prevent or to treat LPS mediated diseases or conditions that are characterized by TLR activation.
  • IBD inflammatory bowel disease
  • sepsis sepsis
  • periodontal disease mucositis
  • mucositis mucositis
  • acne cardiovascular disease
  • chronic obstructive pulmonary disease arthritis
  • cystic fibrosis bacterial-induced infections
  • viral-induced infections mycoplasma-associated diseases
  • post herpetic neuralgia ischemia/reperfusion injury
  • asthma stroke
  • stroke brain injury
  • bed sores leprosy
  • atopic dermatitis psoriasis
  • trauma neurodegenerative disease
  • amphotericin B-induced fever and nephritis coronary artery bypass grafting
  • atherosclerosis atherosclerosis
  • compositions comprising carbohydrate based molecules in an effective amount that achieves the desired therapeutic effect for a particular condition, patient and mode of administration.
  • the dosage level selected depends on the route of administration and the severity of the condition being treated.
  • the doses are generally from about 0.01 to about 100mg/kg, desirably 0.1-lmg/kg by inhalation, desirably 0.5-10mg/kg per day by oral administration, and desirably 0.1-lmg/kg body weight per day by intravenous administration.
  • Doses are determined for each particular case using standard methods in accordance with factors unique to the patient, including age, weight, general state of health, and other factors that can influence the efficacy of the compound(s) of the invention.
  • administration of the compounds of the present invention is not limited to mammal, including humans, be limited to a particular mode of administration, dosage, or frequency of dosing.
  • the present invention encompasses all modes of administration, including oral, intraperitoneal, intramuscular, intravenous, intra-articular, intralesional, subcutaneous, or nasally, rectally, buccally, or any other route sufficient to provide a dose adequate to prevent or treat excess or undesired TLR activity.
  • the present invention also contemplates that one or more compounds may be administered to a mammal in a single dose or multiple doses. When multiple doses are administered, the doses may be separated from one another by, for example, several hours, one day, one week or one month. It is to be understood that, for any particular subject, specific dosage regimes should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of a pharmaceutical composition that includes a compound of the invention.
  • compositions of phenanthrene derivatives being TLR antagonists which may be prepared by conventional methods using one or more pharmaceutically acceptable excipients or adjuvants which may comprise inert diluents, sterile aqueous media and/or various non toxic solvents.
  • pharmaceutically acceptable carrier or diluents may be used as described in literature' (The Science and Practice of Pharmacy (20th ed.), ed. A. R. Gennaro, Lippincott Williams & Wilkins, 2000, Philadelphia; Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988 1999, Marcel Dekker, New York).
  • compositions may be presented in the form of tablets, pills, granules, powders, aqueous solutions or suspensions, injectable solutions, elixirs, or syrups, and the compositions may optionally contain one or more agents chosen from the group comprising sweeteners, flavorings, colorings, and stabilizers in order to obtain pharmaceutically acceptable preparations.
  • excipients such as lactose, sodium citrate, calcium carbonate, and dicalcium phosphate and disintegrating agents such as starch, alginic acids, and certain complex silicates combined with lubricants (e.g., magnesium stearate, sodium lauryl sulfate, and talc) may be used for preparing tablets.
  • lubricants e.g., magnesium stearate, sodium lauryl sulfate, and talc
  • lactose and high molecular weight polyethylene glycols When aqueous suspensions are used, they may contain emulsifying agents that facilitate suspension.
  • Diluents such as sucrose, ethanol, polyethylene glycol, propylene glycol, glycerol, chloroform, or mixtures thereof may also be used.
  • emulsions, suspensions, or solutions of the compositions of the invention in vegetable oil e.g., sesame oil, groundnut oil, or olive oil
  • aqueous- organic solutions e.g., water and propylene glycol
  • injectable organic esters e.g., ethyl oleate
  • sterile aqueous solutions of the pharmaceutically acceptable salts are used.
  • the solutions of the salts of the compositions of the invention are especially useful for administration by intramuscular or subcutaneous injection.
  • Aqueous solutions that include solutions of the salts in pure distilled water may be used for intravenous administration with the proviso that (i) their pH is adjusted suitably, (ii) they are appropriately buffered and rendered isotonic with a sufficient quantity of glucose or sodium chloride, and (iii) they are sterilized by heating, irradiation, or microfiltration.
  • Suitable compositions containing a compound of the invention may be dissolved or suspended in a suitable carrier for use in a nebulizer or a suspension or solution aerosol, or may be absorbed or adsorbed onto a suitable solid carrier for use in a dry powder inhaler.
  • Solid compositions for rectal administration include suppositories formulated in accordance with known methods and containing at least one compound of formula I or II.
  • Dosage formulations of a compound of the invention to be used for therapeutic administration must be sterile. Sterility is readily accomplished by filtration through sterile membranes (e.g., 0.2 micron membranes) or by other conventional methods. Formulations typically are stored in lyophilized form or as an aqueous solution.
  • the pH of the compositions of this invention is typically between 3 and 11, more desirably between 5 and 9, and most desirably between 7 and 8, inclusive. While a desirable route of administration is by injection such as intravenously (bolus and/or infusion), other methods of administration may be used.
  • compositions may be administered subcutaneously, intramuscularly, colonically, rectally, nasally, or intraperitoneally in a variety of dosage forms such as suppositories, implanted pellets or small cylinders, aerosols, oral dosage formulations, and topical formulations such as ointments, drops, and dermal patches.
  • dosage forms such as suppositories, implanted pellets or small cylinders, aerosols, oral dosage formulations, and topical formulations such as ointments, drops, and dermal patches.
  • a compound of the invention is desirably incorporated into shaped articles such as implants, including but not limited to valves, stunts, tubing, and prostheses, which may employ inert materials such as synthetic polymers or silicones, (e.g., Silastic, silicone rubber, or other commercially available polymers).
  • Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxy-propyl- methacrylamide-phenol, polyhydroxyethyl-aspartamide-phenol, or polyethyleneoxide- polylysine substituted with palmitoyl residues.
  • a TLR2 inhibitor of the invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and cross linked or amphipathic block copolymers of hydrogels.
  • biodegradable polymers useful in achieving controlled release of a drug
  • a drug for example polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and cross linked or amphipathic block copolymers of hydrogels.
  • a compound of the invention may also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles.
  • Liposomes can be formed from a variety of lipids, such as cholesterol, stearylamine, or phosphatidylcholines.
  • a compound of the invention may also be delivered using antibodies, antibody fragments, growth factors, hormones, or other targeting moieties to which the compound molecules are coupled (e.g., see Remington: The Science and Practice of Pharmacy, vide supra), including in vivo conjugation to blood components of a compound of the formula I or II, as described herein.
  • compositions that can be used in the therapeutic methods of the invention can be identified in screening methods.
  • cell-based screening methods can be used, in which cells expressing TLR are contacted with a candidate agent and the impact of the agent on the activation of TLR in the cells is determined.
  • the effect of an agent on the activation of TLR by a known ligand e.g., a lipopeptide,
  • Agents that are found to decrease or to block activation of the receptor by the ligand can then be considered for further analysis and/or for use as TLR inhibitors in therapeutic methods.
  • Activation of TLR in these methods can be measured using, for example, a reporter system.
  • cells used in the screening assay can include a reporter gene that is under the control of a promoter that is inducible by a signaling pathway triggered by TLR activation.
  • candidate agents can be tested in animal model systems. This may be desirable, for example, if an agent has been found to have antagonist activity in a cell-based assay or to bind to TLR in an in vitro assay (see below).
  • test agents can be administered to an animal model concurrently with a molecule known to activate TLR (e.g., lipopeptide), and the impact of the agent on a response in the animal that is normally triggered by activation of the receptor (e.g., cytokine induction) can be determined.
  • in vitro methods can be used.
  • a candidate compound can be assayed for whether it binds to TLR or a fragment of the receptor that includes at least a portion of the ligand binding site.
  • assays can be carried out using, for example, columns or beads to which the receptor or fragment is bound.
  • TLR antagonists can be identified in methods in which candidate compounds are compared for TLR antagonist activity with any of the TLR antagonists described herein. Further, in addition to being compared for TLR antagonist activity, the candidate compounds can be compared with TLR2 antagonists with respect to specificity for TLR versus other receptors.
  • TLR2 antagonists with respect to specificity for TLR versus other receptors.
  • Candidate compounds identified as having TLR antagonist activity that is, for example, similar to or greater than the activity of the antagonists described herein (and/or with similar or greater levels of specificity for TLR2 versus TLR4) in these assays can be tested further, for example, in appropriate animal model assays for any of the diseases or conditions described herein, as well as in human clinical studies.
  • a compound that is selective for TLR2 over TLR4 is one that has, for example, an IC 50 value in a TLR2 antagonist assay, such as is described herein, that is less than that found in a TLR4 antagonist assay.
  • the IC 5O in the TLR2 assay can be at least 5, 10, 25, or 50-fold less than the value for the same compound tested in the TLR4 assay.
  • Compounds that are dual antagonists are those that have, for example, IC 50 values that are within a 5-fold range of one another using.
  • dual antagonists include those that have activities that are 1 :5 5:1 with respect to one another (e.g., 1:4, 1 :3, 1:2, 1:1, 2:1, 3:1, and 4:1).
  • the invention also includes the use of TLR2 antagonists such as those described herein in the study of physiological and molecular pathways involved in or affected by TLR2 activation (or inactivation).
  • Agents that can be screened using the methods of the invention include, for example, compounds that are present in compound libraries (e.g., random libraries), as well as analogs of known TLR2 ligands (e.g., lipopeptides) that are modified to prevent rather than activate TLR2. Further, peptides that correspond to the binding site of TLR2 or its ligands, which can competitively inhibit ligand binding, can be tested. Further, antibodies or antibody fragments to the ligand or the ligand binding site of the receptor can be screened. The following examples are included to demonstrate embodiments of the invention.
  • EXAMPLE - 1 Isolation of l,5-dihydoxy-2, 7-dimethoxy-9,10-dihydrophenanthrene (Eulophiol) (RSCL-520) and 2,7-dihydroxy-3,4-dimethoxyphenanthrene (Nudol) (RSCL-518)
  • Tubers of Eulophia ochreata were collected in the month of October 2006.
  • Herbarium voucher specimen (Accession No. 157) was authenticated and deposited at herbarium of Dhirubhai Ambani Life Sciences Center, Navi Mumbai, Maharashtra.
  • Residue obtained after removal of solvents were tested for free radical scavenger activity using curcumin as reference standard and DPPH (2, 2-Diphenyl-l-picryl-hydrazyl) as radical.
  • Test solution preparation DPPH was dissolved in methanol (AR grade) to a concentration of 1OmM.
  • Sample preparation Weighed about one mg of extract residue/curcumin and dissolved in lOO ⁇ l of DMSO and added methanol in order to make concentration lmg/ml. Pipetted out 50 ⁇ l sample solution in a 96-well micro titre plate to which added 200 ⁇ l of above prepared DPPH test solution. The plate was incubated in dark for half an hour. Absorbance measured at 540nm on Eliza plate reader. The corresponding blank readings were also taken for calculating the percentage antioxidant activity. Antioxidant activity in percentage was calculated by the formula: (1 -(Absorbance of sample / Absorbance of DPPH) X lOO.
  • Table 1 Free radical scavenging activity of different extracts in DPPH assay.
  • nudol (II) (0.02Og, 0.07 mmol) in dichloromethane (20ml) was added acetic anhydride (0.020ml), pyridine (0.020ml) and stirred overnight at R.T. (20-25 0 C). To this was then added water (50ml) and the organic layer washed with dilute hydrochloric acid (10ml), water (10ml), dried over sodium sulfate and concentrated under reduced pressure, which gave pure compound (IV) (RSCL-0519) (0.108g) in 82.44% yield (w/w). M.P. 151 0 C, (151°C; Bhandari et al.
  • reaction was quenched by the addition of IN hydrochloric acid (pH changed to 4-5), diluted with brine (20ml), extracted with ethyl acetate (3 X 20ml), dried over sodium sulfate, and the product, an oil, was isolated by silica gel column chromatography (pet- ether-ethyl acetate gradient).
  • RSCL-0520 (Tab. 1) showed the best TNF inhibiting activity, which we took forward for further in-vitro studies.
  • the present invention has checked the effect of various TLR ligands on THP-I monocytes, PBMCs and their ability to activate and release TNF- ⁇ . For this purpose about 9 TLR ligands were used. These ligands were obtained from Apotech Cat; APO- 54N-018-KI01 and assayed for TNF- ⁇ release in culture supernatants. THP-I (Fig. Ia) and PBMCs (2 x 10 5 cells/well) (Fig. Ib) were plated in 96-well plate. The cells were pretreated with RSCL-0520 in DMSO lhr prior to TLR ligand treatment.
  • TLR2 TLR2, TLR5 and TLR6 -75ng/ml each, TLR3-75 ⁇ g/ml, TLR4-750ng/ml, TLR7, TLR8 and TLR9-7.5 ⁇ g/ml
  • the culture supernatant were collected after the stipulated time and assayed for TNF- ⁇ release using a Duoset Enzyme-Linked Immunosorbent Assay (ELISA) detection Kit (R&D systems, MN 55413, USA; Cat: DY-210 E).
  • ELISA Duoset Enzyme-Linked Immunosorbent Assay
  • the present invention has studied the ability of RSCL-0520 to inhibit TNF- ⁇ secretion from LPS (250ng/ml) induced THP-I monocytes (Fig. 2a).
  • THP-I 2 x 10 5 cells/well was plated in 96-well plate.
  • the cells were pretreated with RSCL-0520 at various concentrations (lOO ⁇ M, 50 ⁇ M, lO ⁇ M and l ⁇ M) lhr prior to LPS stimulation.
  • As a control group cells were treated with LPS alone and cells treated with RSCL-0520 alone were used.
  • TNF- ⁇ secretion was estimated in the culture supernatants following 24hr LPS stimulation using Duoset ELISA detection Kit (R&D systems, MN, USA). ***P ⁇ 0.001 values are comparisons for LPS treated vs. RSCL-0520 treated, NS indicates not significant.
  • RSCL-0520 The toxicity of RSCL-0520 (Fig. 2b) was also by treating cells with RSCL-0520 by MTT assay. LPS induced TNF- ⁇ secretion was inhibited by RSCL-0520 in a concentration dependent manner. The viability of the cells (analyzed in tandem by MTT) was not affected by RSCL-0520 indicating its non-toxic nature.
  • RSCL-0520 To evaluate the ability of RSCL-0520 to inhibit LPS induced TNF- ⁇ secretion in a physiological scenario, the present invention has tested the same in PBMC isolated from human blood (Fig. 3) by standard methodology. The inhibition was similar to that observed in THP-I cells. The TNF levels were not detectable in PBMCs without LPS and with RSCL-0520 treatment alone, indicating its specificity in LPS induced TNF- ⁇ through a TLR mediated process. ***P ⁇ 0.001 value is for LPS treated vs. RSCL-0520 treated.
  • the present invention has also conducted experiments to check the ability of RSCL-0520 to inhibit LPS induced TNF- ⁇ secretion from THP-I cells, wherein the studies involved the stimulation of THP-I cells with increasing concentrations of LPS (31.25ng/ml to lOOOng/ml) with and without pre-treatment of cells with different concentrations of RSCL-0520. It is clearly evident (Fig. 4) that with an increased secretion of TNF- ⁇ with increasing concentration of LPS is inhibited by RSCL-0520 to varied extents on a concentration dependent manner. Further, even RSCL-0520(10 ⁇ M) was effective to inhibit LPS (1000ng/ml) induced TNF- ⁇ secretion; clearly indicating it's potency as an antagonist to LPS induced processes.
  • THP-I cells 3 x 10 6 cells/well
  • RSCL-0520 50 ⁇ M
  • Total RNA was isolated from these cells and cDNA was synthesized. LPS treated cells acted as positive control.
  • All quantitative real-time PCR (TaqManTM) primers and probes were obtained from Applied Biosystems(ABI), Rothstadt, Germany.
  • PCR was performed utilizing 1 ⁇ l cDNA per reaction in triplicates of 25 ⁇ l volume on an ABI 7500 Realtime PCR machine using a 2- step PCR protocol.
  • RSCL-0520 on mRNA was checked on pro-inflammatory genes like intercellular adhesion molecule 1 (ICAM-I), cyclooxygenase 2 (COX-2), IL-I ⁇ and IL-8.
  • IAM-I intercellular adhesion molecule 1
  • COX-2 cyclooxygenase 2
  • IL-I ⁇ IL-8.
  • THP-I cells (3 xlO 6 cells) were seeded in a 6-well dish were treated with RSCL-0520 (50 ⁇ M) for lhr followed by incubation with or without LPS (250ng/ml). Following two washes with ice-cold PBS, the cells were harvested and total cellular RNA was isolated using TRIZOL Reagent (Invitrogen) according to the manufacturer's instructions.
  • cDNA was synthesised using high capacity cDNA reverse transcription kit (ABI systems). Amplification of ICAM-I, COX-2, IL- l ⁇ and IL-8 genes from the cDNA was carried out using the respective gene specific primers.
  • RSCL-0520 inhibited mRNA expression levels of the tested genes (Fig. 5b), indicating that its mechanism of action is via NF- ⁇ B. However, cells treated with RSCL-0520 in the absence of LPS, did not alter the gene expressions of the genes in consideration highlighting its specificity.
  • COX-2 is the key enzyme regulating the production of prostaglandins, which act as central mediators of inflammation.
  • Our earlier in- vitro data clearly demonstrated that the present invention inhibits expression of COX-2.
  • COX-2 pathway inhibitors were regarded as promising nonsteroidal anti-inflammatory drugs (NSAIDs). So we decided to test the ability of RSCL-0520 to block the COX-2 pathway to substantiate our earlier mRNA observation. For that purpose we chose A549 cells, a human lung cancer cell line where COX-2 is activated by AA in serum-free stimulation established by Yao et al for 12hr.
  • A549 cells 50 x 10 4 cells/well) in serum free medium were pretreated with different concentrations (lO ⁇ M, 5 ⁇ M, 2.5 ⁇ M, and 1.25 ⁇ M) of the NSAIDs and RSCL-0520 for 30min.
  • concentrations lO ⁇ M, 5 ⁇ M, 2.5 ⁇ M, and 1.25 ⁇ M
  • the cells were incubated with AA (lO ⁇ M) for another 30 min.
  • Prostaglandin E2 (PGE2), a metabolite of AA through the Cox pathway, was assayed in an enzyme immunoassay (EIA) kit from R&D systems.
  • EIA enzyme immunoassay
  • RSCL-0520 shows a concentration dependent inhibition of PGE2 release.
  • RSCL-0520 is effective even at 1.25 ⁇ M enhancing its potential as potent anti-inflammatory molecule.
  • RAW 264.7 cells LPS induces iNOS transcription and transduction, and then the NO production. Furthermore, LPS stimulation is well known to induce IKB proteolysis and NF- ⁇ B nuclear translocation. (Freeman & Natanson. Antiinflammatory therapies and sepsis and septic shock. Expert Opin. Invest. Drugs. 2000; 9: 1651-63). Therefore, RAW264.7 cells provide an excellent model for evaluations of potential inhibitors on the pathway leading the induction of iNOS and NO production. Nitric oxide production was determined in RAW 264.7 cells from the American Type Culture Collection (Manassas, VA) cultured in color-free DMEM with standard supplements by measuring the amount of nitrite from cell culture supernatant.
  • Manassas, VA American Type Culture Collection
  • RAW264.7 cells (5 x 10 4 per well) were stimulated for 24hr with or without LPS (250ng/ml) in the absence of presence of the RSCL-0520.
  • Nitrite a stable end product of NO, was then measured using the Griess reaction (Green LC, Wagner D, Glogowski J, Skipper PL, Wishnok JS, Tannenbaum SR. Analysis of nitrate, nitrite, and [15-N] nitrate in biological fluids. Anal Biochem. 1982; 126: 131—38.).
  • lOO ⁇ l of cell culture supernatant was reacted with lOO ⁇ l of Griess reagent followed by spectrophotometric measurement at 550nm.
  • Nitrite concentrations in the supernatants were determined by comparison with a sodium nitrite standard curve.
  • Fig. 6 show that NO secretion induced by LPS stimulation was inhibited by RSCL-0520 in a concentration dependent manner.
  • RSCL-0520 exhibits better anti-oxidant potency than any of them.
  • the reactive free radical NO synthesized by iNOS is a major macrophage derived inflammatory mediator and also has been reported to be involved in the development of inflammatory diseases. (Xie QW, Cho H, Kashiwabara Y, Baum M, Weidner JR, Elliston K, Mumford R, Nathan C.
  • NF- ⁇ B/I ⁇ B complexes are present in the cytoplasm under unstimulated conditions. Following stimulation with LPS, we see phosphorylation and subsequent degradation of IKB allowing the free NF- ⁇ B to translocate into the nucleus to activate genes with NF- ⁇ B binding regions. Therefore, the effect of RSCL-0520 on blocking of NF- ⁇ B nuclear translocation was checked. Serum- starved THP-I cells were stimulated with LPS (250ng/ml) for the indicated time (Fig. 7a) in the presence and absence of RSCL-0520 (50 ⁇ M). RSCL-0520 treatment was lhr prior to LPS treatment.
  • Total protein was isolated from the treated cells, and an equal amount of protein from each sample was used for immunoblots to determine protein levels of NEMO and I ⁇ B- ⁇ . Blots were stripped and reprobed using ERK- 1/2 antibody to normalize the protein loading.
  • RSCL-0520 blocked signaling to NEMO, possibly blocking phosphorylation of IKK. Which resulted in blocking p65 dissociation from I ⁇ B- ⁇ (Fig. 7a lanes 6-8). This accumulation of I ⁇ B- ⁇ lead to inhibition of subsequent down stream signaling pathways.
  • lhr LPS- stimulated cells with and without pretreatment of RSCL-0520 (50 ⁇ M) Further processing was done as per FACS staining procedure. The cells were then stained with phospho p65 monoclonal antibody tagged with Alexa Fluor 488 (Cell Signaling Technology, Inc, Ma, USA) for lhr at 37°C, followed by washing. Cells were then resuspended in PBS and stained cells were acquired in BD FACS Calibur. Results (Fig. 7b) clearly show its ability to inhibit phospho-p65 in LPS treated cells, substantiating its role in NF- ⁇ B signaling inhibition.
  • the nuclear fractions were obtained from LPS stimulated THP-I cells at the indicated times to evaluate the role of RSCL-0520 in inhibiting p65 subunit translocation into the nucleus.
  • the nuclear proteins electrophoresed were processed for immunoblots using a NF- ⁇ B specific antibody. Immunoblotting profile of nuclear extracts for p65 in a time dependent manner (Fig. 6c) clearly shows that RSCL-0520 blocked translocation of NF- KB into the nucleus.
  • Serum - starved THP-I cells were stimulated with LPS (250ng/ml) for lhr in the presence and absence of RSCL-0520.
  • Total RNA was isolated from treated cells post LPS exposure.
  • the cDNA was used for PCR against specific primers for the TLR related genes and ⁇ -actin was used as internal control.
  • TLR4 TLR4 signaling
  • MyD88 myeloid differentiation primary response protein 88
  • TIRAP Toll receptor IL-IR domain-containing adapter protein
  • TIR domain-containing adapter inducing IFN ⁇ TIR
  • TRM Trif-related adapter molecule
  • TLR related genes are upregulated upon stimulation with LPS (Fig. 8a lane 3); while pretreatment with RSCL-0520(Fig. 8a lane 4) inhibits the mRNA expression levels of TIRAP, MyD88, TRAF6, IL- lR-associated kinase 1 (IRAKI) and IRAK4.
  • RSCL-0520 inhibits MyD88 dependent signaling of TLR4 by LPS.
  • mice Balb/c (5 - 6 weeks) mice were injected with LPS (225 ⁇ g) intraperitoneally with and without pretreatment of RSCL-0520 (10 and 20mg/kg). The compound was injected intraperitoneally 30min before LPS treatment. The mice were monitored for lhr post LPS treatment. Untreated mice served as controls. Blood from retro-orbital plexus was collected under anesthesia lhr post LPS injection. Serum collected was analysed for TNF- ⁇ secretion using an ELISA detection kit. In untreated mice, LPS injection led to the secretion of large amounts on TNF- ⁇ in the serum.
  • compositions and methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the methods described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents that are chemically or physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention.

Landscapes

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

Abstract

The present invention relates to novel synthetic toll like receptor antagonist. The present invention in particular provides compounds, methods and compositions for specifically inhibiting immune stimulation involving TLR ligands, especially TLR-4. The compounds are potentially useful in treatment of inflammation, autoimmunity, allergy, asthma, graft rejection, graft versus host disease, infection, sepsis, cancer and immunodeficiency.

Description

"TOLL LIKE RECEPTOR (TLR) SIGNALING ANTAGONIST" CROSS-REFERENCE TO RELATED PATENT APPLICATIONS
The present application claims benefit of the filing date of Indian Provisional Patent Application No. 1853/MUM/2007 filed September 24, 2007, and application number 2541 /MUM/2007 filed December 24, 2007 which is entirely incorporated herein by reference.
FIELD OF THE INVENTION
The present invention relates to novel molecules, compositions and methods for preparation and modulation of immune functions mediated through Toll-like receptor (TLR) signaling.
BACKGROUND OF THE INVENTION:
The innate or natural immune system recognizes a wide spectrum of pathogens without a need for prior exposure. Cells of the innate immune system effectively prevent free growth of bacteria within the body; however, many pathogens have evolved mechanisms allowing them to evade the innate immune system, but unlike the adaptive immune system, it does not confer long-lasting or protective immunity to the host. Innate immune systems provide immediate defense against infection, and are found in all classes of plant and animal life. The main cells responsible for innate immunity are monocytes /macrophages and neutrophils, which phagocytose microbial pathogens and are responsible for triggering the innate, inflammatory, and specific immune responses.
Toll-like receptor are a family of receptors involved in the recognition of a wide range of microbial molecules e.g. Lipopolysaccharides (LPS) from Gram- negative bacteria and peptidoglycan from Gram- positive bacteria. The prototype receptor Toll was first identified in the fruit fly Drosophila but several TLR was found in mammals, particularly on mononuclear phagocytes. Toll-like receptors (TLRs) are a class of single membrane- spanning non-catalytic which are designated TLR2, TLR 4, TLR5 and each receptor recognizes a small range of structurally conserved molecules once they have breached physical barriers such as the skin or intestinal tract mucosa, and activate immune cell responses. They are believed to play a key role in the innate immune system. TLRs are a type of pattern recognition receptors (PRRs) and recognize molecules that are broadly shared by pathogens but distinguishable from host molecules, collectively referred to as pathogen-associated molecular patterns (PAMPs).
The discovery of the Toll-like receptors finally identified the innate immune receptors that were responsible for many of the innate immune functions that had been studied for many years. Interestingly, TLRs seem only to be involved in the cytokine production and cellular activation in response to microbes, and do not play a significant role in the adhesion and phagocytosis of microorganisms. Binding of TLR leads to the production of inflammatory cytokines, including TNF-alpha and IL- 12 and enhances the cells' antimicrobial killing mechanisms and antigen presenting capacity. The function of the TLRs was discovered by Beutler and colleagues. (Poltorak A, He X, Smirnova I, Liu MY, Van Huffel C, Du X, Birdwell D, Alejos E, Silva M, Galanos C, Freudenberg M, Ricciardi-Castagnoli P, Layton B, Beutler B. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science. 1998; 282: 2085-88.) These workers used positional cloning to prove that mice that could not respond to LPS had mutations that abolished the function of TLR4. This identified TLR4 as a key component of the receptor for LPS, and strongly suggested that other Toll-like receptors might detect other signature molecules of microbes, such as those mentioned above.
The chemical structure and the molecular basis of the recognition of LPS by serum proteins has gained attention in recent years, which has led to the discovery of a family of receptors like Toll like receptors. It has been estimated that most mammalian species have between ten and fifteen types of Toll-like receptors. Thirteen TLRs (named simply TLRl to TLRl 3) have been identified in humans and mice together, and equivalent forms of many of these have been found in other mammalian species' (Du,X., Poltorak, A., Wei,Y., and Beutler,B. Three novel mammalian toll-like receptors: gene structure, expression, and evolution. Eur. Cytokine Netw. 2000; 11 : 362-71; Chuang,T.H., and Ulevitch,R.J. Cloning and characterization of a sub-family of human toll-like receptors: hTLR7, hTLR8 and hTLR9. Eur. Cytokine Netw. 2000; 11 :372-78; Tabeta,K., Georgel,P., Janssen,E., Du5X., Hoebe,K., Crozat,K., Mudd,S., Shamel,L., Sovath,S., Goode,J. et al. Toll-like receptors 9 and 3 as essential components of innate immune defense against mouse cytomegalovirus infection. Proc. Natl Acad. ScL U. S. A. 2004; 101: 3516-21.).
The significance of the toll like receptors in the immune response to LPS has further demonstrated specifically two receptors TLR2 and TLR4' (Yang RB, Mark MR, Gray A, Huang A, Xie MH, Zhang M, Goddard A, Wood WI, Gurney AL, Godowski PJ. Toll-like receptor-2 mediates lipopolysaccharide-induced cellular signalling. Nature. 1998; 395: 284-88; Kirschning CJ, Wesche H, Merrill Ayres T, Rothe M. Human toll-like receptor 2 confers responsiveness to bacterial lipopolysaccharide. J Exp Med. 1998; 188: 2091-97.). Further reports concluded that TLR4 was required for a response to LPS. (Poltorak A, et al., Science. 1998; 282: 2085-88; Qureshi ST, Lariviere L, Leveque G, Clermont S, Moore KJ, Gros P, MaIo D. Endotoxin-tolerant mice have mutations in Toll-like receptor 4(Tlr4) J Exp Med. 1999; 189: 615-25).
Although LPS is an immύnomodulatroty agent, its medicinal use is limited due to its extreme toxicity including the induction of systemic inflammatory response syndrome. The biologically active endotoxio endotoxic sub-structural moeity of LPS is a lipid-A, a phosphorylated, multiple fatty acid acylated glucosamine disaccharide that serves to anchor the entire structure in the outer membrane of the gram-negative bacteria. The toxic effects of the lipid A was addressed by selective chemical modification of the lipid A to produce monophosphoryl lipid A compounds (MPL®: vaccine adjuvant and immunostimulant from Corixa (Seattle, Wash., US) and structurally like MPL® compounds) which is described in U.S. Patent: 4,436,727; 4,877,611; 4,866,034; 4,912,094; 4,987,237; and Johnson et.al. (Johnson DA, Keegan DS, Sowell CG, Livesay MT, Johnson CL, Taubner LM, Harris A, Myers KR, Thompson JD, Gustafson GL, Rhodes MJ, Ulrich JT, Ward JR, Yorgensen YM, Cantrell JL, Brookshire VG. 3-O- Desacyl monophosphoryl lipid A derivatives: synthesis and immunostimulant activities. J Med Chem. 1999; 42: 4640-49). US Pub. patent App. 20070167409 is based on discovery that animals that do not express Toll-like receptor 2 (TLR2) are protected from dextran sulfate sodium (DSS) induction of colitis, a model for inflammatory bowel disease (IBD). The invention relates to the agents that block activation of TLR2 to treat or to prevent colitis and related diseases or conditions, as well as other diseases or conditions characterized by activation of TLR2.
The European patent EP 1635846 has methods and compositions useful for modulating signaling through Toll-like receptors that involve contacting a TLR-expressing cell with a small molecule having a core structure including at least two rings. Certain of the compounds are 4-primary amino quinolines.
US Pub. patent App. US20060058365 relates to the treatment of inflammatory bowel disease (IBD) and related gastrointestinal pathologies that are cytokine-mediated ,or associated with Toll-like receptor 4 using methimazole derivatives and tautomeric cyclic thiones. Further US Pub. patent App. 20050004144 provides a broad-spectrum, long- lasting, and non-toxic combination of synthetic immunostimulatory agents, which are useful for activating the immune system of a mammal and treating diseases such as cancer and autoimmune disease involving 7-substituted, 8-substituted and 7,8-di substituted 7-deazaguanosines.
US Pub. patent App. 20030139364 involves administration of an imidazoquinoline agent in combination with another therapeutic agent in synergistic amounts to enhance ADCC, stimulate immune responses and/or patient and treat certain disorders.
Different phenanthrene derivatives that have been used in the prior art are discussed below:
The PCT patent WO 2006027345 discloses novel 3-Thia-10-aza-phenanthrene derivatives as novel effective PDE4 inhibitors. The PCT WO2006089881 has described novel phenanthrene derivatives as antiinflammatory agents and WO 9113855 has proivided a new phenanthrene derivative having IL-I inhibiting activity and useful for the treatment of chronic inflammatory diseases.
US patent 3683091 specifically provides di-7-hydroxy or methyl-2,3,4,4a,9,10- hexahydrophenanthren-2-one and 4a-alkyl derivatives, useful as specific anti-acne agents.
UK patent GB 1069067 provides novel phenanthrene derivatives having analgesic and morphine antagonistic activity.
US patent 476678 discloses phenathrene derivatives possessing valuable fungicidal properties useful in agriculture, horticulture and other antifungal compositions.
US patent 4808625 discloses aminoalkanol derivatives of containing a polycarbocyclic aromatic ring system such as phenanthrene, as biocidal agents, particularly antitumor agents.
Japanese patent JP 8067626 discloses hydrogenated condensed ring hydrocarbons such as hydrogenated phenanthrene (e.g. 1,2,3,4,5,6,7,8-octahydrophenanthrene), a hydrogenated anthracene (e.g. 1,2-dihydroanthracene), a hydrogenated naphthalene (e.g. 1,2- dihydronaphthalene), etc as capable of inhibiting the carcinogenesis induced by a carcinogenic organic compound without accompanied by side effects
UK patent GB2186570 provides 9, 10-dihydrophenanthrene derivatives which are useful in treating diseases characterized by an immunological imbalance and bacterial and viral infections in mammals.
Certain phenanthrene derivatives from plants have also been used in the prior art for immune system disorders such as 9, 10- dihydrophenanthrene called eulophiol isolated from the tubers of Eulophia nuda' (Bhandari SR and Kapadi AH. A 9,10- dihydrophenanthrene from tubers of Eulophia nuda; Phytochemistry. 1983; 22: 747-48; Tuchinda P, Udchachon J, Khumtaveeporn K, Taylor WC, Engelhardt LM, White, A.H. Phenanthrenes of Eulophia nuda. Phytochemistry. 1988; 27: 3267-71; Majumder PL, Sen S, Majumder S. Phenanthrene derivatives from the orchid Coelogyne cristata. Phytochemistry. 2001; 58: 581-586).
Japanese patent JP 7267895 has provided plant extracts containing phenanthrene derivatives from Raikoutou (a root or leaf of Tripterygium wilfordii Hook. F.) a Chinese herbal drug. These compounds have been found to be useful as a therapeutic agent for diseases owing to leukotriene of pollinosis, bronchial asthma, arthritis etc.
A number of phenanthrene derivatives have been extracted as a principal constituent from plant species Orchidaceae family has been reported' ^Majumder PL and Sen RC. Pendulin, a polyoxygenated phenanthrene derivative from the Orchidaceae Cymbidum pendulum. Phytochemistry. 1991; 30: 2432-2434; Shimizu M, Shogawa H, Hayashi T, Arisawa M, Suzuki S, Yoshizaki M, Morita N, Ferro E, Basualdo I, Berganza LH. Antiinflammatory constituents of topically applied crude drugs. III. Constituents and antiinflammatory effect of Paraguayan crude drug "Tamanda cuna"(Catasetum barbatum LINDLE). Chem Pharm Bull (Tokyo). 1988; 36: 4447-52.). These extracts have been used as antipyretic, antioxidant and antispasmolytic agents.
PCT application WO 2006089881 has focused on methoxy phenanthrene derivatives from Tamus communis as anti-inflammatory agents.
The past decade has seen an explosion in TLR antagonist research, including their potential implication in auto-immune and chronic inflammatory diseases.
OBJECT OF THE INVENTION
It is the object of the present invention to provide small molecules for modulation of immune functions through Toll like receptors. It is the object of this invention to provide small molecules for inhibitions of TLR signaling in response to TLR ligands.
It is the object of the present invention to provide phenanthrene derivatives and its analogs for modulation of the immune functions through toll like receptors.
It is the object of the present invention to provide methods of preparation or isolation of these phenanthrene derivatives and its analogs.
It is the object of the present invention to provide compositions of the phenanthrene derivatives and its analogs.
It is the object of the present invention to provide compositions useful for the prevention or treatment of inflammation, wounds, autoimmunity, allergy, asthma, graft rejection, graft versus host disease, infection, sepsis, cancer and immunodeficiency.
It is also the object of this invention to provide a method and composition for affecting the TLR mediated signaling in response to a TLR ligand.
It is the object of the present invention to provide phenanthrene based molecules for inhibition of TLR ligand, which can be used in combinations with other agents.
SUMMARY OF THE INVENTION
The inventors of the present invention have designed novel phenanthrene derivatives that act as antagonists of Toll-like receptors and methods and compositions for modulating the immune functions through Toll-like receptor. The novel phenanthrene derivatives of the present invention have found their potential in inhibiting signaling of Toll-like receptors.
The present invention focuses on novel derivatives of phenanthrenes for potential use in inhibition of immune stimulation involving toll like receptor ligands. These molecules have been developed for potential use in treatment of inflammation, autoimmunity, allergy, asthma, graft rejection, graft versus host disease, infection, sepsis, cancer and immunodeficiency. More specifically, whereas the agents described herein have been discovered to affect TLRs directly and thus directly affect TLR-bearing cells, e.g., antigen-presenting cells (APCs), such agents can be used in conjunction with additional agents which affect non-APC immune cells, e.g., T lymphocytes (T cells). Such an approach effectively introduces an immunomodulatory intervention at two levels: innate immunity and acquired immunity.
The present invention relates to phenanthrene derivatives, methods of their preparation, and compositions for use in TLR mediated immune conditions. The present invention also relates to compositions and methods for modulating immune functions mediated through Toll-like receptor (TLR) molecules.
In one embodiment the present invention provides compositions that are useful for the prevention or treatment of inflammation, wounds, autoimmunity, allergy, asthma, graft rejection, graft versus host disease, infection, sepsis, cancer, and immunodeficiency. In the preferred embodiments the compositions as described in the present invention or the compositions are useful as for inhibition of TLR signaling in response to TLR ligands.
In the preferred embodiments the compositions for inhibition of TLR signaling as described in the present invention in a therapeutically effective amount and pharmaceutically inert adjuvants, diluents or carriers.
In one embodiment the compositions as described in the present invention or composition and the method of manufacture comprising the same is believed to have the ability to inhibit TLR signaling under physiological conditions, and thereby would have corresponding effectiveness for prevention or treatment of inflammation, wounds, autoimmunity, allergy, asthma, graft rejection, graft versus host disease, infection, sepsis, cancer, and immunodeficiency. In yet other embodiments, the compositions as described in the present invention can be used to prevent or treat clinical manifestations and diseases caused by microbial pathogens.
In yet other embodiments on the basis of inhibition of TLR signaling, the composition can be used in veterinary medicine to prevent or treat clinical manifestations and diseases caused by microbial pathogens.
In preferred embodiments the present invention also provides pharmaceutical formulations either by itself or in a suitable pharmaceutically acceptable adjuvant useful for inhibition of TLR mediated clinical manifestations.
As a feature of the present invention, the methods of the invention can be combined with administration of additional agents to achieve synergistic effect on TLR-mediated immunostimulation. More specifically, whereas the agents described herein have been discovered to affect TLRs directly and thus directly affect TLR-bearing cells, e.g., antigen-presenting cells (APCs), such agents can be used in conjunction with additional agents which affect non-APC immune cells, e.g., T lymphocytes (T cells). Such an approach effectively introduces an immunomodulatory intervention at two levels: innate immunity and acquired immunity. Since innate immunity is believed to initiate and support acquired immunity, the combination intervention is synergistic
In another embodiment of the invention, a method of affecting TLR-mediated signaling in response to a TLR ligand is provided.
In one embodiment of the invention, a method of inhibiting TLR-mediated immunostimulatory signaling is provided.
In another embodiment, the invention provides a method of affecting TLR-mediated immunostimulation in a subject.
Methods of treatment for variety of conditions involving autoimmunity, inflammation, allergy, asthma, graft rejection, graft-versus-host disease (GvHD), infection, sepsis, cancer, and immunodeficiency. Generally, methods useful in the treatment of conditions involving infection, cancer, and immunodeficiency will employ small molecules that augment TLR-mediated signaling in response to a suitable TLR ligand. In some instances the methods can be used to inhibit or promote TLR-mediated signaling in response to a TLR ligand or TLR signaling agonist. In some instances the methods can be used to inhibit TLR-mediated immunostimulatory signaling in response to a TLR ligand or TLR signaling agonist. In some instances the methods can be used to inhibit or promote TLR- mediated immunostimulation in a subject. In some instances the methods can be used to inhibit TLR-mediated immunostimulation in a subject. In some instances the methods can be used to inhibit an immunostimulatory nucleic acid-associated response in a subject.
In one embodiment, the present invention provides molecules and methods useful for modulating TLR-mediated signaling. The molecules of the present invention are applicable to alter any TLR-mediated signaling in response to a suitable ligand or signaling agonist.
In one embodiment the present invention also provides methods for identifying agents that decrease or inhibit activation of Toll-like receptor 2. These methods involve (i) contacting a cell expressing the receptor with a candidate agent in the presence of an activator of the receptor ( in vitro or in vivo) and (ii) determining the effect of the agent on activation of the receptor. Detection of a decrease in activation of the receptor by the activator in the presence of the agent indicates the identification of agent that can be used to decrease or inhibit activation of the receptor. In these methods, the effect of the agent on the activation of the receptor can be determined by analysis of the expression of a reporter gene that is under the control of a promoter that is induced in a signaling pathway triggered by activation of the receptor.
In one aspect, the present invention provides compounds which can be isolated from plant species such as Eulophia. Preferably this base compound Eulophiol (RSCL-0520) is extracted and further derivatives can be prepared by synthetic routes. In another aspect the present invention also provides compounds, which can be prepared by synthetic routes.
In one aspect of the invention, a method of affecting TLR-mediated signaling in response to a TLR ligand is provided. The method according to this aspect involves contacting a cell expressing a TLR with an effective amount of a compound of Formula I
Figure imgf000012_0001
in which R1, R2, R3, R4, R5, R6, R7, R8, R9 and R10 are identical or different and may each be hydrogen, hydroxy, C1-6 alkoxy, C1-6 alkyl, halogen, haloalkyl, acyloxy, hydroxyalkyl, alkenyl, alkenyloxy, carboxyl, carbalkoxy, carbamido, a conjugated group, substituted or unsubstituted phenyl, substituted or unsubstituted heterocyclic group, nitro, amino, acylamino, dialkylamino, nitric oxide (NO) - releasing moiety, pharmaceutically acceptable salts, amides and esters thereof. Ring A, Ring B, and Ring C may be aliphatic or aromatic.
Some of the compounds of the present invention but not limited to the above general formula are listed below:
1. 2, 7-dihydroxy-3,4-dimethoxyphenanthrene.(RSCL-0518)
2. 2, 7-diacetoxy-3,4-dimethoxyphenanthrene (RSCL-0519)
3. 2,3,4,7-tetramethoxyphenanthrene.(RSCL-0575)
4. 1 ,5-dihydroxy-2,7-dimethoxy-9, 10-dihydrophenanthrene (RSCL-0521 )
5. l,5-diacetoxy-2,7-dimethoxy-9,10-dihydrophenanthrene (RSCL 0520)
6. 3,4-dimethoxyphenanthrene-2,7-bis-[(2E)-3-[3,4-bis(acetyloxy)phenyl]acrylate. (RSCL-0522) 7. 1 ,5-dibenzyloxy-2,7-dimethoxy phenanthrene (RSCL-0638)
The present invention and other objects, features, and advantages of the present invention will become further apparent in the following Detailed Description of the Invention and the accompanying Figures and embodiments.
BRIEF DESCRIPTION OF THE DRAWINGS
The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present disclosure, the inventions of which can be better understood by reference to one or more of these drawings in combination with the detailed description of specific embodiments presented herein. Tab. 1 Comparative analysis of anti-TNF activity of isolated/synthesized molecules
FIG. Ia shows that RSCL-0520 inhibits TLR4 induced TNF-α secretion in human monocytic (THP-I) cells.
FIG. Ib shows that RSCL-0520 inhibits TLR2 and TLR4 induced TNF-α secretion in and peripheral blood monocytes (PBMCs).
FIG. 2a shows inhibition of TNF-α secretion in THP-I cells by RSCL-0520 is dose- dependent
FIG. 2b indicates that RSCL-0520 is not toxic to THP-I cells
FIG. 3 shows RSCL-0520 inhibits LPS induced TNF- α release in PBMC
FIG. 4 shows the dose dependent effect of RSCL-0520 on different concentrations of LPS in THP-I cells
FIG. 5a shows effect of RSCL-0520 on TNF-α mRNA expression in THP-I cells in real time FIG. 5b shows effect of RSCL-0520 on mRNA expression of pro-inflammatory genes in THP-I Cells
FIG. 5c Demonstrates ability of RSCL-0520 to inhibit Arachidonic acid induced PGE2 release in A549 cells
FIG. 6 shows RSCL-0520 suppresses LPS induced Nitric oxide (NO) release in RAW 264.7 cells
FIG. 7a represents effect of RSCL-0520 on activation of NEMO and degradation of I kappa B-alpha (IκB-α).
FIG. 7b represents effect of RSCL-0520 activation of NF-κB.
FIG. 7c represents effect of RSCL-0520 on translocation of NF-κB to the nucleus
FIG. 8a represents effect of RSCL-0520 on TLR related genes
FIG. 8b represents RSCL-0520 inhibits MyD88 dependent TLR signaling by LPS.
FIG. 9a shows pre-treatment of RSCL-0520 suppresses LPS induced TNF- α release in Balb/c mice
FIG. 9b Treatment with RSCL-0520 post LPS induction suppresses the induced TNF- α release in Balb/c mice
DETAILED DESCRIPTION OF THE INVENTION
Definitions:
The term "synthetic small molecules" as used herein refers to molecules with basic phenanthrene backbone. The term "LPS" as used herein refers to Lipopolysaccharide. LPS, which is contained in the outer membrane of the cell wall of various gram-negative bacteria, consists of a glycolipid called "Lipid A" to which various saccharides are bonded. It has been known for along time that LPS is the main component of endotoxins.
The term "TLR" as used herein refers to Toll like receptor.
The term "pharmaceutically acceptable salt," as use herein, refers to those salts which are, within the scope of sound medical judgement, suitable for use in contact with the tissues of humans and animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al. describes pharmaceutically acceptable salts in detail. (Berge SM, Bighley LD, Monkhouse DC. Pharmaceutical salts. J Pharm Sci. 1977; 66:1-19). The salts can be prepared in situ during the final isolation and purification of a compound of the invention or separately by reacting the free base group with a suitable organic acid. Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphersulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3- phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, suberate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. The term "pharmaceutically acceptable ester," as used herein, represents esters that hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic, and alkanedioic acids, in which each alkyl or alkenyl group preferably has not more than 6 carbon atoms. Examples of particular esters include formates, acetates, propionates, butyates, acrylates, and ethylsuccinates. Amides and esters could also be prepared by coupling the compounds of the present invention with phenocli acids such as Non-steroidal anti-inflammatory drugs (NSAIDs) etc. The term "pharmaceutically acceptable pro-drugs," as used herein, means pro-drugs of the compounds of the present invention which are, within the scope of sound medical judgement, suitable for use in contact with the tissues of humans and animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention.
The term "prodrug," as used herein, represents compounds that are transformed in vivo into a parent compound of the above formula, for example, by hydrolysis in blood. A thorough discussion of prodrugs is provided in T. Higuchi and V. Stella (Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, Edward B. Roche, ed., "Bioreversible Carriers in Drug Design," American Pharmaceutical Association and Pergamon Press, 1987), and Judkins et al" (Synthetic Communications. 1996; 26: 4351- 67), each of which is incorporated herein by reference.
Asymmetric or chiral centers may exist in the compounds of the present invention. The present invention includes the various stereoisomers and mixtures thereof. Individual stereoisomers of compounds or the present invention may be prepared synthetically from commercially available starting materials that contain asymmetric or chiral centers or by preparation of mixtures of enantiomeric compounds followed by resolution well-known to those of ordinary skill in the art. These methods of resolution are exemplified by (1) attachment of a racemic mixture of enantiomers, designated (+/-), to a chiral auxiliary, separation of the resulting diastereomers by recrystallization or chromatography and liberation of the optically pure product from the auxiliary or (2) direct separation of the mixture of optical enantiomers on chiral chromatographic columns. Enantiomers are designated herein by the symbols "R" or "S," depending on the configuration of substituents around the chiral carbon atom, or are drawn by conventional means with a bolded line defining a substituent above the plane of the page in three-dimensional space and a hashed or dashed line defining a substituent beneath the plane of the printed page in three-dimensional space. If no stereochemical designation is made, it is to be assumed that the structure definition includes both stereochemical possibilities.
COMPOUNDS OF THE PRESENT INVENTION:
The present invention relates to the compounds of formula (I) and derivatives thereof including but not limited to polymorphs, isomers and prodrugs thereof, geometric or optical isomers thereof, and pharmaceutically acceptable esters, ethers, carbamates of such compounds, all solvates and hydrates thereof and all salts thereof.
Particularly the present invention provides the compounds of formula I which are represented by structure numbers as follows
A compound represented by the formula (I):
Figure imgf000017_0001
in which R1, R2, R3, R4, R5, R6, R7, R8, R9 and R10 are identical or different and may each be hydrogen, hydroxy, C1-6 alkoxy, C1-6 alkyl, halogen, haloalkyl, acyloxy, hydroxyalkyl, alkenyl, alkenyloxy, carboxyl, carbalkoxy, carbamido, a conjugated group, substituted or unsubstituted phenyl, substituted or unsubstituted heterocyclic group, nitro, amino, acylamino, dialkylamino, nitric oxide (NO) — releasing moiety, pharmaceutically acceptable salts, amides and esters thereof. Ring A, Ring B, and Ring C may be aliphatic or aromatic.
The compounds of the present invention may contain asymmetric or chiral centers, and therefore may exist in different stereoisomeric forms. All suitable optical isomers and stereoisomeric forms of the compounds of the present invention as well as mixtures thereof, including racemic mixtures, form part of the present invention. In addition, the present invention embraces all geometric and positional isomers. Moreover, some compounds of the present invention may exhibit polymorphism. The present invention includes all polymorphic forms of the compounds according to the invention, which forms the further aspect of the invention. It is to be understood that the present invention encompasses any and all racemic, optically-active, polymorphic and stereoisomeric forms, or mixtures thereof, which form or forms possess properties useful in the treatment of the conditions indicated herein.
Furthermore, the present invention also include isotopically-labeled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
PREPARATION:
All of the starting materials used in any of these methods are commercially available from chemical vendors such as Aldrich, Sigma, Nova Biochemicals, Bachem
Biosciences, Advanced ChemTech, and the like, or may be readily synthesized by known procedures.
The reaction products are isolated and purified by conventional methods, typically by solvent extraction into a compatible solvent. The products may be further purified by column chromatography or other appropriate methods, including medium pressure or high pressure liquid chromatography.
The compounds and methods of the invention are described in further detail, as follows: Synthesis of TLR antagonists:
A general method of synthesis includes the preparation of phenanthrene molecules by any of the following approaches:
1. A mixture of substituted benzaldehyde, 3, 4, 5-trimethoxyphenyl acetic acid, acetic anhydride and triethylamine is refluxed and then acidified to obtain acid (A) which is crystallized from suitable solvent. The acid (A) obtained is mixed with quinoline and copper chromite and refluxed under inert atmosphere and then precipitated with ethyl acetate. After filtration, the filtrate is washed with dilute hydrochloric acid, water, and brine, dried (sodium sulphate), and evaporated under reduced pressure to isolate the product. (B) A stirred solution of the product obtained (B) and iodine in ethanol is irradiated at 254 run. The solution is then evaporated under reduced pressure and the product (C) is isolated by silica gel column chromatography. (Pet-ether-ethyl acetate gradient)
SCHEME I:
Figure imgf000019_0001
A B C
2. An alternate approach to the above reaction is Suzuki coupling of the appropriate reactants 1 and 2 gives a biaryl dialdehyde 3. The compound 3 on Wittig olefination, and ring closing metathesis of the resultant diene 4 with the Grubbs second-generation ruthenium catalyst gives 5.
SCHEME II
Figure imgf000020_0001
3. An alternate approach is Pschorr synthesis wherein substituted nitroaldehyde, 3,4,5- trimethoxyphenyl acetic acid, acetic anhydride and triethylamine are refluxed. The mixture is then acidified and stirred. The precipitated solid is filtered off and washed with hot water, leaving acid (A). A solution of the acid (A) in 5N ammonium hydroxide is added to a slurry of iron (II) sulphate, and concentrated ammonium hydroxide at 80- 9O0C. After heating, the product is filtered and the precipitate is washed with 5N- ammonium hydroxide. The cooled filtrate is acidified with glacial acetic acid and the precipitate is filtered off and vacuum dried (B). To a mixture of amine (B), ethanol and 5N-hydrochloric acid at O0C is added a 50% ethanolic solution of isopentyl nitrite. The mixture stirred and then diluted with water, further copper powder is added and stirred at 500C. The cooled mixture is then extracted with ethyl acetate, which on concentration under reduced pressure gives acid (C). A mixture of acid (C), quinoline and basic copper carbonate is refluxed, and extracted with ethyl acetate. The ethyl acetate layer is washed with acid, alkali, and evaporated. The product (P) is purified by column chromatography on silica (with pet-ether-ethyl acetate gradient).
SCHEME III
Figure imgf000021_0001
Therapeutic Use of TLR antagonist
The present invention provides agents that can be used to prevent or to treat LPS mediated diseases or conditions that are characterized by TLR activation.
The conditions that are prevented or treated but are not limited to inflammatory bowel disease (IBD), sepsis, periodontal disease, mucositis, acne, cardiovascular disease, chronic obstructive pulmonary disease, arthritis, cystic fibrosis, bacterial-induced infections, viral-induced infections, mycoplasma-associated diseases, post herpetic neuralgia, ischemia/reperfusion injury, asthma, stroke,' brain injury, necrotizing enterocolitis, bed sores, leprosy, atopic dermatitis, psoriasis, trauma, neurodegenerative disease, amphotericin B-induced fever and nephritis, coronary artery bypass grafting, and atherosclerosis.
Delivery and dosage of the TLR antagonist:
The present invention provides compositions comprising carbohydrate based molecules in an effective amount that achieves the desired therapeutic effect for a particular condition, patient and mode of administration. The dosage level selected depends on the route of administration and the severity of the condition being treated.
For example: For adults, the doses are generally from about 0.01 to about 100mg/kg, desirably 0.1-lmg/kg by inhalation, desirably 0.5-10mg/kg per day by oral administration, and desirably 0.1-lmg/kg body weight per day by intravenous administration. Doses are determined for each particular case using standard methods in accordance with factors unique to the patient, including age, weight, general state of health, and other factors that can influence the efficacy of the compound(s) of the invention.
Further the administration of the compounds of the present invention is not limited to mammal, including humans, be limited to a particular mode of administration, dosage, or frequency of dosing.
The present invention encompasses all modes of administration, including oral, intraperitoneal, intramuscular, intravenous, intra-articular, intralesional, subcutaneous, or nasally, rectally, buccally, or any other route sufficient to provide a dose adequate to prevent or treat excess or undesired TLR activity.
The present invention also contemplates that one or more compounds may be administered to a mammal in a single dose or multiple doses. When multiple doses are administered, the doses may be separated from one another by, for example, several hours, one day, one week or one month.. It is to be understood that, for any particular subject, specific dosage regimes should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of a pharmaceutical composition that includes a compound of the invention.
The present invention provides compositions of phenanthrene derivatives being TLR antagonists which may be prepared by conventional methods using one or more pharmaceutically acceptable excipients or adjuvants which may comprise inert diluents, sterile aqueous media and/or various non toxic solvents. The pharmaceutically acceptable carrier or diluents may be used as described in literature' (The Science and Practice of Pharmacy (20th ed.), ed. A. R. Gennaro, Lippincott Williams & Wilkins, 2000, Philadelphia; Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988 1999, Marcel Dekker, New York).
The compositions may be presented in the form of tablets, pills, granules, powders, aqueous solutions or suspensions, injectable solutions, elixirs, or syrups, and the compositions may optionally contain one or more agents chosen from the group comprising sweeteners, flavorings, colorings, and stabilizers in order to obtain pharmaceutically acceptable preparations.
The choice of vehicle and the content of active substance in the vehicle are generally determined in accordance with the solubility and chemical properties of the product, the particular mode of administration, and the provisions to be observed in pharmaceutical practice. For example, excipients such as lactose, sodium citrate, calcium carbonate, and dicalcium phosphate and disintegrating agents such as starch, alginic acids, and certain complex silicates combined with lubricants (e.g., magnesium stearate, sodium lauryl sulfate, and talc) may be used for preparing tablets. To prepare a capsule, it is advantageous to use lactose and high molecular weight polyethylene glycols. When aqueous suspensions are used, they may contain emulsifying agents that facilitate suspension. Diluents such as sucrose, ethanol, polyethylene glycol, propylene glycol, glycerol, chloroform, or mixtures thereof may also be used.
For parenteral administration, emulsions, suspensions, or solutions of the compositions of the invention in vegetable oil (e.g., sesame oil, groundnut oil, or olive oil), aqueous- organic solutions (e.g., water and propylene glycol), injectable organic esters (e.g., ethyl oleate), or sterile aqueous solutions of the pharmaceutically acceptable salts are used. The solutions of the salts of the compositions of the invention are especially useful for administration by intramuscular or subcutaneous injection. Aqueous solutions that include solutions of the salts in pure distilled water may be used for intravenous administration with the proviso that (i) their pH is adjusted suitably, (ii) they are appropriately buffered and rendered isotonic with a sufficient quantity of glucose or sodium chloride, and (iii) they are sterilized by heating, irradiation, or microfiltration. Suitable compositions containing a compound of the invention may be dissolved or suspended in a suitable carrier for use in a nebulizer or a suspension or solution aerosol, or may be absorbed or adsorbed onto a suitable solid carrier for use in a dry powder inhaler. Solid compositions for rectal administration include suppositories formulated in accordance with known methods and containing at least one compound of formula I or II.
Dosage formulations of a compound of the invention to be used for therapeutic administration must be sterile. Sterility is readily accomplished by filtration through sterile membranes (e.g., 0.2 micron membranes) or by other conventional methods. Formulations typically are stored in lyophilized form or as an aqueous solution. The pH of the compositions of this invention is typically between 3 and 11, more desirably between 5 and 9, and most desirably between 7 and 8, inclusive. While a desirable route of administration is by injection such as intravenously (bolus and/or infusion), other methods of administration may be used. For example, compositions may be administered subcutaneously, intramuscularly, colonically, rectally, nasally, or intraperitoneally in a variety of dosage forms such as suppositories, implanted pellets or small cylinders, aerosols, oral dosage formulations, and topical formulations such as ointments, drops, and dermal patches. A compound of the invention is desirably incorporated into shaped articles such as implants, including but not limited to valves, stunts, tubing, and prostheses, which may employ inert materials such as synthetic polymers or silicones, (e.g., Silastic, silicone rubber, or other commercially available polymers). Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxy-propyl- methacrylamide-phenol, polyhydroxyethyl-aspartamide-phenol, or polyethyleneoxide- polylysine substituted with palmitoyl residues. Furthermore, a TLR2 inhibitor of the invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and cross linked or amphipathic block copolymers of hydrogels.
A compound of the invention may also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles. Liposomes can be formed from a variety of lipids, such as cholesterol, stearylamine, or phosphatidylcholines. A compound of the invention may also be delivered using antibodies, antibody fragments, growth factors, hormones, or other targeting moieties to which the compound molecules are coupled (e.g., see Remington: The Science and Practice of Pharmacy, vide supra), including in vivo conjugation to blood components of a compound of the formula I or II, as described herein.
In vitro application in identification of TLR antagonists:
Pharmaceutical agents that can be used in the therapeutic methods of the invention can be identified in screening methods. For example, cell-based screening methods can be used, in which cells expressing TLR are contacted with a candidate agent and the impact of the agent on the activation of TLR in the cells is determined. In one example of such a method, the effect of an agent on the activation of TLR by a known ligand (e.g., a lipopeptide,) is determined. Agents that are found to decrease or to block activation of the receptor by the ligand can then be considered for further analysis and/or for use as TLR inhibitors in therapeutic methods. Activation of TLR in these methods can be measured using, for example, a reporter system. For example, cells used in the screening assay can include a reporter gene that is under the control of a promoter that is inducible by a signaling pathway triggered by TLR activation.
In addition to cell-based methods, candidate agents can be tested in animal model systems. This may be desirable, for example, if an agent has been found to have antagonist activity in a cell-based assay or to bind to TLR in an in vitro assay (see below). For example, in animal studies, test agents can be administered to an animal model concurrently with a molecule known to activate TLR (e.g., lipopeptide), and the impact of the agent on a response in the animal that is normally triggered by activation of the receptor (e.g., cytokine induction) can be determined. Further, in vitro methods can be used. For example, a candidate compound can be assayed for whether it binds to TLR or a fragment of the receptor that includes at least a portion of the ligand binding site. Such assays can be carried out using, for example, columns or beads to which the receptor or fragment is bound.
In addition to the methods described above, additional TLR antagonists can be identified in methods in which candidate compounds are compared for TLR antagonist activity with any of the TLR antagonists described herein. Further, in addition to being compared for TLR antagonist activity, the candidate compounds can be compared with TLR2 antagonists with respect to specificity for TLR versus other receptors. Candidate compounds identified as having TLR antagonist activity that is, for example, similar to or greater than the activity of the antagonists described herein (and/or with similar or greater levels of specificity for TLR2 versus TLR4) in these assays can be tested further, for example, in appropriate animal model assays for any of the diseases or conditions described herein, as well as in human clinical studies.
Also included in the invention are compounds that are selective for TLR2 over TLR4, as well as compounds that are dual antagonists (i.e., antagonists of both TLR2 and TLR4). A compound that is selective for TLR2 over TLR4 is one that has, for example, an IC50 value in a TLR2 antagonist assay, such as is described herein, that is less than that found in a TLR4 antagonist assay. For example, the IC5O in the TLR2 assay can be at least 5, 10, 25, or 50-fold less than the value for the same compound tested in the TLR4 assay. Compounds that are dual antagonists are those that have, for example, IC50 values that are within a 5-fold range of one another using. Thus, dual antagonists include those that have activities that are 1 :5 5:1 with respect to one another (e.g., 1:4, 1 :3, 1:2, 1:1, 2:1, 3:1, and 4:1). The invention also includes the use of TLR2 antagonists such as those described herein in the study of physiological and molecular pathways involved in or affected by TLR2 activation (or inactivation).
Agents that can be screened using the methods of the invention include, for example, compounds that are present in compound libraries (e.g., random libraries), as well as analogs of known TLR2 ligands (e.g., lipopeptides) that are modified to prevent rather than activate TLR2. Further, peptides that correspond to the binding site of TLR2 or its ligands, which can competitively inhibit ligand binding, can be tested. Further, antibodies or antibody fragments to the ligand or the ligand binding site of the receptor can be screened. The following examples are included to demonstrate embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
The invention will now be illustrated with the aid of following non-limiting examples. It should be understood, however, that the invention is not limited to the solely to the particular examples given below. It will be apparent that those skill in the art that any modifications, both to the materials and methods, may be practiced without departing from the purpose and interest of this invention a) All operations which were carried out at room temperature or ambient temperature were in the range of 18 to 250C. b) Evaporation of the solvent was carried out under reduced pressure (600-4000 pascals; 4.5-30mm Hg) with a bath temperature up to 4O0C. c) The course of the reaction was monitored by thin layer chromatography (TLC) and reaction times are given for illustration only. d) Melting points are uncorrected, the melting points are given for the materials prepared as described, and polymorphism may result in isolation of materials with different melting points in some preparations. e) The structure and purity of all final products were assured by at least one of the following techniques: TLC, NMR (nuclear magnetic resonance) spectroscopy, IR (Infrared spectroscopy), or microanalytical data and HPLC f) Yields are given for illustration only. g) When given, NMR data is in the form of delta (.delta. )values for major diagnostic protons given in parts per million (ppm) relative to tetramethylsilane (TMS) as internal standard determined at 300 MHz or 400 MHz using the indicated solvent. h) Chemical symbols have their usual meanings; the following abbreviations have also been used: v(volume), w(weight), B.P.(boiling point), M.R.(Melting range), M.pt.(melting point), L(liters), ml(milliliters), gms(grams), mg(milligrams), mol (moles), mmol(millimoles) eq (equivalents) deg C (degree centigrade), cone. HCl (concentrated hydrochloric acid) any other
GENERAL PROCEDURE FOR PREPARATION OF TLR ANTAGONISTS
EXAMPLE - 1: Isolation of l,5-dihydoxy-2, 7-dimethoxy-9,10-dihydrophenanthrene (Eulophiol) (RSCL-520) and 2,7-dihydroxy-3,4-dimethoxyphenanthrene (Nudol) (RSCL-518)
1. Collection of plant materials:
Tubers of Eulophia ochreata were collected in the month of October 2006. Herbarium voucher specimen (Accession No. 157) was authenticated and deposited at herbarium of Dhirubhai Ambani Life Sciences Center, Navi Mumbai, Maharashtra.
2. Extraction of plant materials: (Scheme - 1)
Tubers were chopped in small pieces, shade dried and pulverized. Powder (lkg) was extracted under stirring with 61tr of pet ether (60-80) at room temperature for 7hr. The reaction mixture was filtered by using buchner funnel. Organic filtrate was stored and the cake was reextracted with 61tr of pet ether (60-80) under above conditions. The extraction was repeated one more time. The three extracts of pet ether were combined (about 15 ltr) and concentrated under vacuum to get dark yellow residue (4.6g). The cake was further extracted with dichloromethane three times using 61tr of solvent each time. The three extracts of dichloromethane (about 15 ltr) were mixed and concentrated under vacuum to yield dark brown residue (24g). Cake obtained after dichloromethane extraction was further extracted three times with ethyl acetate and followed by methanol using 61tr solvent each time. The removal of ethyl acetate (about 161tr) under vacuum gave dark black solid (6.4g) and concentration of methanol extract (about 171tr) under vacuum gave dark brown lump (4.9g). 3. Testing of free radical scavenging activity of various fractions:
Residue obtained after removal of solvents were tested for free radical scavenger activity using curcumin as reference standard and DPPH (2, 2-Diphenyl-l-picryl-hydrazyl) as radical.
Test solution preparation: DPPH was dissolved in methanol (AR grade) to a concentration of 1OmM.
Sample preparation: Weighed about one mg of extract residue/curcumin and dissolved in lOOμl of DMSO and added methanol in order to make concentration lmg/ml. Pipetted out 50μl sample solution in a 96-well micro titre plate to which added 200μl of above prepared DPPH test solution. The plate was incubated in dark for half an hour. Absorbance measured at 540nm on Eliza plate reader. The corresponding blank readings were also taken for calculating the percentage antioxidant activity. Antioxidant activity in percentage was calculated by the formula: (1 -(Absorbance of sample / Absorbance of DPPH) X lOO.
Table 1: Free radical scavenging activity of different extracts in DPPH assay.
Figure imgf000029_0001
* Reference standard.
4. Isolation of EULOPHIOL and NUDOL from dichloromethane fraction:
Based on free radical scavenging activity of different fractions as above, the residue of both dichloromethane and ethyl acetate extracts were combined and chromatographed over 30Og silica gel (60-120 mesh) in a glass column (90 x 5 cm). A mixture of pet ether and ethyl acetate in different combinations were used as mobile phase. The eluted fractions with pet ether and ethyl acetate (89:11 v/v) containing Eulophiol were collected where as the fractions eluted with pet ether and ethyl acetate (89:15 v/v) were collected for nudol. TLC monitoring system was pet ether and ethyl acetate (45:55, v/v) as the mobile phase. RF values calculated for eulophiol and nudol were 0.7 and 0.6 respectively.
5. Purification and characterization: Eulophiol:
All fractions containing Eulophiol were mixed and concentrated to remove the total solvent under vacuum. The syrupy mass left at room temperature for a week to get white crystals. The crystals were suspended in dichloro methane (5ml), filtered, washed and dried in oven under vacuum to get Eulophiol. Yield: 0.150g (0.014 %) (Reported9 0.21%). M.P. 201 - 203 0C (lit9., 202-203); 1H NMR: (400MHz, CD3COCD3, δ ppm) δ 2.59-2.75 (m, 4H, H-9 and H-10), δ 3.82 (S, 3H, OMe), δ 3.85 (S, 3H, OMe), δ 6.39 (d, IH, J=2.4Hz, meta coupled, H-6), δ 6.45 (d, IH, J=2.4Hz, meta coupled, H-8), δ 6.78 (d, IH, J=8.4Hz, ortho coupled, H-3), δ 7.28 (br s, IH, exch.D2O, OH), δ 7.72 (d, IH, J=8.4Hz, ortho coupled, H-4), δ 8.37 (br s, IH, exch.D2O, OH); MS: m/z 273 (M+l)
HPLC analysis showed 98.89% purity. Analysis was performed on column Hypersil GOLD Cl 8, 5μm, 4.6 x 250 mm, Wave length: 265 nm, Mobile phase: Buffer : Acetonitrile, Buffer: 2ml TEA = 1 litre water. Adjust pH to 3.0 with phosphoric acid, Run time: 60 min, Injection volume: 20μl, Sample preparation: O.lmg/ml, Diluent: Acetonitrile.
Structure:
Figure imgf000031_0001
Eulophiol (RSCL-0520) Nudol:
All the fractions containing Nudol were mixed and concentrated to minimum volume of 3-4ml and added 10- 12ml of pet ether to form the precipitate. Precipitate was stirred in pet ether, filtered and washed. Dark brownish hard crystals of nudol got separated on filter paper. Yield: 0.663g (0.063 %). (Reported yield is 0.003%; Bhandari SR, Kapadi AH, Majumder PL, Joardar, Mukta, Shoolery JN. Nudol, a phenanthrene of the orchids Eulophia nuda, Eria carinata and Eria stricta. Phytochemsitry.1985; 24: 801-804.). M. P: 253 0C (lit., 253; Id); 1H NMR: (400MHz, CDCl3, δ ppm) δ 3.95 (S, 3H, OMe), δ 4.05 (s, 3H, OMe), δ 5.08 (br s, IH, exch.D2O, OH), δ 6.01 (br s, IH, exch.D2O, OH), δ 7.08 (s, IH, H-I), δ 7.18 (dd, 1H,J=9.2 Hz, J=2.8Hz, ortho and meta coupled, H-6), δ 7.22 (d, IH, J=2.8Hz, meta coupled, H-8), δ 7.53 (AB-quartet, 2H, J=9.0Hz, H-9 and H-IO), δ 9.33 (d, IH, J=9.2Hz, ortho coupled, H-5); MS: m/z 271 (M+l).
HPLC analysis showed 97.02% purity. Analysis was performed on column Hypersil GOLD C18, 5μm, 4.6 x 250 mm, Flow rate: lml/min, Wave length: 257, 275 run, Mobile phase: Buffer : Acetonitrile, Buffer: 2ml TEA = 1 litre water. Adjust pH to 3.0 with phosphoric acid, Run time: 35min, Injection volume: 20μl, Sample preparation: O.lmg/ml, Diluent: Acetonitrile.
Figure imgf000032_0001
Nudol (RSCL-0518)
Scheme I
Extraction and isolation:
Figure imgf000032_0002
EXAMPLE 2: Derivatization of isolated molecules
A. Synthesis of l-S-diacetoxy^^-dimethoxy-^lO-dihydrophenanthrene^RSCL- 0521)
To a solution of eulophiol (I) (0.02Og, 0.07 mmol) in dichloromethane (20ml) was added acetic anhydride (0.020ml), pyridine (0.020ml) and stirred overnight at R.T. (20-250C). To this was then added water (50ml) and the organic layer washed with dilute hydrochloric acid (10ml), water (10ml), dried over sodium sulfate and concentrated under reduced pressure, which gave pure compound (III) (RSCL-0521) (0.022g) in 84.61% yield (w/w). M.P. (143-144 0C (reported Bhandari SR et al. Phytochemistry. 1983; 22: 747-48)., 143 0C; 1U NMR: (400MHz, CD3COCD3, δ ppm) δ 2.25 (S, 3H, OAc), δ 2.29 (S, 3H, OAc), δ 2.59-2.70 (m, 4H, H-9 and H-10), δ 3.84 (S, 3H, OMe), δ 3.89 (S, 3H, OMe), δ 6.66 (d, IH, J=2.4Hz, meta coupled, H-6), δ 6.78 (d, IH, J=2.4Hz, meta coupled, H-8), δ 6.96 (d, IH, J=8.4Hz, ortho coupled, H-3), δ 8.16 (d, IH, J=8.4Hz, ortho coupled, H-4), MS: m/z 357 (M+l).
Figure imgf000033_0001
B. Synthesis of 2, 7-diacetoxy-3,4-dimethoxyphenanthrene2 ( RSCL-0519)
To a solution of nudol (II) (0.02Og, 0.07 mmol) in dichloromethane (20ml) was added acetic anhydride (0.020ml), pyridine (0.020ml) and stirred overnight at R.T. (20-250C). To this was then added water (50ml) and the organic layer washed with dilute hydrochloric acid (10ml), water (10ml), dried over sodium sulfate and concentrated under reduced pressure, which gave pure compound (IV) (RSCL-0519) (0.108g) in 82.44% yield (w/w). M.P. 1510C, (151°C; Bhandari et al. Phytochemsitry.1985; 24: 801-804.); 1H . NMR: (400MHz, CDCl3, δ ppm) δ 2.37 (s, 3H, OAc), δ 2.46 (s, 3H, OAc), δ 3.92 (s, 3H, OMe), δ 3.98 (s, 3H, OMe), δ 7.15 (s, IH, H-I), δ 7.36 (dd, IH, J=9.2Hz, J=2.8Hz, ortho and meta coupled, H-6), δ 7.58 (d, IH, J=2.8Hz,meta coupled, H-8), δ 7.64 (apparent singlet (AB-quartet), 2H, H-9 and H-IO), δ 9.42 (d, IH, J=8.8Hz, ortho coupled, H-5) MS: m/z 355 (M+l).
Scheme III
Figure imgf000034_0001
C. Synthesis of 2,3,4,7-tetramethoxyphenanthrene2 (RSCL-0575)
To a solution of nudol (II) (0.027g, O.lmmol) in acetone (20ml) was added Potassium Carbonate (0.069g, 0.5 mmol), Methyl Iodide (0.5ml) and stirred at RT (20-250C) overnight. The reaction mixture was then filtered and the compound (V) (RSCL-0575) was isolated by silica gel column chromatography (Pet-ether-Ethyl acetate gradient). Yield = 0.0236g (79.19% w/w) M.P. 148, (148 °C; Bhandari et al. Phytochemsitry.1985; 24: 801-804.); 1H NMR: (400MHz, CDCl3, δ ppm) δ 3.95 (s, 3H, OMe), δ 4.00 (s, 3H, OMe), δ 4.03 (s, 3H, OMe), δ 4.05(s, 3H, OMe), δ 7.00 (s, IH, H-I), δ 7.16 (dd, J=9.2Hz, J=2.8Hz, ortho and meta coupled, H-6), δ 7.19 (d, IH, J=2.8Hz, meta coupled, H-8), δ 7.51 (apparent singlet (AB-quartet), 2H, H-9 and H-IO), δ 9.34 (d, IH, J=9.2Hz, ortho coupled , H-5), MS: m/z 299 (M+l). Scheme IV
Figure imgf000035_0001
EXAMPLE 3: SYNTHESIS OF NOVEL TLR ANTAGONISTS
1. Synthesis of 3,4-dimethoxyphenanthrene-2,7-bis-r (2E)-3-F 3,4-bis (acetyloxy) phenyl 1 acrylate (IX) ( RSCL-0522)
A. Synthesis of 3,4-Di-O-acetyleaffeic acid (VII)
To a solution of 3,4-Dihydroxycinnamic acid (VI) (1.8g, lOmmol) in anhydrous pyridine (1.8ml) was added acetic anhydride (2.25g, 22 mmol) and stirred overnight at room temperature (20-250C). The reaction was quenched by adding water (5ml) and extracted with ethyl acetate (3 X 50ml). The ethyl acetate layer washed successively with saturated sodium bicarbonate, brine and dried over anhydrous sodium sulfate. After concentration under reduced pressure the product (VII) was recrystallized from methanol. Yield 2.2g (83%), M.P 198-1990C, 1H NMR: (400MHz, CDCl3, δ ppm} 2.32 (s, 6H, two OAc), 6.40 (d, IH, Hα,Jαβ=16.0 Hz), 7.25 (d, IH, H-5, J5,6=8.8Hz), 7.39 (d, IH, H-2, J2;6=2.0Hz), 7.44 (dd, IH, H-6, J6,5=8.8 Hz, J6,2=2.0 Hz), 7.72 (d, IH, Hβ, Jβα=16.0 Hz,) MS: m/z 287 (M+Na)
B. Synthesis of 3,4-Di-O-acetylcaffeoyl chloride (VIII)
To a solution of 3,4-di-O-acetylcaffeic acid (VII) (0.063g, 0.24mmol) in anhydrous ethylene dichloride (10ml) were added thionyl chloride (ImI), N,N'-dimethyl formamide
(Cat) and refluxed in an oil bath (80-90 C) for lhr. The excess solvent and reagent were distilled off & pure acyl chloride (VIII) was immediately used as such without further purification.
C. Synthesis of 3,4-dimethoxyphenanthrene-2, 7-bis-[ (2E)-3-[ 3,4-bis (acetyloxy) phenyl ] acrylate (IX)
To the above acyl chloride (VIII) in dichloromethane (50ml) was added nudol (II) (0.027g, 0.1 mmol) and stirred overnight at room temperature (20-250C). The reaction was quenched by adding water (10ml) and extracted with dichloromethane (3 X 50ml). The dichloromethane layer was washed successively with saturated sodium bicarbonate, brine and dried over anhydrous sodium sulfate. After concentration under reduced pressure the product (IX) (RSCL-0522) was isolated by silica gel column chromatography (Pet-ether-ethyl acetate gradient). Yield 0.036g (47 %), M.P. 174-1750C, 1H NMR: (400MHz, CDCl3, δ ppm} δ 2.33 (S, 12H, OAc), δ 3.95 (S, 3H, OMe), δ 3.99 (S, 3H, OMe), δ 6.65 (d, lH,J=16Hz), δ 6.76 (d, IH, J=16Hz), δ 7.19 (S, IH), δ 7.28 (d, IH), δ 7.30 (d, IH), δ 7.40-7.55 (m, 5H), δ 7.68 (d, 3H), δ 7.87 (d, IH, J=I 6Hz), δ 7.93 (d, IH, J=16.4Hz), δ 9.47 (d, IH, J=9.2Hz) MS: m/z 785 (M+Na)
Scheme V
Figure imgf000036_0001
Figure imgf000037_0001
2. Synthesis of l,5-dibenzyloxy-2,7-dimethoxy phenanthrene (RSCL-0638)
A. Synthesis of the intermediates 6,6'-dibenzyloxy-4,4'-dimethoxy-biphenyl-2,2'- dicarbaldehyde(XII), 3,6'-dibenzyloxy-4,4'-dimethoxy-biphenyl-2,2'- dicarbaldehyde) (XIII), (3,3'-dibenzyloxy-4,4'-dimethoxy-biphenyl-2,2'- dicarbaldehyde) (XIV).
Synthesis of the intermediates 6,6'-dibenzyloxy-4,4'-dimethoxy-biphenyl-2,2'- dicarbaldehyde(XII), 3 ,6 ' -dibenzyloxy-4,4 ' -dimethoxy-biphenyl-2,2 ' -dicarbaldehyde) (XIII), (3,3'-dibenzyloxy-4,4'-dimethoxy-biphenyl-2,2'-dicarbaldehyde) (XIV were first carried out as per the procedure reported in the literature (Spencer B. Jones, Liwen He, and Steven L. Castle, Total Synthesis of(±)-Hasubanoine, Org.lett. 2006; 8: 3757-3760.). A three necked flask previously flushed with argon was charged with bis(pinacolato diboron) (3.6 Ig, 14.21 mmol), Potassium acetate (3.60g, 36.71 mmol), PdCl2 (dppf), CH2Cl2(0.29g, 0.35 mmol), dimethyl sulphoxide (35ml), stirred for 5-10 min, added a solution of 3-Benzyloxy-2-bromo-5-methoxybenzaldehyde (X) (3.8g, 11.84 mmol) in dimethyl sulphoxide (15ml) slowly over a period of 10-15min. (Mark A. Rizzacasa and Melvyn V. Sargent, Synthetic Approaches to the Alkaloids of the Ancistrocladaceae. Part3. The Total Synthesis of (-)-Ancistrocladinine: Control of the Diastereoisomer Excess in the Synthesis of Axially Chiral Biaryls, J.Chem.Soc.Perkin. Trans.I. 1991; 2773-2781) The resulting mixture was heated gently for two hours at 800C. Cooled to room temperature (20-25 C), added Potassium carbonate (5.24g, 37.9 mmol), PdCl2(dppf) CH2Cl2(LlOg, 1.42 mmol), a solution of 2-Benzyloxy-6-bromo-3- methoxybenzaldehyde (IV) (4.55g, 14.17 mmol) in 100ml dimethyl sulphoxide (90ml +
10ml rinse) and heated gently for two hours at 800C. (Takeshi Nakanishi, Masanobu
Suzuki, Akihiro Mashiba, Keizou Ishikawa and Takashi Yokotsuka, Synthesis of NKl 09, an anticancer Benzo(c)phenanthridine Alkaloid, J.Org.Chem. 1998; 63: 4235-4239) The reaction quenched by adding water (200ml), added excess sodium chloride & extracted with ethyl acetate (3 x 50ml), washed the ethyl acetate layer with brine (50ml), dried over sodium sulfate, and the mixture was subjected to silica gel column chromatography (pet ether-ethyl acetate gradient). This gave compound XII (0.9g), compound XIII (2.Og) and compound XIV (0.6g) as semi solid materials which solidified on standing for 24 hr at
R.T (20-250C).
Compound XII (6,6'-dibenzyloxy-4,4'-dimethoxy-biphenyl-2,2'-dicarbaldehyde)
M.P. 90-950C, 1HNMR: (400MHz, CDCl3, δ ppm) δ 3.81(s, 6H, two OCH3), 4.91(m, 4H,
CH2), 6.73 (d, 2H, J=2.4 Hz, ArH), 7.0-7.2(m, 12H), 9.64(s, 2H, CHO),
MS: m/z 483(M+l)
Compound XIII (3,6' -dibenzy loxy-4,4 ' -dimethoxy-bipheny 1-2 ,2 ' -dicarbaldehyde)
M.P. 78-850C, 1HNMR: (400MHz, CDCl3, δ ppm) δ 3.79(s, 3H, OCH3), 3.79(s, 3H,
OCH3), 3.90(s, 3H, OCH3), 4.86(s, 2H, CH2), 5.1(m, 2H, CH2), 6.62-7.40 (m, 14H, ArH),
9.55(s, IH, CHO), 10.10(s, IH, CHO), MS: m/z 483(M+l)
Compound XFV (3,3'-dibenzyloxy-4,4'-dimethoxy-biphenyl-2,2'-dicarbaldehyde)
M.P. 68-7O0C, 1HNMR: (400MHz, CDCl3, δ ppm) δ 3.87(s, 6H, two OCH3) 5.10(m, 2H,
CH2), 5.1(m, 2H, CH2), 6.75 (d, 2H, J=8.4 ArH), 7.20-7.40(m, 1OH, ArH), 10.03(s, 2H,
CHO), MS: m/z 483(M+1)
B. Synthesis of 6,6'-dibenzyloxy-4,4'-dimethoxy-2,2'-divinyl-biphenyl (XV) To a suspension of methyltriphenylphosponium bromide (0.37g, 1.037 mmol) in anhydrous tetrahydrofuran (11.6ml) was added dropwise n-Butyl Lithium (0.74 mLl, 1.6 M in hexanes, 1.18 mmol) at 0°C. The solution was warmed to RT (20-25°C) and stirred for three hours, then cooled to -78°C and added a solution of 6,6'-dibenzyloxy-4,4'- dimethoxy-biphenyl-2,2' -dicarbaldehyde (XII) (O.lg, 0.207 mmol) in anhydrous tetrahydrofuran (10.4ml) dropwise over a period of ten minutes. The resultant mixture was stirred for ten minutes, then warmed to RT (20-25°C) and stirred overnight. The reaction was quenched by the addition of IN hydrochloric acid (pH changed to 4-5), diluted with brine (20ml), extracted with ethyl acetate (3 X 20ml), dried over sodium sulfate, and the product, an oil, was isolated by silica gel column chromatography (pet- ether-ethyl acetate gradient). Yield- 0.058g (58.58%), 1HNMR: (400MHz, CDCl3, δ ppm) δ 3.77(s, 6H, two OCH3), 4.88(s, 4H, CH2), 5.00(d, 2H, J=I 0.8), 5.54(d, 2H, J=I 8.0 Hz), 6.30(m, 2H), 6.4 l(d, 2H, J=2.4 Hz), 6.76(d, 2H, J=2.4 Hz), 7.0-7.2(m, 10H) MS: m/z 479(M+l)
C. Synthesis of l,5-dibenzyloxy-2,7-dimethoxy phenanthrene (XVI)
To a solution of 6,6'-dibenzyloxy-4,4'-dimethoxy-2,2'-divinyl-biphenyl (XV) (0.048g, 0.1 mmol) in dichloromethane (10ml) was added the Grubbs second-generation ruthenium catalyst (0.0025g, 0.003 mmol), refluxed for two hours under argon, concentrated the reaction mixture in vacuo and the product was isolated by silica gel column chromatography (pet-ether-ethyl acetate gradient). Yield=0.020g (44%) M.P. 75-8O0C, 1HNMR: (400MHz, CDCl3, δ ppm) δ 3.86(s, 6H, two OCH3), 4.80(s, 4H, CH2), 6.62(d, 2H, J=2.4, ArH), 6.77(d, 2H, J=2.4 Hz, ArH), 7.0-7.2(m, 1OH, ArH), 7.45(s, 2H, H-9 & H-10), MS: m/z 451(M+1) Scheme VI
Figure imgf000039_0001
EXAMPLE 9: In-Vitro testing
Experiment 1: Comparative TNF inhibition activity chart of isolated and synthesized molecules:
The molecules isolated and synthesized were initially pre-screened for TNF inhibition activity to identify the most effective anti-inflammatory molecule. RSCL-0520 (Tab. 1) showed the best TNF inhibiting activity, which we took forward for further in-vitro studies.
Experiment 2: Effect of RSCL- 0520 on various TLR ligands
The present invention has checked the effect of various TLR ligands on THP-I monocytes, PBMCs and their ability to activate and release TNF-α. For this purpose about 9 TLR ligands were used. These ligands were obtained from Apotech Cat; APO- 54N-018-KI01 and assayed for TNF-α release in culture supernatants. THP-I (Fig. Ia) and PBMCs (2 x 105 cells/well) (Fig. Ib) were plated in 96-well plate. The cells were pretreated with RSCL-0520 in DMSO lhr prior to TLR ligand treatment. Following lhr pre-treatment, the cells were treated with TLR ligands [TLR2, TLR5 and TLR6 -75ng/ml each, TLR3-75μg/ml, TLR4-750ng/ml, TLR7, TLR8 and TLR9-7.5μg/ml) for 24hrs. The culture supernatant were collected after the stipulated time and assayed for TNF-α release using a Duoset Enzyme-Linked Immunosorbent Assay (ELISA) detection Kit (R&D systems, MN 55413, USA; Cat: DY-210 E). Simultaneously, supernatants from cells treated with ligands without RSCL-0520 pre-treatment and RSCL-0520 without ligand treatment were collected which served as respective controls. We detected TNF-α secretion from cells stimulated with TLR2, TLR4 and TLR6. No detectable TNF-α was observed following stimulations with other ligands. In THP-I cells pretreated with RSCL-0520, TNF-α production decreased by -50% following TLR4 ligand stimulation. Further, the same inhibitory effect was seen in PBMCs (~%50 TLR4 following TLR4 ligand stimulation. These results show that RSCL-0520 selectively inhibits TNF-α production mediated mainly by TLR4 in THP-I and its inhibition in addition to TLR4 extended to TLR2 in PBMCs. This clearly indicates that RSCL-0520 exerts its inhibition primarily TLR-4 signaling mechanisms. Experiment 3: Inhibition of TNF-α secretion in THP-I cells by RSCL-0520 is dose- dependent
To confirm whether the inhibitory effect of RSCL-0520 is dose-dependent, the present invention has studied the ability of RSCL-0520 to inhibit TNF-α secretion from LPS (250ng/ml) induced THP-I monocytes (Fig. 2a).
THP-I, 2 x 105 cells/well was plated in 96-well plate. The cells were pretreated with RSCL-0520 at various concentrations (lOOμM, 50μM, lOμM and lμM) lhr prior to LPS stimulation. As a control group, cells were treated with LPS alone and cells treated with RSCL-0520 alone were used. TNF-α secretion was estimated in the culture supernatants following 24hr LPS stimulation using Duoset ELISA detection Kit (R&D systems, MN, USA). ***P < 0.001 values are comparisons for LPS treated vs. RSCL-0520 treated, NS indicates not significant.
The toxicity of RSCL-0520 (Fig. 2b) was also by treating cells with RSCL-0520 by MTT assay. LPS induced TNF-α secretion was inhibited by RSCL-0520 in a concentration dependent manner. The viability of the cells (analyzed in tandem by MTT) was not affected by RSCL-0520 indicating its non-toxic nature.
Experiment 4: Effect of RSCL-0520 on LPS induced TNF- release in human PBMC
To evaluate the ability of RSCL-0520 to inhibit LPS induced TNF-α secretion in a physiological scenario, the present invention has tested the same in PBMC isolated from human blood (Fig. 3) by standard methodology. The inhibition was similar to that observed in THP-I cells. The TNF levels were not detectable in PBMCs without LPS and with RSCL-0520 treatment alone, indicating its specificity in LPS induced TNF-α through a TLR mediated process. ***P < 0.001 value is for LPS treated vs. RSCL-0520 treated.
Experiment 5: Effect of RSCL-0520 on TNF response to increasing concentration of LPS
The present invention has also conducted experiments to check the ability of RSCL-0520 to inhibit LPS induced TNF-α secretion from THP-I cells, wherein the studies involved the stimulation of THP-I cells with increasing concentrations of LPS (31.25ng/ml to lOOOng/ml) with and without pre-treatment of cells with different concentrations of RSCL-0520. It is clearly evident (Fig. 4) that with an increased secretion of TNF-α with increasing concentration of LPS is inhibited by RSCL-0520 to varied extents on a concentration dependent manner. Further, even RSCL-0520(10μM) was effective to inhibit LPS (1000ng/ml) induced TNF-α secretion; clearly indicating it's potency as an antagonist to LPS induced processes.
Experiment 6: Effect of RSCL-0520 on TNF-α mRNA expression in THP-I Cells
To determine whether the suppressive effect of RSCL-0520 on the cytokine production occurs at mRNA expression level, quantitative real- time PCR was used to examine TNF- α mRNA expression in THP-I cells stimulated with LPS. THP-I cells (3 x 106 cells/well) were stimulated with 250ng/ml LPS in the presence or absence of RSCL-0520 (50μM) for the lhr. Total RNA was isolated from these cells and cDNA was synthesized. LPS treated cells acted as positive control. All quantitative real-time PCR (TaqMan™) primers and probes were obtained from Applied Biosystems(ABI), Weiterstadt, Germany. For detection of TNF-α, pre-developed assay reagents (Universal master mix as obtained from Applied Biosystems included all reagents including Taq-polymerase apart from specific primers and probes) were used. The PCR was performed utilizing 1 μl cDNA per reaction in triplicates of 25μl volume on an ABI 7500 Realtime PCR machine using a 2- step PCR protocol.
Using the comparative threshold cycle method and standard software mRNA Quantization was carried out and the data expressed as fold change of TNF-α after correction with internal control β-actin. We observed that lhr post LPS stimulation, RSCL-0520 down regulated TNF-α to almost control levels (Fig. 5a). On the other hand, TNF-α mRNA expression increased ~7 fold after the stimulation with LPS. ***P < 0.001 value is for LPS treated vs. RSCL-0520 treated followed by LPS treatment and NS is for Control Vs RSCL-0520.
Experiment 7: Effect of RSCL-0520 inhibits mRNA expression of pro-inflammatory genes in THP-I Cells.
The inhibitory effect of the present invention, RSCL-0520 on mRNA was checked on pro-inflammatory genes like intercellular adhesion molecule 1 (ICAM-I), cyclooxygenase 2 (COX-2), IL-I β and IL-8. THP-I cells (3 xlO6 cells) were seeded in a 6-well dish were treated with RSCL-0520 (50μM) for lhr followed by incubation with or without LPS (250ng/ml). Following two washes with ice-cold PBS, the cells were harvested and total cellular RNA was isolated using TRIZOL Reagent (Invitrogen) according to the manufacturer's instructions. cDNA was synthesised using high capacity cDNA reverse transcription kit (ABI systems). Amplification of ICAM-I, COX-2, IL- lβ and IL-8 genes from the cDNA was carried out using the respective gene specific primers. RSCL-0520 inhibited mRNA expression levels of the tested genes (Fig. 5b), indicating that its mechanism of action is via NF-κB. However, cells treated with RSCL-0520 in the absence of LPS, did not alter the gene expressions of the genes in consideration highlighting its specificity.
Experiment 8: Effect of RSCL-0520 on Arachidonic acid (AA) induced PGE2 release in A549 cells.
COX-2 is the key enzyme regulating the production of prostaglandins, which act as central mediators of inflammation. Our earlier in- vitro data clearly demonstrated that the present invention inhibits expression of COX-2. It is well documented COX-2 pathway inhibitors were regarded as promising nonsteroidal anti-inflammatory drugs (NSAIDs). So we decided to test the ability of RSCL-0520 to block the COX-2 pathway to substantiate our earlier mRNA observation. For that purpose we chose A549 cells, a human lung cancer cell line where COX-2 is activated by AA in serum-free stimulation established by Yao et al for 12hr. A549 cells (50 x 104 cells/well) in serum free medium were pretreated with different concentrations (lOμM, 5μM, 2.5μM, and 1.25μM) of the NSAIDs and RSCL-0520 for 30min. (Yao JC, Duan WG, Yun Y, Liu de Q, Yan M, Jiang ZZ, Zhang LY. Screening method for nonsteroidal antiinflammatory drugs based on the cyclooxygenase 2 pathway activated by serum-free stimulation in A549 cells. Yakugaku Zasshi. 2007; 127: 527-32.) Then the cells were incubated with AA (lOμM) for another 30 min. Prostaglandin E2 (PGE2), a metabolite of AA through the Cox pathway, was assayed in an enzyme immunoassay (EIA) kit from R&D systems. The results (Fig. 5c) indicate that RSCL-0520 shows a concentration dependent inhibition of PGE2 release. RSCL-0520 is effective even at 1.25μM enhancing its potential as potent anti-inflammatory molecule. Also, in the absence of AA, we do not see any PGE2 release clearly demonstrating that RSCL-0520 inhibits the PGE2 generated following AA stimulation.
Experiment 9: Effect of RSCL-0520 on LPS induced Nitric oxide (NO) release in RAW 264.7 cells
In murine macrophage RAW 264.7 cells, LPS induces iNOS transcription and transduction, and then the NO production. Furthermore, LPS stimulation is well known to induce IKB proteolysis and NF-κB nuclear translocation. (Freeman & Natanson. Antiinflammatory therapies and sepsis and septic shock. Expert Opin. Invest. Drugs. 2000; 9: 1651-63). Therefore, RAW264.7 cells provide an excellent model for evaluations of potential inhibitors on the pathway leading the induction of iNOS and NO production. Nitric oxide production was determined in RAW 264.7 cells from the American Type Culture Collection (Manassas, VA) cultured in color-free DMEM with standard supplements by measuring the amount of nitrite from cell culture supernatant. RAW264.7 cells (5 x 104 per well) were stimulated for 24hr with or without LPS (250ng/ml) in the absence of presence of the RSCL-0520. Nitrite, a stable end product of NO, was then measured using the Griess reaction (Green LC, Wagner D, Glogowski J, Skipper PL, Wishnok JS, Tannenbaum SR. Analysis of nitrate, nitrite, and [15-N] nitrate in biological fluids. Anal Biochem. 1982; 126: 131—38.). lOOμl of cell culture supernatant was reacted with lOOμl of Griess reagent followed by spectrophotometric measurement at 550nm. Nitrite concentrations in the supernatants were determined by comparison with a sodium nitrite standard curve.
Results (Fig. 6) show that NO secretion induced by LPS stimulation was inhibited by RSCL-0520 in a concentration dependent manner. We have compared our molecule with the existing known strong anti-oxidants (Trolox, Vit E, Catechin and Ascorbic acid) and as is evident from the figure, RSCL-0520 exhibits better anti-oxidant potency than any of them. The reactive free radical NO synthesized by iNOS is a major macrophage derived inflammatory mediator and also has been reported to be involved in the development of inflammatory diseases. (Xie QW, Cho H, Kashiwabara Y, Baum M, Weidner JR, Elliston K, Mumford R, Nathan C. Carboxyl terminus of inducible nitric oxide synthase. Contribution to NADPH binding and enzymatic activity. J Biol Chem. 1994; 269: 28500- 05.). Further, it is also reported that the production of TNF-α is crucial for the synergistic induction of NO synthesis in IFN-g and/or LPS-stimulated macrophages. (Jun CD, Choi BM, Kim HM, Chung HT. Involvement of protein kinase C during taxol-induced activation of murine peritoneal macrophages. J Immunol. 1995; 154:6541-47) Our data corroborates these observations, thus clearly showing that RSCL-0520 has strong anti- inflammatory and anti-oxidant activity.
Experiment 10: Effect of RSCL-0520 on activation of NEMO, degradation of IκB-α and activation of NF-κB.
NF-κB/IκB complexes are present in the cytoplasm under unstimulated conditions. Following stimulation with LPS, we see phosphorylation and subsequent degradation of IKB allowing the free NF-κB to translocate into the nucleus to activate genes with NF-κB binding regions. Therefore, the effect of RSCL-0520 on blocking of NF-κB nuclear translocation was checked. Serum- starved THP-I cells were stimulated with LPS (250ng/ml) for the indicated time (Fig. 7a) in the presence and absence of RSCL-0520 (50μM). RSCL-0520 treatment was lhr prior to LPS treatment. Total protein was isolated from the treated cells, and an equal amount of protein from each sample was used for immunoblots to determine protein levels of NEMO and IκB-α. Blots were stripped and reprobed using ERK- 1/2 antibody to normalize the protein loading. RSCL-0520 blocked signaling to NEMO, possibly blocking phosphorylation of IKK. Which resulted in blocking p65 dissociation from IκB-α (Fig. 7a lanes 6-8). This accumulation of IκB-α lead to inhibition of subsequent down stream signaling pathways.
Further, for the detection of intracellular location of phospho NF-κB p65 subunits, lhr LPS- stimulated cells with and without pretreatment of RSCL-0520 (50μM) Further processing was done as per FACS staining procedure. The cells were then stained with phospho p65 monoclonal antibody tagged with Alexa Fluor 488 (Cell Signaling Technology, Inc, Ma, USA) for lhr at 37°C, followed by washing. Cells were then resuspended in PBS and stained cells were acquired in BD FACS Calibur. Results (Fig. 7b) clearly show its ability to inhibit phospho-p65 in LPS treated cells, substantiating its role in NF-κB signaling inhibition.
The nuclear fractions were obtained from LPS stimulated THP-I cells at the indicated times to evaluate the role of RSCL-0520 in inhibiting p65 subunit translocation into the nucleus. The nuclear proteins electrophoresed were processed for immunoblots using a NF-κB specific antibody. Immunoblotting profile of nuclear extracts for p65 in a time dependent manner (Fig. 6c) clearly shows that RSCL-0520 blocked translocation of NF- KB into the nucleus.
Experiment 11: Effect of RSCL-0520 on Mv 1)88 dependent TLR signaling
Serum - starved THP-I cells were stimulated with LPS (250ng/ml) for lhr in the presence and absence of RSCL-0520. Total RNA was isolated from treated cells post LPS exposure. The cDNA was used for PCR against specific primers for the TLR related genes and β-actin was used as internal control.
With the primary TLR involved identified as TLR4, we have studied the intracellular signaling accessory/adaptor molecules involved in its TLR signaling. Various adapters [myeloid differentiation primary response protein 88(MyD88), Toll receptor IL-IR domain-containing adapter protein (TIRAP), TIR domain-containing adapter inducing IFNβ (TRIF), and Trif-related adapter molecule (TRAM)] and signaling molecules are involved in TLR-4 signaling. (Dunne A, O'Neill LA. Adaptor usage and Toll-like receptor signaling specificity. FEBS Lett. 2005; 579: 3330-35) Further, two signaling pathways; MyD88-dependent and MyD88-independent pathways have been elucidated downstream of TLR2 and 4, with MyD88 dependent pathway shown to be the most predominant one. We have shown that the present invention exerts its inhibitory effect in a MyD88 dependent manner.
All these TLR related genes are upregulated upon stimulation with LPS (Fig. 8a lane 3); while pretreatment with RSCL-0520(Fig. 8a lane 4) inhibits the mRNA expression levels of TIRAP, MyD88, TRAF6, IL- lR-associated kinase 1 (IRAKI) and IRAK4. However, treatment of cells with RSCL-0520 did not show any effect in any of the genes at both mRNA level. These data suggest that RSCL-0520 inhibits MyD88 dependent signaling of TLR4 by LPS.
Immunoblotting further corroborated the mRNA results. Serum - starved THP-I cells were stimulated with LPS (250ng/ml) for the indicated time in the presence and absence of RSCL-0520. RSCL-0520 treatment was lhr prior to LPS treatment. Immunoblotting of total protein was carried out as before to determine protein levels of TIRAP and MyD88. Down regulation of TIRAP and also MyD88 at protein levels (similar to mRNA level) following LPS stimulation was observed in RSCL-0520 pretreated cells suggesting that RSCL-0520 inhibits MyD88 dependent TLR signaling by LPS (Fig. 8b).
EXAMPLE 12: In vivo testing
Experiment 1: Effect of RSCL- 0520 on LPS induced TNF-α release in Balb/c mice
The present invention has studied the ability of RSCL-0520 to exert protection against inflammatory agents in a mice (Balb/c) model. Balb/c (5 - 6 weeks) mice were injected with LPS (225μg) intraperitoneally with and without pretreatment of RSCL-0520 (10 and 20mg/kg). The compound was injected intraperitoneally 30min before LPS treatment. The mice were monitored for lhr post LPS treatment. Untreated mice served as controls. Blood from retro-orbital plexus was collected under anesthesia lhr post LPS injection. Serum collected was analysed for TNF-α secretion using an ELISA detection kit. In untreated mice, LPS injection led to the secretion of large amounts on TNF-α in the serum. However, pretreated mice significantly reduced TNF production (-66% and -70%) Fig. 9a) reconfirming our in- vitro data. *** P value < 0.001 represents LPS treated vs. RSCL-0520 treated. The present invention shows clinical application potential to effectively attenuate LPS mediated TNF release, a key factor in various inflammatory diseases such as arthritis, sepsis and inflammatory bowel disease.
Our results have more significance from the post LPS treatment of RSCL-0520. Here we have stimulated the inflammation process in mice with an injection of LPS (225 μg) intraperitoneally and 30 min post LPS, we have challenged the ability of RSCL-0520 at two different doses (10mg/kg, 20mg/kg) to inhibit the TNF-α secretion. We see a dose dependent inhibition of TNF secretion (-38% and 49% respectively, Fig. 9b.) *** P value < 0.001 represents LPS treated vs. RSCL-0520 treated, NS represents non significant.
Both our ln-vitro and In-vivo results are corroborative and clearly show our molecule to be a promising TLR antagonist and an anti-inflammatory molecule. All of the compositions and methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the methods described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents that are chemically or physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention.

Claims

CLAIMSWe claim:
1. A compound of formula I :
Figure imgf000049_0001
[Formula I]
in which R1, R2, R3, R4, R5, R6, R7, R8, R9 and R10, identical or different, are a hydrogen, a hydroxy, a C1-6 alkoxy, a Ci-6 alkyl, a halogen, a haloalkyl, a acyloxy, a hydroxyalkyl, an alkenyl, an alkenyloxy, a carboxyl, a carbalkoxy, a carbamido, a conjugated group, a substituted or unsubstituted phenyl, a substituted or unsubstituted heterocyclic group, a nitro, an amino, an acylamino, a dialkylamino, a nitric oxide (NO)-releasing moiety, wherein Ring A, Ring B, and Ring C are aliphatic or aromatic and derivatives, and polymorphs, isomers, prodrugs, pharmaceutically acceptable salts, amides and esters thereof.
2. The compound of claim 1, wherein one or more atoms are replaced with an isotope of the atom.
3. The compound of claim 1, selected from the group consisting of: 2, 7-dihydroxy-3 ,4-dimethoxyphenanthrene (RSCL-0518);
2, 7-diacetoxy-3,4-dimethoxyphenanthrene (RSCL 0519);
2,3 ,4,7-tetramethoxyphenanthrene (RSCL-0575);
1 ,5-dihydroxy-2,7-dimethoxy-9, 10-dihydrophenanthrene(RSCL-0521 );
1 ,5-diacetoxy-2,7-dimethoxy-9, 10-dihydrophenanthrene (RSCL-0520); 3,4-dimethoxyphenanthrene-2,7-bis-[(2E)-3-[3,4-bis(acetyloxy)phenyl]acrylate (RSCL-
0522); and l,5-dibenzyloxy-2,7-dimethoxy phenanthrene (RSCL-0638).
4. The compound of claim 1, wherein the compound is isolated from a plant.
5. The compound of claim 4, wherein the compound is Eulophiol (RSCL-0520)
6. A pharmaceutical composition comprising a compound according to claim 1 or 3 or a salt thereof, together with a pharmaceutically acceptable carrier, adjuvant or diluent.
7. The pharmaceutical composition of claim 4, for use in preparing a medicament for prevention or treatment of a disease or condition selected from the group consisting of autoimmunity, inflammation, allergy, asthma, graft rejection, graft- versus-host disease (GvHD), infection, sepsis, cancer, and immunodeficiency.
8. The pharmaceutical composition of claim 7, for use in preparation of medicaments for use in the treatment of conditions involving unwanted immune activity comprising inflammatory or autoimmune disorders.
9. The pharmaceutical composition of claim 4, comprising an effective amount of the compound for modulating immune system activity mediated by Toll-like receptors (TLRs).
10. The pharmaceutical composition of claim 9, comprising an effective amount of the compound sufficient for inhibition of TLR signaling in response to TLR ligand or TLR signaling agonist.
11. The pharmaceutical composition of claims 7 or 9, comprising an effective amount of the compound sufficient for inhibition of TLR signaling under physiological conditions.
12. The pharmaceutical composition of claim 9, comprising an effective amount of the compound sufficient for inhibiting immune stimulation via TLR antagonism.
13. The pharmaceutical composition of claim 4, wherein the compound is selected from RSCL-0518, RSCL-0519, RSCL— 0575, RSCL-0521, RSCL-0520, RSCL-0638, RSCL- 0522, and/or a pharmaceutically acceptable salt, hydrate, solvate, stereoisomer, amorphous solid thereof, or any combination thereof.
14. The pharmaceutical composition of claim 4, wherein the compound is an antagonist of a toll-like receptor (TLR) selected from the group consisting of TLR2, TLR3, TLR4, TLR6, TLR7, TLR8, TLR9 and TLRlO.
15. The pharmaceutical composition of claim 14, wherein the compound selectively inhibits TLR4 over TLR2.
16. The pharmaceutical composition of claim 7, for use in preparation of medicaments for treatment of an autoimmune disorder selected from the group consisting of: systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, Sjogren's syndrome, polymyositis, vasculitis, Wegener's granulomatosis, sarcoidosis, ankylosing spondylitis, Reiter's syndrome, psoriatic arthritis, and Behget's syndrome.
17. The pharmaceutical composition of claim 7, for use in preparation of medicaments for treating a lipopolysaccharide (LPS)-mediated disease selected from the group consisting of inflammatory bowel disease (IBD), sepsis, periodontal disease, mucositis, acne, cardiovascular disease, chronic obstructive pulmonary disease, arthritis, cystic fibrosis, bacterial-induced infections, viral-induced infections, mycoplasma-associated diseases, post herpetic neuralgia, ischemia/reperfusion injury, asthma, stroke, brain injury, necrotizing enterocolitis, bed sores, leprosy, atopic dermatitis, psoriasis, trauma, neurodegenerative disease, amphotericin B-induced fever and nephritis, coronary artery bypass grafting, and atherosclerosis.
18. An unit dose of the pharmaceutical composition of claims 7 or 9, comprising an 0.01 to 100mg/kg of adult body weight.
19. The unit dose of claim 18 wherein the amount of the compound is selected from the group consisting of: 0.1-1.0 mg/kg body weight for inhalable dose, 0.5-10 mg/kg for oral dose, and 0.1-1.0 mg/kg for intravenous dose.
20. An article of manufacture comprising the pharmaceutical composition of claim 4 provided in a form suitable for administration to a patient in need thereof.
21. A formulation comprising (a) the pharmaceutical composition of claim 4 and (b) a second agent affecting non-antigen presenting cells bearing TLRs, in amounts sufficient to exhibit a synergistic effect on TLR-mediated immunostimulation.
22. The formulation of claim 21 , wherein the second agent inhibits the proliferation of T cells.
23. A method of modulating immune system activity mediated by Toll-like receptors (TLRs) comprising contacting a cell expressing a TLR with an effective amount of a compound of Formula I
Figure imgf000052_0001
[Formula I]
in which R1, R2, R3, R4, R5, R6, R7, R8, R9 and R10, identical or different, are a hydrogen, a hydroxy, a C1-6 alkoxy, a C1-6 alkyl, a halogen, a haloalkyl, a acyloxy, a hydroxyalkyl, an alkenyl, an alkenyloxy, a carboxyl, a carbalkoxy, a carbamido, a conjugated group, a substituted or unsubstituted phenyl, a substituted or unsubstituted heterocyclic group, a nitro, an amino, an acylamino, a dialkylamino, a nitric oxide (NO)-releasing moiety, wherein Ring A, Ring B, and Ring C are aliphatic or aromatic and derivatives, and polymorphs, isomers, prodrugs, pharmaceutically acceptable salts, amides and esters thereof.
24. The method of claim 23, wherein the compound is selected from the group consisting of: 2, 7-dihydroxy-3,4-dimethoxyphenanthrene (RSCL-0518); 2, 7-diacetoxy-3,4-dimethoxyphenanthrene (RSCL-0519);
2,3,4,7-tetramethoxyphenanthrene (RSCL-0575);
1 ,5-dihydroxy-2,7-dimethoxy-9, 10-dihydrophenanthrene (RSCL-0521 ); l,5-diacetoxy-2,7-dimethoxy-9,10-dihydrophenanthrene (RSCL 0520);
3,4-dimethoxyphenanthrene-2,7-bis-[(2E)-3-[3,4-bis(acetyloxy)phenyl]acrylate (RSCL-
0522); and l,5-dibenzyloxy-2,7-dimethoxy phenanthrene (RSCL-0638).
25. The method of claim 23, wherein the TLR-mediated immunostimulatory signaling in response to a ligand for the TLR is inhibited.
26. The method of claim 23, wherein the TLR-mediated immunostimulatory signaling in response to an agonist for the TLR is inhibited.
27. The method of claim 23, wherein TLR-mediated immunostimulation in a subject is inhibited.
28. The method of claim 23 or 24, further comprising inhibiting signaling by a Toll-like receptor (TLR) and comprising the step of contacting a cell expressing a functional TLR with an effective amount of the compound.
29. The method of claim 23 or 24, further comprising treating an autoimmune disorder selected from the group consisting of: systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, Sjogren's syndrome, polymyositis, vasculitis, Wegener's granulomatosis, sarcoidosis, ankylosing spondylitis, Reiter's syndrome, psoriatic arthritis, and Behget's syndrome, by administering a therapeutically effective amount of the compound to an individual in need thereof.
30. The method of claim 23 or 24, further comprising treating or preventing a disease or condition selected from the group consisting of autoimmunity, inflammation, allergy, asthma, graft rejection, graft- versus-host disease (GvHD), infection, sepsis, cancer, and immunodeficiency, by administering a therapeutically effective amount of the compound to an individual in need thereof.
31. The method of claim 23 or 24, further comprising treating a lipopolysaccharide (LPS)- mediated disease selected from the group consisting of inflammatory bowel disease (IBD), sepsis, periodontal disease, mucositis, acne, cardiovascular disease, chronic obstructive pulmonary disease, arthritis, cystic fibrosis, bacterial-induced infections, viral-induced infections, mycoplasma-associated diseases, post herpetic neuralgia, ischemia/reperfusion injury, asthma, stroke, brain injury, necrotizing enterocolitis, bed sores, leprosy, atopic dermatitis, psoriasis, trauma, neurodegenerative disease, amphotericin B-induced fever and nephritis, coronary artery bypass grafting, and atherosclerosis, by administering a therapeutically effective amount of the compound to an individual in need thereof.
32. The method of claim 23 or 24, further comprising treating or preventing a clinical condition or disease caused by microbial pathogens, by administering a therapeutically effective amount of the compound to an individual in need thereof.
33. The method of claim 23 or 24, further comprising administering 0.01 to lOOmg of the compound per kg of adult body weight.
34. The method of claim 33 further comprising administering the compound to an individual in need thereof an amount selected from the group consisting of: 0.1-1.0 mg/kg body weight for an inhalable dose, 0.5-10 mg/kg for an oral dose, and 0.1-1.0 mg/kg for an intravenous dose.
35. The method of claim 32, wherein the individual is a veterinary animal.
36. The method of claim 32, wherein the individual is a human.
37. The method of claim 23 or 24, comprising administering the compound by a route selected from oral, intraperitoneal, intramuscular, intravenous, intra-articular, intralesional, subcutaneous, nasal, rectal, buccal, or a route sufficient to provide an amount sufficient to inhibit TLR activity
38. A method for screening agents that inhibit Toll-like receptor activation, the method comprising: contacting a cell expressing a TLR with a candidate agent in the presence of a TLR activator or agonist; determining the effect of the candidate agent on activation of TLR.
39. The method of claim 38, wherein the candidate agent is a compound of Formula I, according to claim 1.
40. The method of claim 38, wherein the candidate agent is a phenanthrene compound.
41. The method of claim 38, further comprising comparing the activity of the candidate agent with an activity of a compound of claim 3.
42. The compounds, compositions, and methods for the preparation and modulation of immune functions mediated through Toll-like receptor (TLR) signaling as claimed above exemplified herein substantially in the examples and figures.
PCT/IN2008/000608 2007-09-24 2008-09-24 Toll like receptor (tlr) signaling antagonist WO2009047791A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
IN1853/MUM/2007 2007-09-24
IN1853MU2007 2007-09-24
IN2541/MUM/2007 2007-12-24
IN2541MU2007 2007-12-24

Publications (2)

Publication Number Publication Date
WO2009047791A2 true WO2009047791A2 (en) 2009-04-16
WO2009047791A3 WO2009047791A3 (en) 2009-08-06

Family

ID=40549710

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IN2008/000608 WO2009047791A2 (en) 2007-09-24 2008-09-24 Toll like receptor (tlr) signaling antagonist

Country Status (2)

Country Link
US (1) US20090215908A1 (en)
WO (1) WO2009047791A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012011100A1 (en) 2010-07-19 2012-01-26 Yeda Research And Development Co. Ltd. Peptides based on the transmembrane domain of a toll-like receptor (tlr) for treatment of tlr-mediated diseases
WO2014111069A1 (en) * 2013-01-17 2014-07-24 USTAV ORGANICKE CHEMIE A BIOCHEMIE AKADEMIE VED CR, v.v.i. Helquat derivatives, preparation thereof, and use thereof as medicaments
WO2019228794A1 (en) * 2018-05-31 2019-12-05 Societe Des Produits Nestle S.A. Direct ampk activator compounds

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010127367A2 (en) * 2009-05-01 2010-11-04 Alfagene Bioscience, Inc. Human gastrointestinal stem cell-derived primary intestinal epithelial cell system and methods of use thereof
CN109516903A (en) * 2017-09-19 2019-03-26 北京理工大学 One kind is for treating the stilbenes compound, preparation method and its application of hepatic injury
CA3144902A1 (en) 2019-08-14 2022-01-19 Andreas Thess Rna combinations and compositions with decreased immunostimulatory properties
TWI830426B (en) * 2022-10-06 2024-01-21 長庚醫療財團法人高雄長庚紀念醫院 Use of toll-like receptor 7 and/or 9 antagonist for treating and/or preventing hearing loss

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3971651A (en) * 1973-02-12 1976-07-27 Rikagaku Kenkyusho Plant growth modifier and a process for preparation thereof
US4434073A (en) * 1981-12-08 1984-02-28 Merck Patent Gesellschaft Mit Beschrankter Haftung Perhydrophenanthrene derivatives, their preparation, and their use in liquid-crystalline dielectrics and electro-optical display elements
US4830664A (en) * 1986-12-23 1989-05-16 Janssen Pharmaceutica N.V. Tricyclic 1h-imidazole-5-carboxylic acid derivatives and use thereof as herbicides
WO2005007672A2 (en) * 2003-06-20 2005-01-27 Coley Pharmaceutical Gmbh Small molecule toll-like receptor (tlr) antagonists
WO2006088716A1 (en) * 2005-02-15 2006-08-24 Eli Lilly And Company Substituted tetralins as selective estrogen receptor-beta agonists
WO2006089881A1 (en) * 2005-02-23 2006-08-31 Pierre Fabre Dermo-Cosmetique Use of phenanthrene derivatives as anti-inflammatory agents, synthesis method and intermediate products
WO2006131466A1 (en) * 2005-06-10 2006-12-14 Ciba Specialty Chemicals Holding Inc. Hydroxyphenyltriazines with an aromatic carbocyclic fused ring system
US20070167409A1 (en) * 2003-10-24 2007-07-19 Eisai Co., Ltd. Compounds and Methods for Treating Toll-Like Receptor 2-Related Diseases and Conditions

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3683091A (en) * 1967-11-13 1972-08-08 Wataru Nagata Certain phenanthrene compounds for treatment of acne
US4866034A (en) * 1982-05-26 1989-09-12 Ribi Immunochem Research Inc. Refined detoxified endotoxin
US4436727A (en) * 1982-05-26 1984-03-13 Ribi Immunochem Research, Inc. Refined detoxified endotoxin product
US4808625A (en) * 1983-05-17 1989-02-28 Burroughs Wellcome Co. Phenanthrene derivatives
US4987237A (en) * 1983-08-26 1991-01-22 Ribi Immunochem Research, Inc. Derivatives of monophosphoryl lipid A
US4877611A (en) * 1986-04-15 1989-10-31 Ribi Immunochem Research Inc. Vaccine containing tumor antigens and adjuvants
US4912094B1 (en) * 1988-06-29 1994-02-15 Ribi Immunochem Research Inc. Modified lipopolysaccharides and process of preparation
DK0472733T3 (en) * 1990-03-06 1995-03-13 Otsuka Pharma Co Ltd Phenanthrend derivatives
EP1478371A4 (en) * 2001-10-12 2007-11-07 Univ Iowa Res Found Methods and products for enhancing immune responses using imidazoquinoline compounds
WO2005016235A2 (en) * 2003-04-14 2005-02-24 The Regents Of The University Of California Combined use of impdh inhibitors with toll-like receptor agonists
US7928132B2 (en) * 2004-08-06 2011-04-19 Ohio University Methods for the amelioration of episodes of acute or chronic ulcerative colitis

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3971651A (en) * 1973-02-12 1976-07-27 Rikagaku Kenkyusho Plant growth modifier and a process for preparation thereof
US4434073A (en) * 1981-12-08 1984-02-28 Merck Patent Gesellschaft Mit Beschrankter Haftung Perhydrophenanthrene derivatives, their preparation, and their use in liquid-crystalline dielectrics and electro-optical display elements
US4830664A (en) * 1986-12-23 1989-05-16 Janssen Pharmaceutica N.V. Tricyclic 1h-imidazole-5-carboxylic acid derivatives and use thereof as herbicides
WO2005007672A2 (en) * 2003-06-20 2005-01-27 Coley Pharmaceutical Gmbh Small molecule toll-like receptor (tlr) antagonists
US20070167409A1 (en) * 2003-10-24 2007-07-19 Eisai Co., Ltd. Compounds and Methods for Treating Toll-Like Receptor 2-Related Diseases and Conditions
WO2006088716A1 (en) * 2005-02-15 2006-08-24 Eli Lilly And Company Substituted tetralins as selective estrogen receptor-beta agonists
WO2006089881A1 (en) * 2005-02-23 2006-08-31 Pierre Fabre Dermo-Cosmetique Use of phenanthrene derivatives as anti-inflammatory agents, synthesis method and intermediate products
WO2006131466A1 (en) * 2005-06-10 2006-12-14 Ciba Specialty Chemicals Holding Inc. Hydroxyphenyltriazines with an aromatic carbocyclic fused ring system

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
CUSHMAN M ET AL: "SYNTHESIS AND EVALUATION OF ANALOGUES OF (Z)-1-(4-METHOXYPHENYL)-2-(3 ,4,5-TRIMETHOXYPHENYL)ETHENE AS POTENTIAL CYTOTOXIC AND ANTIMITOTIC AGENTS" JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, WASHINGTON, US, vol. 35, no. 12, 12 June 1992 (1992-06-12), pages 2293-2306, XP000571677 ISSN: 0022-2623 *
P. TUCHINDA ET AL.: "Phenanthrenes of Eulophia Nuda" PHYTOCHEMISTRY, vol. 27, no. 10, 1988, pages 3267-3271, XP002529778 *
S. ESTRADA; J.J LOPEZ-GUERRERO; R. VILLALOBOS-MOLINA; R. MATA: "Spasmolytic stilbenoids from Maxillaria densa" FITOTERAPIA, vol. 75, 2004, pages 690-695, XP002529775 *
S.R. BHANDARI, A.H. KAPADI: "A 9,10-Dihydrophenanthrene from Tubers of Eulophia Nuda" PHYTOCHEMISTRY, vol. 22, no. 3, 1983, pages 747-748, XP002529774 cited in the application *
S.R. BHANDARI; A.H. KAPADI; P.L. MUJUMDER; M. JOARDAR; J.N. SHOOLERY: "Nudol; a Phenanthrene of the Orchids Eulophia Nuda, Eria Carinata and Eria Stricta" PHYTOCHEMISTRY, vol. 24, no. 4, 1985, pages 801-804, XP002529777 *
SHIMIZU M ET AL: "ANTI-INFLAMMATORY CONSTITUENTS OF TOPICALLY APPLIED CRUDE DRUGS III. CONSTITUENTS AND ANTI-INFLAMMATORY EFFECT OF PARAGUAYAN CRUDE DRUG TAMANDA CUNA CATASETUM-BARBATUM LINDLE" CHEMICAL AND PHARMACEUTICAL BULLETIN, PHARMACEUTICAL SOCIETY OF JAPAN, TOKYO, JP, vol. 36, no. 11, 1 January 1988 (1988-01-01), pages 4447-4452, XP009052878 ISSN: 0009-2363 cited in the application *
Y.-W. LEONG; L.J. HARRISON; A.D. POWELL: "Phenanthrene and other aromatic constituents of Bulbophyllum vaginatum" PHYTOCHEMISTRY, vol. 50, 1999, pages 1237-1241, XP002529776 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012011100A1 (en) 2010-07-19 2012-01-26 Yeda Research And Development Co. Ltd. Peptides based on the transmembrane domain of a toll-like receptor (tlr) for treatment of tlr-mediated diseases
WO2014111069A1 (en) * 2013-01-17 2014-07-24 USTAV ORGANICKE CHEMIE A BIOCHEMIE AKADEMIE VED CR, v.v.i. Helquat derivatives, preparation thereof, and use thereof as medicaments
US9340543B2 (en) 2013-01-17 2016-05-17 USTAV ORGANICKE CHEMIE A BIOCHEMIE AKADEMIE VED CR, v.v.i. Helquat derivatives, preparation thereof, and use thereof as medicaments
WO2019228794A1 (en) * 2018-05-31 2019-12-05 Societe Des Produits Nestle S.A. Direct ampk activator compounds
US11583507B2 (en) 2018-05-31 2023-02-21 Societe Des Produits Nestle S.A. Direct AMPK activator compounds

Also Published As

Publication number Publication date
US20090215908A1 (en) 2009-08-27
WO2009047791A3 (en) 2009-08-06

Similar Documents

Publication Publication Date Title
US20090215908A1 (en) Toll like receptor (tlr) signaling antagonist
KR100840925B1 (en) NOVEL INTERLEUKIN-1 AND TUMOR NECROSIS FACTOR-αMODULATORS, SYNTHESES OF SAID MODULATORS AND METHODS OF USING SAID MODULATORS
US20110014699A1 (en) Prostratin analogs, bryostatin analogs, prodrugs, synthetic methods, and methods of use
Spivak et al. Synthesis and activity of new triphenylphosphonium derivatives of betulin and betulinic acid against Schistosoma mansoni in vitro and in vivo
JP2021513532A (en) Steroidal derivative modulator, its manufacturing method and application
AU2010303822A1 (en) Novel TRPA1 antagonists
AU2006334359A2 (en) Triterpenequinone and triterpenephenol derivatives and their application for the treatment of tumors and parasitic diseases
CN108026141B (en) Ligands for orphan nuclear receptor Nur77 and uses thereof
Idhayadhulla et al. Synthesis of novel and diverse mollugin analogues and their antibacterial and antioxidant activities
JP7123417B2 (en) Anxiolytic deuterium compound and its medicinal use
WO2012171114A1 (en) Synthetic epigallocatechin gallate (egcg) analogs
EP2879669A1 (en) Antagonists of the toll-like receptor 1/2 complex
AU2005281640B2 (en) Novel derivatives of 3,5-seco-4-norcholestane and use thereof
JP2022501445A (en) Terpenoid derivatives and their uses
Wang et al. Garcinol and its analogues: Synthesis, cytotoxic activity and mechanistic investigation
JP4620652B2 (en) Novel polygalatenoside compound and antidepressant containing the same
Misawa et al. Structure–activity relationships of benzhydrol derivatives based on 1′-acetoxychavicol acetate (ACA) and their inhibitory activities on multiple myeloma cell growth via inactivation of the NF-κB pathway
EP2051958A1 (en) Phenyl-prenyl-ether derivatives for the treatment of cognitive, neurodegenerative or neuronal diseases or disorders
WO2007020631A2 (en) Tetracyclic benzofuran derivatives with therapeutic activities
Primahana et al. Synthesis of three quinine fatty acid ester derivatives and its cytotoxic activity evaluation
RU2479582C1 (en) Labdane-type 6-hydroxynaphthoquinones, having cytotoxic activity on human tumour cells
Venditti et al. Trials of phenanthrene opioids functionalization with hindered polycyclic carboxylic acids
AU2012288404A1 (en) Indene derivatives for use in the treatment of inflammatory bowel disease
WO2003099269A1 (en) Cinnamic acid dimers, their preparation and the use thereof for treating neurodegenerative disease
JP2006508098A (en) Therapeutic compounds and methods

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

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

Ref document number: 08836797

Country of ref document: EP

Kind code of ref document: A2

122 Ep: pct application non-entry in european phase

Ref document number: 08836797

Country of ref document: EP

Kind code of ref document: A2