WO2009096905A1 - Method for treating fibrosis and cancer with imidazolium and imidazolinium compounds - Google Patents

Method for treating fibrosis and cancer with imidazolium and imidazolinium compounds Download PDF

Info

Publication number
WO2009096905A1
WO2009096905A1 PCT/SG2009/000037 SG2009000037W WO2009096905A1 WO 2009096905 A1 WO2009096905 A1 WO 2009096905A1 SG 2009000037 W SG2009000037 W SG 2009000037W WO 2009096905 A1 WO2009096905 A1 WO 2009096905A1
Authority
WO
WIPO (PCT)
Prior art keywords
imidazolium
benzyl
methyl
compound
cells
Prior art date
Application number
PCT/SG2009/000037
Other languages
French (fr)
Inventor
Lang Zhuo
Chunyan Zhang
Yugen Zhang
Jackie Y. Ying
Began Gopalan
Zhiyuan Ke
Zhaobing Ding
Original Assignee
Agency For Science, Technology And Research
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Agency For Science, Technology And Research filed Critical Agency For Science, Technology And Research
Priority to EP09706910A priority Critical patent/EP2249832A4/en
Priority to US12/865,668 priority patent/US9072729B2/en
Priority to JP2010544928A priority patent/JP5559701B2/en
Priority to CN2009801120807A priority patent/CN101983057A/en
Publication of WO2009096905A1 publication Critical patent/WO2009096905A1/en
Priority to US14/789,928 priority patent/US20160331727A9/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41881,3-Diazoles condensed with other heterocyclic ring systems, e.g. biotin, sorbinil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/439Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom the ring forming part of a bridged ring system, e.g. quinuclidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • 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
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/56Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring carbon atoms
    • C07D233/58Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring carbon atoms with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/56Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring carbon atoms
    • C07D233/60Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring carbon atoms with hydrocarbon radicals, substituted by oxygen or sulfur atoms, attached to ring nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/56Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring carbon atoms
    • C07D233/61Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring carbon atoms with hydrocarbon radicals, substituted by nitrogen atoms not forming part of a nitro radical, attached to ring nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/06Benzimidazoles; Hydrogenated benzimidazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/22Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains four or more hetero rings

Definitions

  • the present invention relates to methods for delivering an anti-fibrotic or an anti-cancer agent to a cell, including methods for treating fibrotic disease or cancer.
  • HSCs Hepatic stellate cells
  • Hepatic fibrogenesis may be incurred as a result of insults arising from oxidative stress, chemical toxicity or viral infection.
  • fibrotic disease including cell and tissue fibrosis, is among the physiological disorders that have been associated with oxidative stress.
  • oxidative stress resulting from the metabolic generation of ROS has been linked to HSC activation and liver fibrosis (Britton et al. 1994, Tsukamoto et al. 1995, Galli et al. 2005). It has been shown that products of lipid peroxidation lead to increased collagen synthesis by HSCs (Casird et al. 1997 andJParolaet al. 1996).
  • ROS Reactive oxygen species
  • Oxidative stress occurs when there is an excess amount of ROS due to an imbalance between the generation and the elimination or neutralization of these molecules.
  • Excess amounts of ROS may damage cell lipids, proteins and DNA resulting in the inhibition of normal cell function or the development of abnormal cellular behaviour which can in turn lead to a variety of diseases and conditions, depending on the cells affected.
  • ROS have been implicated in many physiological disorders and in the onset and development of a number of diseases.
  • N-acetyl-L- cysteine (NAC), a synthetic precursor of glutathione (GSH) that has been clinically used as an antioxidant, showed anti-fibrogenic properties through the suppression of TGF- ⁇ signalling transduction as well (Meurer et al. 2005).
  • Oxidative stress has also been associated with the onset and progression of cancer. It has been reported that oxidative stress as indicated by reduced anti-oxidant enzyme activities is associated with the deyelopment of primary carcinogenesis and metastasis in clinical patients (VaIi et al. 2008). ROS have been found to cause DNA damage increasing the risk of DNA mutation and thus the development of cancer (Hussain et al. 2005). Tea polyphenols are anti-oxidants and have been shown to inhibit carcinogen-induced DNA damage in animal models of skin, lung, colon, liver and pancreatic cancers (Frei et al. 2003). ⁇ In addition, anti-oxidants PBN and NXY-059 have demonstrated anti-cancer activity in hepatocellular carcinoma. (Floyd 2006). However, the effectiveness of the anti-oxidants investigated remains unclear (Valko et al, 2004).
  • the inflammatory response is the immune system's response to infection or injury and involves the activation of cells of the immune system which produce mediators, such as cytokines, that further activate other cells, leading to a cascade of immune reactions that fight off the infection or repair the injury.
  • mediators such as cytokines
  • inflammatory stresses have been associated with fibrotic disease and cancer (Rakoff-Nahoum 2006; Tsukamoto et al. 1999; Bachem et al. 1992; Vasiliou et al. 2000).
  • HSCs play a critical role. Responding to liver injury, HSCs undergo a process called "activation" and trans-differentiate to myofibroblast- like cells. This process is characterized by phenotypic changes including cell proliferation, over- expression of smooth muscle actin- ⁇ (SMAA), and deposition of extracellular matrix (ECM) proteins, including collagen type ⁇ l (I) (collal) and fibronectin.
  • SMAA smooth muscle actin- ⁇
  • ECM extracellular
  • Inflammatory cytokines represent major mediators for HSC activation.
  • TGF- ⁇ 1 and interleukin 6 (TL-6) have been categorized as profibrogenic cytokines mainly responsible for the induction of ECM proteins (Tsukamoto 1999).
  • HSCs respond to TGF- ⁇ 1 secreted from Kupffer cells and endothelial cells during liver injury, and themselves via autocrine action, resulting in HSC activation and liver fibrosis (Bachem et al. 1992).
  • HSCs can also be attributed to chronic hepatic inflammation through the secretion of proinflammatory cytokine IL-6, leading to cirrhosis (Vasiliou et al. 2000) and hepatocellular carcinoma (Naugler et al, 2007).
  • Transcription factor nuclear factor kappa B (NF- ⁇ B) is an important regulator for the secretion of inflammatory cytokines, and its subunit NF- ⁇ B p65 was reported to mediate liver fibrosis (Vasiliou et al. 2000).
  • TGF- ⁇ is a major mediator for HSC activation and TGF- ⁇ signalling is affected by both oxidative stress and the induction of an inflammatory response (Tsukamoto et al.l999, Bachem et al. 1992, Chen et al. 2002, Nakanuta et al. 2005, Fu et al. 2006, Meurer et al. 2005).
  • NF- ⁇ B Another key molecule in the development of fibrosis, is NF- ⁇ B which is an important regulator of the secretion of inflammatory cytokines but is also known to be sensitive to oxidative stress. Most agents activating NF- ⁇ B are either modulated by ROS or oxidant themselves. It has been reported that treatment with anti-oxidant resveratrol (Chavez et al. 2007) or vitamin E (Liu et al. 1995) attenuated NF-icB elevation induced in carbon tetrachloride experimental fibrotic rodents.
  • Oxidative stress and inflammatory response have also been found to be interrelated in the pathogenesis of cancer. Oxidative stress can cause DNA mutations, some of which will result in the formation of cancer cells. However other mutations will result in cell death, stimulating an inflammatory response. In turn, an inflammatory response will not only provide survival and proliferative signals to cancer cells but may also induce the production of ROS (Rakoff-Nahoum 2006).
  • Dietary anti-oxidants have been widely used as a general approach to ameliorate excessive oxidative stress both in animal models and humans.
  • resveratrol has been shown to extend the lifespan of various species, and to be effective at improving the health and survival of mice on a high-calorie diet (Baur et al. 2006).
  • a method for delivering an anti-fibrotic or anti-cancer agent to a cell comprising contacting the cell with an effective amount of a compound of general formula I or an oligomer or polymer thereof:
  • the dashed line is absent or is present as a bond to form a second bond between the carbon to which R 1 and R 3 are attached and the carbon to which R 2 and R 4 are attached;
  • (i) are each independently H, straight or branched C 1 -C 6 alkyl, straight or branched
  • R 1 and R 5 together with their ring atoms and R 2 and R 6 together with their ring atoms each form a 5- to 10-membered fused saturated, unsaturated or aromatic ring system;
  • R 3 and R 4 are both H, or, when R 1 and R 2 together with their ring atoms form a 6- to 10-membered fused aromatic ring system or when the dashed line is present as a bond, R 3 and R 4 are absent;
  • Ci-C 6 alkyl straight or branched Ci-C 6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C 2 -C 6 alkynyl, C3-C 1 8 cycloalkyl including fused cycloalkyl ring systems, C 6 -CiO aryl, C 6 -Ci O aryl-Ci-C6 alkyl, C 6 -CiO 8TyI-C 2 -C 6 alkenyl, or C 6 -Ci 0 aryl-C 2 -C 6 alkynyl, Ci-C 6 alkyl-C 6 -Cio aryl, C 2 -C 6 alkenyl-C 6 -Ci 0 aryl, or C 2 -C 6 alkynyl-C 6 -Cio aryl;
  • R 7 is H, Ci-C 6 alkyl, phenyl, substituted Ci-C 6 alkyl or halo;
  • any of R 1 to R 7 where applicable, optionally has one or more carbon atoms replaced with " a heteroatom selected from N, O, S and P and is optionally substituted with one or more of straight or branched Ci-C 6 alkyl, straight or branched C 2 -Cg alkenyl, straight or branched C 2 -C 6 alkynyl, C3- Cis cycloalkyl including fused cycloalkyl ring systems, C 6 -Ci O aryl, fluoro, tri-fluoro-methyl, .
  • Ci-C 6 acyloxy Ci-C 6 acyl, carbonyl, amino, acetyl, acetoxy, oxo, nitro, hydroxy.
  • Ci-C 6 alkylcarboxy Ci-C 6 alkoxy, C 2 -C 6 alkenoxy, C 2 -C 6 alkynoxy;
  • the compound is an imidazolium or an imidazolium. In another embodiment, the compound is an imidazolinium or an irnidazolinium salt.
  • R 5 is the same as R 6 .
  • R 5 and R 6 are hydrocarbons.
  • the pharmaceutically acceptable salt of general formula I may be a chloride, bromide, tetrafluoroborate or hexafmorophosphate salt.
  • the compound may be l-ethyl-3-methylimidazolium, 1,3- bisbenzylimidazolium, 1,3-d ⁇ sopropylimidazolium, 1,3-di-tert-butylimidazolinium, l,3-bis(l- adamantyl)imidazolium, 1 ,3 -bis(2,4,6-1iimemylphenyl)-imidazolinium, 1 ,3-bis(2,6-diisopropyl- phenyl)-irnidazoHnium, 1,3-diallylimidazolium, l-benzyl-3-me ⁇ ylimidazolium, l-butyl-3- methylimidazolium, l-(l-adamantyl)-3-(2,4,6-trimethylphenyl)-4,5-dihydroimidazolium, 2- benzyUmidazo[l,5-
  • the compound is a dimer of a compound having a structure of general formula I.
  • the compound may be l,3-dibenzyl-2 ⁇ (l,3 ⁇ dibenzyl-lH-imidazol- 2(3H)-ylidene)-2,3-dihydro- lH-imidazole, (l,2-4,5-d ⁇ imdazottum)-N,N' ,N" , N" ' -tetrabenzyl- benzene, 2,6-di-(3-benzyl-imidazolium)-pyridine or 2,2'-di-(3-benzyl-imidazolium)-l,r- binaphthalene, or a pharmaceutically acceptable salt thereof.
  • the compound is a trimer of a compound having a structure of general formula I.
  • the compound is l,3,5-tris(4-me1hyl-imidazolium)-Mnked cyclophane or a pharmaceutically acceptable salt thereof.
  • the cell may be in vitro.
  • the cell may be in vivo.
  • the method may comprise administering the agent to a subject for the treatment of fibrotic disease or the treatment of cancer.
  • the fibrotic disease may be hepatic fibrosis amd the compound may be for example, 3-d ⁇ sopropylimidazolium or a pharmaceutically acceptable salt thereof.
  • the cancer may be hepatocellular carcinoma, lung cancer, breast cancer, stomach cancer or glioma or the compound may be for example, 3-Bisbenzylimidazolium, 3-dibenzyl-2- methylimidazolium, l-(2,4,6-trimethylphenyl)-3-(4-acetate-benzyl)-imidazo]ium, l-benzyl-3-(4- acetate-benzyl)-2-methyl-imidazolium, l-benzyl-3-(4-methylbenzyl)-imidazolium, l-benzyl-3-(2- trifluorometh ⁇ ylbenzyl)-2-memylimidazolium or l,3-di(2-phenylethyl)-imidazolium, or a pharmaceutically acceptable salt thereof.
  • 3-Bisbenzylimidazolium 3-dibenzyl-2- methylimidazolium, l-(2,4,6-trimethylphenyl)
  • the compound is an imidazolium or an . imidazolium salt. In another embodiment, the compound is an imidazolinium or an imidazolinium salt.
  • R 5 is the same as R 6 .
  • R 5 and R 6 are hydrocarbons.
  • the pharmaceutically acceptable salt of general formula I may be a chloride, bromide, tetrafluoroborate or hexafluorophosphate salt.
  • the compound may be l-ethyl-3- methylimidazolium, 1,3-bisbenzylimidazolium, 1,3-diisopropylimidazolium, 1,3-di-tert- butylimidazolinium, l,3-bis(l-adamantyl)hnidazolium, l,3-bis(2,4,6-trimethylphenyl)- imidazolinium, l,3-bis(2,6-d ⁇ sopropyl-phenyl)-imidazolinium, 1,3-diallylimidazolium, l-benzyl-3- methylimidazolium, l-butyl-3-methylimidazolium, l-(l-adamantyl)-3-(2,4,6-trimethylphenyl)-4,5- dihydroimidazolium, 2-benzylimidazo[l,5- ⁇ ]quinolinium,
  • the compound is a dimer of a compound having a structure of general formula I.
  • the compound may be l,3-dibenzyl-2-(l,3-dibenzyl-lH-imidazol- 2(3H)-y ⁇ dene)-2,3-daydro-lH-imidazole, (l,2-4,5-diimidazolium)-N,N',N'', N'''-tetrabenzyl- benzene, 2,6-di-(3-benzyl-imidazolium)-pyridine or 2,2'-di-(3-benzyl-imidazolium)-l,r ⁇ binaphthalene, or a pharmaceutically acceptable salt thereof.
  • the compound is a trimer of a compound having a structure of general formula I.
  • the compound is l,3,5-tris(4-methyl-imidazolium)-linked cyclophane or a pharmaceutically acceptable salt thereof.
  • the anti-fibrotic agent is delivered for the treatment of fibrotic disease.
  • the fibrotic disease may be hepatic fibrosis amd the compound may be for example, 3-diisopropylimidazolium or a pharmaceutically acceptable salt thereof.
  • the anti-cancer agent is delivered for the treatment of cancer.
  • the cancer s hepatocellular carcinoma, lung cancer, breast cancer, stomach cancer or glioma and the compounds may be, for example, 3- Bisbenzylimidazolium, 3-Dibenzyl-2-methylimidazolium, l-(2,4,6-trimethylphenyl)-3-(4-acetate- benzyl)-imidazolium, l-benzyl-3-(4-acetate-benzyl)-2-methyl-imidaz ⁇ lium, l-benzyl-3-(4- methylbenzyl)-imidazolium, l-benzyl-3-(2-trifluoromethylbenzyl)-2-methylimidazolium or 1,3- di(2-phenylethyl)-imidazolium, or a pharmaceutically acceptable salt thereof.
  • a compound that is an imidazolium or imidazolinium that is l-ethyl-3-methylimidazolium, l-methyl-3-(2-hydroxylethyl)-imidazolium, l-methyl-3-(4- isocynatobenzyl)-imidazolium, l-methyl-3-(4-carboxylbenzyl)-imidazolium, l-methyl-3-(4-acetate- benzyl)-imidazolium, l-methyl-3-(2,2-dimethoxylethyl)-imidazolium, l-(2,4,6-trimethylphenyl)-3- (4-acetate-benzyl)-imidazolium, l,3-Dibenzyl-5-phenylimidazolium, l-benzyl-3-(4- carboxylbenzyl)-2-methylimidazolium, l-benzyl-3-
  • FIG. 1 Cytotoxicity of (a) DBZIM and (b) TDBZIM.
  • HSC T6 cells were seeded at a density of 5000 cells/well in 96-well plate, and cultured for 18-24 h in 10% FBS DMEM before the addition of various compounds (XM-OO ⁇ M) in 10% FBS DMEM for 48 h of proliferation before assaying.
  • the signal was normalized against a vehicle control treatment (0 ⁇ M of compound).
  • the data were obtained from four independent experiments, and presented as mean and standard error or mean (SEM), * a P ⁇ 0.05, * b P ⁇ 0.01 and * C P ⁇ 0.005.
  • FIG.2 (a) DBZIM and (b) TDBZIM attenuated cellular ROS level.
  • HSC T6 cells were incubated with DBZIM (10, 50, 100 and 300 ⁇ M), TDBZIM (10, 50 and 100 ⁇ M), NAC (1 mM) and EGCG (25 ⁇ M) for 48 h before assaying for cellular ROS levels.
  • FIG. 3 Effect of DBZIM and TDBZIM on amount of GSH
  • DBZM attenuated total cellular GSH amount, and (b) TDBZIM did not change the total GSH.
  • DBZM and TDBZM both suppressed cellular GSSG (the oxidation product of GSH).
  • DBZM and (f) TDBZM both enhanced the GSH/GSSG ratio.
  • HSC-T6 cells were incubated with compounds of various concentrations for 48 h, and assayed for the total GSH and GSSG amounts. The GSU/GSSG ratio was calculated as (GSH - GSSG)/GSSG.
  • the GSH and GSSG amounts were normalized against the total protein.
  • TDBZIM suppressed (b) GPx activity and (d) CAT activity, (e) DBZIM and (f) TDBZIM had little effect on SOD activity, (g) DBZIM induced GST activity.
  • HSC T6 cells were treated with compounds of various concentrations for 48 h, and homogenized by sonication (60% frequency for 30 s) in PBS (pH 7.4, 1 mM of EDTA) for GPx, GST and CAT assays, or in 20 mM of HEPES buffer containing 1 mM of EGTA, 210 mM of mannitor and 70 mM of sucrose (pH 7.2) for SOD assay. Enzyme activity was normalized against total protein.
  • DBZIM was dissolved in DMSO, and TDBZIM was dissolved in H 2 O.
  • FIG. 5 DBZIM and TDBZIM protected primary HSC cells against oxidative stress induced by 0.2% (v/v) of DMSO.
  • FIG. 6 (a) DBZIM and (b) TDBZIM suppressed the mRNA expression of HSC activation markers (GFAP and SMAA) and fibrotic endpoints (collal and fibronectin) in a dosage-dependent manner.
  • HSC T6 cells were treated with DBZIM (1, 10, 100 and 300 ⁇ M) and TDBZIM (1, 10 and 100 ⁇ M) for 48 h.
  • ⁇ -actin was used as the normalization gene; its expression level was constant under the experimental conditions.
  • the ⁇ Ct method was used for relative quantification with vehicle control sample being the calibrator.
  • FIG. 7 DBZIM (100 ⁇ M) and TDBZIM (100 ⁇ M) suppressed (a) GFAP, (b) SMAA, (c) collal and (d) fibronectin mRNA expression in a time-dependent manner. HSC T6 cells were treated with DBZIM and TDBZIM, and assayed after 8, 24 and 48 h. A transient elevation of SMAA expression was observed for TDBZIM at 24 h.
  • FIG. 8 Western blotting of collal, fibronectin, GFAP and SMAA protein expression in HSC T6 cells treated with DBZIM (100 ⁇ M) and TDBZIM (50 ⁇ M) for 48 h. 15 ⁇ g of total proteins were resolved in 3-8% of tris-acetate gel for collal and fibronectin, or 4-12% gradient PAGE-SDS gel for GFAP and SMAA. The target proteins were recognized by the respective antibodies, and visualized by the ECL method. The membrane was stripped and re- probed for ⁇ -tubulin as the loading control.
  • FIG. 9 DBZIM and TDBZIM suppressed TGF- ⁇ l mRNA.
  • HSC T6 cells were incubated with (a) DBZM (1, 10, 100 and 300 ⁇ M) and (b) TDBZIM (1, 10 and 100 ⁇ M) for 48 h for the dosage-dependent study, and (c) with DBZIM (100 ⁇ M) and TDBZM (100 ⁇ M) for 8, 24 and 48 h for the time-dependent study.
  • FIG. 10 DBZIM and TDBZIM suppressed JL-6 mRNA.
  • HSC T6 cells were incubated with (a) DBZM (1, 10, 100 and 300 ⁇ M) and (b) TDBZM (1, 10 and 100 ⁇ M) for 48 h for the dosage-dependent study, and (c) with DBZM (100 ⁇ M) and TDBZIM (100 ⁇ M) for 8, 24 and 48 h for the time-dependent study.
  • FIG. 11 Western blotting of NF- ⁇ B and AP-I (c-Fos, JunD and Fra-1) protein expressions in HSC T6 cells treated with DBZIM (100 ⁇ M) and TDBZIM (50 ⁇ M) for 48 h. 15 ⁇ g of nuclear protein was resolved in 4-12% gradient PAGE-SDS gel. The target proteins were recognized by the respective antibodies, and visualized by the ECL method. The membrane was stripped and probed for the Tata binding protein TBP used as the loading control.
  • DBZIM 100 ⁇ M
  • TDBZIM 50 ⁇ M
  • FIG. 12 (a) DBZIM and (b) TDBZIM attenuated GFAP-£ ⁇ cZ transgene as reported by ⁇ -galactosidase activity.
  • Stable transfected T6 GFAP-L ⁇ cZ cells were treated with compounds of 1-300 ⁇ M concentrations, and assayed after 24 h and 48 h.
  • FIG. 13 Western blotting of SMAA, collal, fibronectin, TGF- ⁇ l, TGF ⁇ RI protein expressions in HSC T6 cells treated with various IMSs for 48 h.
  • A DBIM (1 mM), AMM (50 ⁇ M), TMPHM (50 ⁇ M), DPPHM (10 ⁇ M) and DBZBM (100 ⁇ M) were dissolved in DMSO with a final DMSO concentration of 0.2% (v/v).
  • DPM (2 mM), EGCG (25 ⁇ M) and DBZtM (100 ⁇ M) were dissolved in H 2 O.
  • Alpha-tubulin was used as the loading control.
  • Densitometric measurement of protein band intensity was normalized against the respective vehicle control.
  • B Relative densitometric quantification of protein band intensity. The vehicle treated sample was normalized to 1.
  • FIG. 14 Real-time PCR quantification of (a) GFAP, (b) SMAA, (c) fibronectin and (d) collal mRNA expressions in HSC T6 cells treated with various IMSs for 48 h.
  • DBIM (1 mM), AM-M (50 ⁇ M), TMPHIM (50 ⁇ M), DPPHIM (10 ⁇ M) and DBZBIM (100 ⁇ M) were dissolved in DMSO with a final DMSO concentration of 0.2% (v/v).
  • DPIM (2 mM), EGCG (25 ⁇ M) and DBZIM (100 ⁇ M) were dissolved in H 2 O.
  • FIG. 15 Effect of DBZEM and DPIM on liver fibrosis in mice.
  • DBZEM was tested on mice induced by (a) liver toxin thioacetamide (TAA) or by (b)bile duct ligation (BDL) .
  • DPIM was also tested on mice induced by TAA (data not shown)) or by (c) bile duct ligation (BDL).
  • TAA liver toxin thioacetamide
  • BDL bile duct ligation
  • BDL bile duct ligation
  • FIG. 16 Representative of images of Sirius red staining in liver sections from different DPIM compound treated group mice. Each treated group had 6-8 mice, except for 500mg/I treated group, which had only one mouse in the group. Red staining areas represent collagen deposits.
  • A sham operated
  • B sham operated + DPIM lg/1 for 4 weeks
  • C BDL for 4 weeks
  • D BDL 4 weeks + 500mg/l DPIM treatment
  • E BDL 4 weeks + 750mg/l DPIM treatment
  • F BDL 4weeks + lg/1 DPIM treatment.
  • FIG. 17 Percentage of Sirius red staining areas. Sirius red staining areas from different treatment group mice were quantified with Image J software, at least 6 different areas from left and middle lobes of liver of each mouse were covered. * P ⁇ 0.05, BDL vs. DPIM treated groups, # P ⁇ 0.05, 750mg/l vs. lg/1 treated groups.
  • FIG. Liver weights of different treatment groups. Each group consisted of 6-8 mice which were 10-12 weeks old at the start of surgery procedure. *P ⁇ 0.05, sham operated control group vs. BDL 4weeks, ** P ⁇ 0.05, DPIM compound treated groups vs-. BDL weeks group, # P
  • FIG. 10 Representative images of H&E staining of liver sections.
  • A control group;
  • B BDL 4 weeks group; and
  • C BDL 4 weeks +DPIMlg/l treatment group.
  • Figure 20 Quantitative real-time PCR for collagen IaI mRNA. DPIM reduced collagen IaI mRNA.
  • DBZM inhibited HCC cell proliferation. HLE cells were seeded at a density of 5000 cells/well in 96-well plate, and cultured for 18-24 h in 10% EBS DMEM before the addition of various compounds (0-1.25 mM) in 10% FBS DMEM for 48 h of proliferation before assaying. Cell count was calculated based on Hoechst dye staining and BrdU incorporation was based on the total fluorescence intensity of antibody staining.
  • B DBZM arrested HCC cells at G0/G1 phase. HLE cells were incubated with DBZIM (0, 1, 3 mM) for 24 hr before fixation and DAPI staining for DNA content (2N vs. 4N) determination.
  • FIG.22 DBZIM induced caspase 3/7 activity in HLE cells (A) and HepG2 (C) and did not cause LDH releasing in HLE (B) and HepG2 (D). HLE or HepG2 cells were incubated with DBZEVI of various concentrations for 6 hr, and assayed for caspase 3/7 activity and LDH releasing (expressed as % toxicity). DBZIM induced apoptosis as characterized by annexin V staining using flow cytometer in HLE (E) and HepG2 (F). Scatter plot and histogram gave the representative information on the cell population distribution while bar graphs quantified the percentage of apoptotic cells from the histogram. HLE and HepG2 cells were treated by DBZIM of various concentrations for 24 hr before harvesting for annexin " V staining and flow cytometric analysis.
  • FIG. 23 (A). DBZIM did riot change caspase 8 activity and induced caspase 9 and 3 activity in HLE cells. HLE cells were grown in a T75 flask and treated with DBZM for 24 hr. The cells were then lysed and cytoplasmic protein was collected for assaying caspase 8, 9 arid 3 using kits from Biovision. The data was normalized against the vehicle control. (B). DBZM did not cause cytochrome c release from mitochondria to cytosol. HLE cells were treated by DBZM of 1.5 mM for 24 hr. Cytosolic and mitochondria protein fractions were collected using a kit from Biovision. 15. ⁇ g of each protein was resolved in 4-12% gradient PAGE-SDS gel. The target proteins were recognized by antibody of cytochrome C, and visualized by the ECL method. The membrane was stripped and probed for the ⁇ -tubulin as the loading control for cytosolic fraction and Cox4 for mitochondria fraction.
  • FIG. 24 DBZIM affected Bcl-2 and IAP protein expression.
  • He ⁇ G2 cells were treated with DBZIM (0, 0.5, 1.0, 1.5 and 2.0 mM) for 24 hr.
  • Total protein was collected using RIPA buffer. 15 ⁇ g of total protein was resolved in 4-12% gradient PAGE-SDS gel. The target proteins were recognized by respective antibodies, and visualized by the ECL method. The membrane was stripped and probed for the ⁇ -tubulin as the loading control.
  • Figure 25 DBZIM induced nucleus to cytoplasm translocation of survivin protein.
  • FIG. 1 Representative immunocytostaining images of HLE cells treated by DBZIM (0 and 1.0 mM) for 24 hr. Arrows indicate the accumulation and the disappearance of survivin in the nucleus of the control and the DBZIM-treated cells, respectively. The images were acquired and analyzed using ArrayScan VTI HCS reader (Thermal Scientific, PA, USA) and Target Activation BioApplication software.
  • FIG. 1 Representative immunocytostaining images of HLE cells treated by DBZIM (0 and 1.0 mM) for 24 hr. Arrows indicate the accumulation and the disappearance of survivin in the nucleus of the control and the DBZIM-treated cells, respectively. The images were acquired and analyzed using ArrayScan VTI HCS reader (Thermal Scientific, PA, USA) and Target Activation BioApplication software.
  • B Quantitation of translocation event. Cells were grown in 96-well plate and treated by DBZIM of various concentrations for 24 hr before fixation and immuno
  • FIG. 26 DBZIM induced cytoplasm-to-nucleus translocation of AIF.
  • A DBZlM translocated ALF from cytoplasm to nucleus.
  • HepG2 cells were treated by DBZIM for 24 hr.
  • Cytoplasmic and nuclear proteins were fractionated using a kit from Pierce. 15 ⁇ g of proteins were resolved in 4—12% gradient PAGE-SDS gel. The target proteins were recognized by the antibody of AIF, and visualized by the ECL method. The membrane was stripped and re-probed for ⁇ -tubulin as the loading control for cytoplasmic fraction and Tata binding protein TBP for nuclear protein.
  • B HLE cells were grown in 96-well plate and treated by DBZIM for 24 hr.
  • the cells were then fixed and stained by antibody of AIF.
  • the images were acquired and analyzed using ArrayScan VTI HCS reader (Thermal Scientific, PA, USA) and Target Activation BioApplication software.
  • the arrows indicate the cytoplasmic and the nuclear location of the ATF staining in the control and the DBZIM, respectively.
  • C Quantification of AIF staining in different subcellular compartments. Higher value in "Cytoplasmic-Nuclear . '
  • FIG. 27 High dose DBZIM induced ROS production in HLE cells.
  • HLE cells were grown in 96-well plate and treated by DBZIM for 24 hr. The cells were then fixed and stained by DHE for quantification of ROS generation. The images were acquired and analyzed using ArrayScan VTI HCS reader (Thermal Scientific, PA 5 USA) and Target Activation BioApplication software.
  • FIG. 28 DBZIM induced up-regulation of AP-I expression in HepG2 cells.
  • HepG2 cells were treated withDBZM for 24 hr.
  • Cytoplasmic and nuclear proteins were fractionated using a kit from Pierce. 15 ⁇ g of nuclear proteins were resolved in 4-12% gradient PAGE-SDS gel. The target proteins were recognized by their respective antibodies, and visualized by the ECL method. The membrane was stripped and re-probed for Tata binding protein TBP as loading control.
  • Figure 29 DMZIM induced apoptosis. Pictures from phase-contrast microscopy show the apoptotic morphology of gastric cancer cell AGS (A) and lung cancer cell H1299 (B) after exposure to DBZM for 72 h, respectively. Extending the length of exposure caused increased loss of confluence with apoptotic cells in cell cultures.
  • FIG. 30 DBZIM inhibited cell proliferation in glioma cell line C6 (A) and U87 MG (C) and induced caspase 3/7 activity in C6 (B) and U87 MG CD).
  • C6 or U87 cells were incubated with DBZlM of various concentrations for 48 hr for proliferation assay and 6 hr for caspase 3/7 activation assay. Proliferation was measured using MTS Mt from Promega and expressed as relative cell proliferation with control group was normalized to 1. Caspase 3/7 activity was quantified using a kit from Promega and also expressed as relative caspase 3/7 activity of treated groups over control group.
  • FIG. 31 DBZIM induces cleavage of caspase-3, caspase-9 and PARP as indication of apoptosis.
  • FIG. 32 DBZIM inhibits phosphorylation of Akt.
  • AGS, MKN28, H1299 and MCF-7 cells were treated with DBZIM at their corresponding IC50 concentration, phosphorylation at serine 473 of Akt was downregulated in p53 wild type cancer cell line AGS (A), in p53 mutant gastric cancer cell MKN 28 (B), in p53 null Lung cancer cell H1299 (C), and in p53 wild type breast cancer cell MCF-7 (D) .
  • Time course inhibition of phosphorylated-AKT was shown by Western blot, ⁇ -actin was used as an internal loading control.
  • FIG. 33 DBZIM reduced HCC tumor in vivo. Tumors in the HCC-xenografted mice in the control group and in the treatment group, 3 weeks post inoculation, are shown in (A). The tumor growth curve and body weight chart are depicted in (B) and (C), respectively. * P ⁇ 0.01.
  • Figure 34 Endogenous expression of p53.
  • Cell lysates from Hep3B (p53 null), Hep G2 (wild type), HIe and PIc (p53 mutant) were subjected to Western blotting using p53 (DO-I) and actin antibodies.
  • Figure 35 Phase-contrast microscopy of cells exposed to IMSs. Pictures from phase-contrast microscopy show cell cultures after exposure to MSs (IBN 15, IBn 19, IBN 24, IBN 25 and IBN 32) for 72 h, respectively. Extending the length of exposure caused increased loss of confiuency in cell cultures.
  • FIG. 36 IMSs induce apoptosis in Hepatocarcinoma cells.
  • FIG. 37 IMSs arrest Hepatocarcinoma cells. Cells were treated witih vehicle and IBN 15, IBN 19, IBN 24, IBN 25 and IBN 32 for 72 h, and cell cycle distribution was assessed by flow cytometry.
  • FIG. 38 IMSs induce cleavage of caspase-3, caspase-9 and PARP.
  • FIG 39 IMSs-induced apoptosis was accompanied by accumulation of p53 in the nucleus.
  • HLE cells were treated with the indicated dose of IBN 19 and IBN 24 for 48 h. The treated cells were fixed with 4% formaldehyde, immunostained with anti.-p53 (green fluorescence) and DAPI (blue fluorescence), and analysed by confocal microscopy.
  • FIG. 40 IMSs-induced accumulation of p53 and p53 phosphorylation on serines 15, 20, 46, and 392 in p53mutant HLE cells.
  • A Hepatocarcinoma cancer cell HLE were treated with indicated concentration of IBN 15, IBN 19, IBN 24, IBN 25 and IBN 32 for 72h. The levels of phosphorylation of p53 protein and Ser-15, Ser-20, Ser-46 and Ser-392 were determined by Western blotting, ⁇ -actin was used as a loading control.
  • FIG. 41 IMSs-induced apoptosis through the initiation of the mitochondrial pathway.
  • Cells were treated with various IMSs and the levels of Bcl-2 and BAX were assessed by Western blot assay.
  • Figure 42 Real-Time PCR array analysis of p53 signaling pathway-associated gene expression.
  • A Anti-apoptotic genes.
  • B Growth inhibition gene SESN2.
  • C Cell cycle check point genes.
  • D Cell proliferation genes.
  • E DNA repair gene ATM. The relative level of gene expression was normalized with housekeeping gene GAPDH. Values are expressed as mean ⁇ standard deviation.
  • Figure 43 Effect of Compound C (DBZMIM) and compound 9 (MABZMDM) on lung cancer and gastric cancer cells.
  • Compound C (DBZMIM) and compound 9 (MABZM3M) induced morphological change and apoptosis in lung cancer cells MDAMB 231 (A) and in gastric cancer cells MKN 28 (B), 120 h after the treatment.
  • FIG 44 Effect of compound 9on HCC-xenografted mice. Tumors in the HCC- xenografted mice in the control group and in the treatment group, 3 weeks post inoculation, were shown in (A). The tumor growth curve and body weight chart was depicted in (B) and (C), respectively. * P ⁇ 0.01.
  • the methods described herein relate to the discovery that the imidazolium and imidazolinium compounds (collectively "IMSs") as described herein, including in the form of imidazolium and imidazolinium salts, may be used as anti-fibrotic and anti-cancer agents or to treat fibrotic disease or cancer.
  • IMSs imidazolium and imidazolinium compounds
  • the inventors have synthesized novel MSs that may be used in the methods described herein.
  • Both imidazolium and imidazoliniums are based on an imidazole ring and both are N,N' -substituted; imidazoliums are N 5 N' -substituted imidazoles, while imidazoliniums are N,N'- substituted imidazolines and do not have the carbon-carbon double bond between positions C4 and C5 that is present in imidazole.
  • Imidazole itself is incorporated in many biological molecules, and synthetic C-substituted imidazoles have become an important part of many pharmaceuticals (Olmos et al. 1999) (Casanovas et al. 2000).
  • MSs in synthetic chemistry
  • the electronic structure and stability, and thus the therapeutic safety and efficacy of IMSs, can be fine-tuned by varying the N-substituents (substituents on the nitrogen atoms of the central ring) and the central ring of the molecule.
  • the "central ring" of MSs refers to the five membered ring containing 2 nitrogen atoms (either the imidazole or imidazoline ring) to which various substituents may be bound to create different IMS molecules.
  • IMSs provide inexpensive and chemically tunable building blocks for the development of novel therapeutics.
  • NHCs N-heterocyclic carbenes
  • IMSs are precursors of N-heterocyclic carbenes (NHCs).
  • NHCs can be easily generated from IMSs having a hydrogen atom at the C2 position of the central ring and substituents on both nitrogen atoms of the central ring i.e. two N-substituents.
  • NHCs are generated from these IMSs by the deprotonation of the IMS under the appropriate conditions. Deprotonation of IMSs may be carried out under basic conditions or in a diluted solution under neutral conditions.
  • the IMSs as described herein may be used as anti- fibrotic or anti-cancer agents or to treat fibrotic disease or cancer. Without being limited to any particular theory, the IMSs as described herein may exhibit anti-oxidative and anti-inflammatory properties. Both fibrotic disease and cancer have been linked to oxidative stress response. In turn, in both fibrotic disease and cancer, oxidative stress has been shown to be closely linked with inflammatory response.
  • IMSs that may be used as anti-fibrotic agents or as anti-cancer agents are compounds having a structure of the general formula I:
  • the dashed line is absent or is present as a bond to form a second bond between the carbon to which R 1 and R 3 are attached and the carbon to which R 2 and R are attached.
  • the carbon to which R 1 and R 3 are attached and the carbon to which R 2 and R 4 are attached may be connected by either a single bond or a double bond.
  • R 1 and R 2 are each independently H, straight or branched Cj-C 6 alkyl, straight or branched C 2 -C 6 alkenyl, straight or branched C 2 -C 6 alkynyl, C 6 -C 10 aryl; or ( ⁇ ) together with their ring atoms form a 6- to 10-membered fused saturated, unsaturated or aromatic ring system; or ( ⁇ i) R 1 and R 5 together with their ring atoms or R 2 and R 6 together with their ring atoms, form a 5- to 10-membered fused saturated, unsaturated or aromatic ring system and the other of R 1 and R 2 is as defined above in (i); or (iv) R 1 and R 5 together with their ring atoms and R 2 and R 6 together with their ring atoms form 5- to 10-membered fused saturated, unsaturated or aromatic ring system.
  • R 3 and R 4 are both H, or, when R 1 and R 2 together with their ring atoms form a 6- to 10- mmeemmbbeeired fused aromatic ring system or when the dashed line is present as a bond, R and R are absent.
  • R 5 and R 6 are each independently straight or branched Ci-C 6 alkyl, straight or branched C 2 -C 6 alkenyl, straight or branched C 2 -C 6 alkynyl, C 3 -C 1 S cycloalkyl including fused cycloalkyl ring systems, C 6 - C 10 aryl, C 6 -C 10 aryl-Ci-C 6 alkyl, C 6 -C 10 aryl-C 2 -C 6 alkenyl, or C 6 -Ci 0 aryl-C 2 -C 6 alkynyl, C 1 -C 6 alkyl-C 6 -C 10 aryl, C 2 -C 6 alkenyl-C 6 -Cio aryl, or C 2 -C 6 alkynyl-C 6 -C 10 aryl.
  • R 7 may be H, C 1 -C 6 alkyl, phenyl, substituted C 1 -C 6 alkyl or halo.
  • any of the above substituents R 1 to R 7 may optionally have one or more carbon atoms replaced with a heteroatom selected from N, O, S and P.
  • any of the above substituents R 1 to R 7 may optionally be substituted with one or more of straight or branched C 1 -C 6 alkyl, straight or branched C 2 -C 6 alkenyl, straight or branched C 2 -C 6 alkynyl, C 3 -C 1 S cycloalkyl including fused cycloalkyl ring systems, C 6 -C 10 aryl, fluoro, tri-fluoro- methyl, cyanato, isocyanato, carboxyl, Ci-C 6 acyloxy, C 1 -C 6 acyl, carbonyl, amino, acetyl, acetoxy, OXO, nitro, hydroxyl, C 1 -C 6 alkylcarboxy, C 1
  • One of the ring carbon atom to which R 1 and R 5 are attached and the ring carbon to which R 2 and R 6 are attached may be optionally replaced with a nitrogen atom.
  • MSs that may be used in the present methods include pharmaceutically acceptable salts of compounds of formula I and oligomers and polymers of such salts.
  • Such salts have a structure of the general formula II:
  • General formula II is as defined above for general formula I, with the further feature of counterion X.
  • X is a pharmaceutically acceptable anion, including but not limited to chloride, bromide, tetrafluoroborate, hexafluorophosphate.
  • fused refers to the sharing of at least two atoms between ring structures.
  • the ring system is fused to the central imidazolium or imidazolinium ring.
  • the dashed line may be absent or is present as a bond to form a second bond between the carbon to which R 1 and R 3 are attached and the carbon to which R 2 and R 4 are attached;
  • R 1 and R 2 are both H or together with their ring atoms form a 6- to 10- membered fused aromatic ring system;
  • R 3 and R 4 are both H or nothing;
  • R 5 and R 6 are each independently straight or branched C 1 -C 6 alkyl, straight or branched C 2 -C 6 alkenyl, straight or branched C 2 -C 6 alkynyl, C 3 -C 18 cycloalkyl including fused cycloalkyl ring systems, C 6 -C 1 O aryl, C 6 - C 10 aryl-CrC 6 alkyl, C 6 -C 1O aryl-C 2 -C 6 alkenyl or C 6 -C 10 3TyI-C 2 -C 6 alkynyl;
  • the compound may be an imidazolium salt, with a double bond between the two ring atoms to which R 1 and R 2 are attached.
  • the compound may be an imidazolinium salt, wherein R 3 and R 4 are hydrogens and there is only a single bond between their ring carbons.
  • R 3 and R 4 are nothing and R 1 and R 2 together with their ring atoms form a 6- to 10-membered fused aromatic ring system.
  • R 5 and R 6 may also vary.
  • the IMS of the present method may be a compound wherein R 5 and R 6 are the same.
  • R 5 or R 6 may be aralkyls, branched alkyls or cycloalkyls including fused ring systems. Ih other embodiments R 5 or R 6 may comprise a phenalkyl group. In other embodiments, one or both of R 5 and R 6 may comprise an adamantanyl group.
  • IMSs that may be used in the present method also include oligomers or polymers formed from compounds of general formula I.
  • oligomer refers to, for example, 2 or more, 3 or more, 4 or more, 5 or more, 10 or more, 15 or more, 20 or more, 25 or more, 50 or more, 100 or more, 150 or more compounds of general formula I connected together in the manner described below.
  • oligomer includes dimers and trimers.
  • Polymers refers to, for example, 100 or more, 150 or more, 200 or more, 250 or more, 500 or more, 1000 or more compounds of general formula I connected together in the manner described below.
  • oligomers or polymers as used herein, compounds of general formula I are connected to each other to form a macromolecular structure, which may be cyclised.
  • trimers as described herein include 1 ,3 ,5-tris(4-methyl-imidazolium)- linked cyclophane or a pharmaceutically acceptable salt thereof (TDBZIM).
  • two compounds of general formula I may be connected together so that one or more R 1 , R 2 , R 5 , R 6 or R 7 substituent is bivalent and is thus shared between the two compounds, rather than have a relevant substituent present in the oligomer or polymer for every compound of general formula I present.
  • R 1 , R 2 , R 5 , R 6 or R 7 substituent is bivalent and is thus shared between the two compounds, rather than have a relevant substituent present in the oligomer or polymer for every compound of general formula I present.
  • two compounds may be connected by a shared bivalent R 1 substituent, and thus in the complete dimer, there is only one occurrence of an R 1 substituent for the two compounds of general formula I.
  • the compounds of general formula I may be j oined to each other via a bivalent substituent such as R 1 , R 2 , R 5 , R 6 , or R 7 .
  • a bivalent substituent such as R 1 , R 2 , R 5 , R 6 , or R 7 .
  • an R 5 substituent on one compound of general formula I may also be R 5 on a second compound of general formula I such that the two compounds are linked via a shared R 5 substituent.
  • oligmers or polymers may be formed by linking compounds via shared R 1 , R 2 , R 5 , R 6 , or R 7 substituents.
  • dimers of this description include 2,6-di-(3-benzyl-imidazolium bromide)-pyridine or a pharmaceutically acceptable salt thereof (IBN-22).
  • Sharing of substituents as described above can include sharing of two substituents that together with their ring carbons form a fused ring system.
  • additional carbons within the ring will be ring carbons from a second compound of general formula I.
  • dimers of this description include benzo(l,2-4,5-diimidazolium)-N,N',N'', N"'- tetrabenzyl-di-bromide or a pharmaceutically acceptable salt thereof (IBN-29).
  • a substituent such as one or more of R 1 , R 2 , R 5 , R 6 or R 7 may be bonded to another such substituent on a second compound in order to connect two compounds together to form an oligomer or polymer, and thus there will be one occurrence of the relevant Unking substituent for each compound of general formula I in the oligomer or polymer.
  • Multiple compounds falling within general formula I may be joined together (oligomerised) in this way, by one or more shared bivalent R 1 , R 2 , R 5 , R 6 or R 7 substituents.
  • such dimers include 2,2' -di-(3 -benzyl-imidazoHum bromide)- 1,1' -binaphthalene or a pharmaceutically acceptable salt thereof (IBN-23).
  • two compounds of general formula I may be attached by a single or double bond between respective carbon atoms to which R 7 is attached in general formula I to form a dimer.
  • These molecules include for example, l,3-dibenzyl-2-(l,3-dibenzyl-lH-imidazol-2(3H)- ylidene)-2,3-dib.ydro-lH-imidazole or a pharmaceutically acceptable salt thereof (compound H).
  • the oligomerised or polymerised compounds may be the same compound, such that all substituents in general formula I are the same in each oligomerised or polymerised compound, or may be different, with one or more substituents differing between compounds, with the exception that shared bivalent substituent or substituents will obviously be the same in the compounds in which the bivalent substituent is shared.
  • Table 1 contains the names and structures of particular examples of the IMSs described herein.
  • the compounds of the present invention may be 1-ethyl- 3-methylimidazolium, 1,3-bisbenzylimidazolium, 1,3-d ⁇ sopropylimidazolium, 1,3-di-tert- butylimidazolinium, l,3-bis(l-adamantyl)imidazolium, l,3-bis(2,4,6-trimethylphenyl)- imidazolinium, l,3-bis(2,6-dusopropyl-phenyl)-imidazolMum, 1,3-diallylimidazolium, l-benzyl-3- methylimidazolium, l-butyl-3-methylimidazolium, l-(l-adamantyl)-3-(2,4,6-trimethylphenyl)-4,5- dihydroimidazolium, 2-benzylimidazo[l,5- ⁇ 3quinoIinium, l,3-
  • the compounds of the present invention may be 1-ethyl- 3-methylimidazolium bromide, 1,3-bisbenzylimidazolium bromide (DBZIM), 1,3- d ⁇ sopropylimidazolium tetrafluoroborate (DPIM), 1,3-di-tert-butylimidazoliniumtetrafluoroborate (DBIM), l,3-bis(l-adamantyl)imidazolium tetrafluoroborate (AMIM), l,3-bis(2,4,6- • ttimemylphenyl)-imidazolimum chloride (TMPHIM), l,3-bis(2,6-diisopropyl-phenyl)- imidazolinium chloride (DPPHIM), 1,3-diallylimidazolium bromide (Compound D), l-benzyl-3- methylimidazoKum bromide (Compound E),
  • the compounds of general formula I include novel compounds, including l-ethyl-3- methylimidazolium, l-methyl-3-(2-hydroxylethyl)-imidazolium,. l-methyl-3-(4-isocynatobenzyl)- imidazolium, l-methyl-3-(4-carboxylbenzyl)-miidazolium, l-methyl-3-(4-acetate-benzyl)- imidazolium, l-methyl-3-(2,2-dimethoxylethyl)-imidazolium, l-(2,4,6-trimethylphenyl)-3-(4- acetate-benzyl)-i ⁇ udazolium, l,3-Dibenzyl-5-phenylimidazolium, l-benzyl-3-(4-carboxylbenzyl)-2- methylimidazolium, l-benzyl-3-(3,4,5-tri
  • the compounds of general formula I include novel compounds, including l-ethyl-3- methylimidazolium bromide, l-methyl-3-(2-hydroxylethyl)-rmidazolium bromide (IBN-2), 1- methyl-3-(4-isocynatobenzyl)-imidazolium chloride (IBN-3), l-methyl-3-(4-carboxylbenzyl)- imidazolium bromide (IBN-4), l-metiiyl-3-(4-acetate-benzyl)-imidazolium chloride (IBN-6), 1- methyl-3-(2,2-dimethoxylethyl)-imidazolium bromide (IBN-8), l-(2,4,6-trimethylphenyl)-3-(4- acetate-benzyl)-imidazolium chloride (IBN-9), l,3-Dibenzyl-5-phenylimidazolium bromide (IBN-9),
  • a method for delivering an anti-fibrotic or anticancer agent to a cell comprising contacting the cell with an effective amount of a compound of general formula I, a pharmaceutically acceptable salt thereof or an oligomer or polymer thereof.
  • delivering an anti-fibrotic or anti-cancer agent to a cell refers to providing the agent in sufficiently close proximity to the cell such that the agent can exert its anti- fibrotic or anti-cancer effects on the cell.
  • the agent may be delivered to the cell by adding the agent to the cell culture media.
  • the agent may be delivered by administering the agent to a subject as a pharmaceutical composition.
  • contacting refers to direct and indirect contact with the cell.
  • Direct contact refers to a direct interaction between the agent and the cell.
  • indirect contact involves "contacting the cell” via interactions with other molecules or compounds.
  • the agent may interact with a molecule, affecting a change in that molecule that causes it to interact with the cell or interact with another molecule which will then interact with the cell to exert an effect.
  • Indirect contact may involve a series of molecular interactions resulting in an effect on the cell.
  • An anti-fibrotic agent as used herein refers to a compound that alters, reduces or inhibits molecules, mechanisms or effects associated with fibrotic disease.
  • the agent may be an anti-inflammatory or an anti-oxidative, induce apoptosis, alter, reduce or inhibit the activity of fibrogenic mediators such as TGF- ⁇ , NF- ⁇ B or TL-6 or neutralize proliferative, fibrogenic, contractile or pro-inflammatory responses of cells.
  • the anti-fibrotic agent may reduce or inhibit fibrotic disease by: (a) reducing inflammation to avoid stimulating activation of cells involved in fibrotic disease such as stellate cells, (b) directly down-regulating activation of cells involved in fibrotic disease, (c) neutralizing proliferative, fibrogenic, contractile or pro-inflammatory responses of cells involved in fibrotic disease, (d) inducing apoptosis such as, for example, Bcl-xL or Fas; or (e) inducing ECM degradation.
  • An anti-cancer agent as used herein refers to a compound that alters, reduces or inhibits molecules, mechanisms or effects associated with cancer.
  • the agent may be an anti-inflammatory or an anti-oxidative, induce apoptosis, inhibit abnormal cell growth, inhibit cell proliferation, inhibit DNA mutation or modify the activity or level of activity of molecules associated with cancer such as survivin, caspase 9, caspase 3, BCI-X L , Bak, BAX, ATM, p53, Cdc25A, Cdc2, SESN2 and AP-I .
  • the cell to which the anti-fibrotic or anti-cancer agent is to be provided may be any cell, including an in vitro cell, a cell in culture, or an in vivo cell within a subject.
  • the term "cell” as used herein refers to and includes a single cell, a plurality of cells or a population of cells where context permits, unless otherwise specified.
  • the cell may be an in vitro cell including a cell explanted from a subject or it may be an in vivo cell in a subject.
  • reference to “cells” also includes reference to a single cell where context permits, unless otherwise specified.
  • the cell may be derived from any organism, for example an insect, a microorganism including a bacterium, or an animal including a mammal including a human.
  • the cell of the present method may be within a subject having fibrotic disease or cancer, a subject requiring treatment for fibrotic disease or cancer or a subject in which prevention of fibrotic disease or cancer is desired.
  • the subject is a human subject.
  • Fibrotic disease will be understood by those skilled in the art to refer to a condition which may be characterized or caused by the development of excess fibrous tissue or over production of extracellular matrix in an- organ or tissue and may include for example liver fibrosis, kidney fibrosis, lung fibrosis, bone fibrosis., systemic sclerosis, mixed connective tissue. Father examples of fibrotic disease are provided in US 2007/0043016 which is herein incoporated by reference.
  • an anti-fibrotic agent may be delivered to a cell in a subject having hepatic fibrosis.
  • the compound may be for example 1,3- diisopropylimidazolium or a pharmaceutically acceptable salt thereof.
  • cancer to encompass a class of diseases in which cells exhibit abnormal cell growth and the potential to invade nearby tissues.
  • the abnormal cells may also spread to other locations in the body.
  • Different types of cancer include for example, breast cancer, colorectal cancer, brain cancer, prostate cancer, cervical cancer, ovarian cancer, bone cancer, skin cancer, lung cancer, pancreatic cancer, bladder cancer, gallbladder cancer, kidney cancer, esophageal cancer, Hodgkin lymphoma, Non-Hodgkin lymphoma, laryngeal cancer, leukemia, multiple myeloma, oral cancer, pleural mesothelioma, small intestine cancer, testicular cancer, uterine cancer, thyroid cancer and stomach cancer.
  • an anti-cancer agent may be deliverd to a cell in a subject having hepatocellular carcinoma, lung cancer, breast cancer, stomach cancer or glioma.
  • the compound may be for example 1,3-Bisbenzylimidazolium, 1, 3,-Dibenzyl-2- methylimidazoHum,l-(2,4,6-trimethylphenyl)-3-(4-acetate-benzyl)-m ⁇ idazoHum, l,3-Dibenzyl-5- phenylimidazolium , l-benzyl-3-(4-acetate-benzyl)-2-methyl-imidazolium, l-benzyl-3-(4- methylbenzyl)-imidazolium, l-benzyl-3-(2-trifluoromemylbenzyl)-2-methylimidazolium or 1,3- di(2-phenylethyl)-imidazolium, or a pharmaceutically acceptable
  • an effective amount means an amount effective, at dosages and periods of time necessary to achieve the desired result, for example to treat fibrotic disease or cancer.
  • the total amount of IMS to be administered will vary, depending on several factors, including the severity and type of the disorder, the mode of administration, and the age and health of the subject. Methods for determining an effective amount of a particular IMS for treating fibrotic disease or cancer will be readily apparent to a person skilled in the art.
  • Treating" fibrotic disease or cancer refers to an approach for obtaining beneficial or desired results, including clinical results.
  • Beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disorder or disease, stabilization of the state of disease, prevention of development of disorder or disease, prevention of spread of disorder or disease, delay or slowing of disorder or disease progression, delay or slowing of disorder or disease onset, amelioration or palliation of the disorder or disease state, and remission (whether partial or total).
  • Treating can also mean prolonging survival of a subject beyond that expected in the absence of treatment.
  • Treating can also mean inhibiting the progression of disorder or disease, slowing the progression of disorder or disease temporarily, although more preferably, it involves halting the progression of title disorder or disease permanently.
  • the IMS may be formulated as an ingredient in a pharmaceutical composition.
  • the compositions may contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives and various compatible carriers or diluents.
  • the proportion and identity of the pharmaceutically acceptable carrier is dependant on a variety of factors including the chosen route of administration, compatibility with the IMS molecule and standard pharmaceutical practice. Generally, the pharmaceutical composition will be formulated with components that will not significantly impair the biological properties of the IMS.
  • the pharmaceutical composition may be administered to a subject in a variety of forms depending on the selected route of administration, as will be understood by those skilled in the art.
  • the composition of the invention may be administered for example, by oral administration, surgically or by injection to the desired site.
  • the composition is administered by injection (subcutaneously, intravenously, intramuscularly, etc.) directly at a desired site, for example in the vicinity of fibrotic disease or cancer that is to be treated.
  • the dose of the pharmaceutical composition that is to be used depends on the particular fibrotic disease or cancer disorder being treated, the severity of the condition, individual patient parameters including age, physical condition, size and weight, the duration of the treatment, the nature of concurrent therapy (if any), the specific route of administration and other similar factors that are within the knowledge and expertise of the health practitioner. These factors are known to those of skill in the art and can be addressed with minimal routine experimentation.
  • compositions may be provided in a variety of dosage forms and thus, in different embodiments, IMSs may be administered in different dosage forms including for example pills, tablets, capsules, solutions, suspensions, powder and injections.
  • Conventional procedures and ingredients for preparing and administering the different dosage forms would be known to a skilled person and are described for example, in Remington's Pharmaceutical Sciences (Remington, The Science and Practice of Pharmacy, 21 st edition, Lippincott Williams & Wilkins, Philadelphia, PA., 2006).
  • IMSs as described herein, including compounds having a structure of general formula I, a pharmaceutically acceptable salt thereof or an oligomer or polymer thereof for delivering an anti-fibrotic or anti-cancer agent to a cell in vivo and in the preparation of a medicament for delivering an anti-fibrotic or anti-cancer agent to a cell in vivo are also contemplated.
  • the IMSs of the present method may exert their anti-fibrotic and anti-cancer effect as a result of anti-oxidative properties, which may function via a multitude of molecular mechanisms.
  • Anti-oxidarits generally exert their effect mainly through three different pathways: (1) neutralization of cellular free radical ROS generated during metabolism and immune response, (2) induction of endogenous anti-oxidative enzymatic activity, and (3) chelation of iron or copper ions that catalyze the generation of hydroxyl radical.
  • the anti-oxidative properties of the IMSs may result, at least in part, due to their activity as a radical scavenger.
  • IMSs may occur after a- series of chemical reactions including (1) the spontaneous conversion of MSs to NHCs preferably under a basic condition, (2) the interaction of the NHCs at the carbon 2-position with free radicals such as ROS resulting in the formation of intermediate active radicals and the neutralization of the free radicals.
  • the IMSs described herein also exhibit anti-inflammatory properties.
  • the dual anti-oxidative and anti-inflammatory properties of the IMSs may be attributable, at least in part, to the interplay between oxidative stress and inflammation.
  • the anti-inflammatory effects of the IMSs compounds defined herein may be attributable to the IMSs' anti-oxidative properties that inhibit HSC activation and thus the resulting . production of pro-inflammatory cytokines.
  • the inhibition of inflammatory- mediator IL-6 by an IMS may be partially attributable to the anti- oxidative properties of the IMS which suppress the activation of transcription factor NF- ⁇ B, an important regulator of the secretion of inflammatory cytokines.
  • the IMSs may exhibit other effects that are useful in treating fibrotic disease and cancer.
  • Anti-oxidants have been found to also have apoptosis-inducing properties.
  • EGCG a green tea extract has shown to induce apoptosis of human hepatocellular carcinoma in cultured cell lines and a xenograft model (Nishikawa et al. 2006) while garlic extract, a natural anti-oxidant, has also been shown to induce apoptosis in human glioblastoma cells (Das et al. 2007) and colon carcinoma cells (Jakubilova et al. 2006) by inducing oxidative stress.
  • the MSs may be effective in treating cancer due to inducement of apoptosis and inhibition of cell proliferation through cell cycle arrest.
  • IMSs may trigger apoptosis and cell cycle arrest by increasing or decreasing the activity and expression of molecules involved in apoptosis and cell cycling including for example survivin, caspase 9, caspase 3, BCI-X L , Bak, BAX, ATM, p53, Cdc25A, Cdc2, SESN2 and AP-I.
  • DBZM, TDBZM and DBZBM were synthesized in-house.
  • DPM, DBM, AMIM, TMPHM and DPPHIM were purchased from Sigma Chemicals (USA).
  • the chemical structures of DBZM (1,3-bisbenzylimidazolium bromide), DBZBM (1,3-bisbenzyl-benzimidazolium bromide) and TDBZM (l,3,5-tris(4-methyl-imidazolium)-Unked cyclophane-3Br) are illustrated in Table 1.
  • DBZM and DBZBM were synthesized using a method disclosed in the literature (Harlow et al. 1996).
  • TDBZIM was synthesized by mixing 4-methylimidazole (123 mg, 1.5 mmol) and 2,4,6- tris(bromomethyl)mesitylene (400 mg, 1 mmol) in 200 ml of N,N'-dimethylformamide (DMF) in a reaction vial. The reaction mixture was heated to 100 0 C for 2 days. Colorless crystals of TDBZIM were precipitated, and collected in 40% yield (160 mg).
  • the stellate cell band was harvested, washed, and IxIO 6 cells/well were seeded in a 6-well culture plate in Dulbecco modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS).
  • DMEM Dulbecco modified Eagle's medium
  • FBS fetal bovine serum
  • the viability of the HSCs was > 95% as determined by trypan blue exclusion staining, and the purity of the HSCs was > 90% as assessed by glial fibrillary acidic protein (GFAP) positive staining in > 200 cells counted.
  • GFAP glial fibrillary acidic protein
  • HSCs Primary HSCs were routinely cultured in DMEM with 10% FBS in a humidified CO 2 incubator at 37°C, and split at 1:4 ratio by trypsinization (0.05% trypsin/0.53 mM ethylenediaminetetraacetic acid (EDTA)) when they grew to confluency.
  • the HSC-T6 cell line was kindly provided by Dr. Scott Friedman of Mount Sinai School of Medicine, New York.
  • a stable T6/GFAP-L ⁇ cZ clonal cell line was established as previously reported (Maubach et al.2006). HSC-T6 and T6/GFAP-Z, ⁇ cZ cells were cultured in the same conditions as the primary HSCs.
  • the ROS level of treated HSC-T6 cells was determined using the dichlorofluorescein (DCF) labeling method (Molecular Probes Inc., OR, USA). Cultured cells were harvested by trypsinization, and resuspended in phenol red-free DMEM Qhvitrogen). Cells were then incubated with 10 ⁇ g/ml of 2' ,7'-dichloro-fluorescein diacetate (DCFH-DA) in DMEM for 15 min before resuspension in DMEM for assaying.
  • DCFH-DA 2' ,7'-dichloro-fluorescein diacetate
  • Total cellular GSH and GSSG levels were determined by using assay kits from Cayman Chemical (Ann Arbor, MI, USA) following the manufacturer's instructions. Briefly, HSC- T6 cells treated with DBZIM, TDBZIM, NAC, or EGCG were scalped and homogenized in phosphate buffered saline (PBS) by sonication at 60% frequency for 30 s, and the supernatant was collected after centrifugation at 16,000xg for 10 min at 4 0 C. Protein content of the samples was determined by bicinchonicic acid(BCA) assay.
  • PBS phosphate buffered saline
  • GSH assays the protein sample was deproteinated by metaphosphoric acid (Aldrich, Catalog No. 23927-5), and the pH was adjusted by triethanolamine (Aldrich, Catalog No. T5830-0) according to the manual. The sample was ready for assaying total GSH including both the reduced and oxidized forms. Quantification of GSSG 5 exclusive of GSH, was performed by first derivatizing GSH with 2-vinylpyridine and assaying separately. Colorimetric signal was recorded at 405 nm with a Tecan Safire ⁇ plate reader. The levels of GSH or GSSG were determined as nmol/ ⁇ g protein.
  • Glutathione Peroxidase GPx
  • Catalase CAT
  • Superoxide Dimutase SOD
  • Activity levels of three anti-oxidant enzymes were determined using assay kits from Cayman Chemical.
  • protein samples were harvested in ice-cold PBS containing 1 mM of EDTA.
  • SOD activity protein samples were prepared in 20 mM of N-(2-hydroxyethyl)-piperazine-N'-2-ethanesulfonic acid (HEPES) buffer containing 1 mM of ethylene glycol-bis(2-aminoethylether)-N,N,N',N'-tetraacetic acid (EGTA), 210 mM of mannitol and 70 mM of sucrose. Protein content was determined by BCA assay.
  • HEPES N-(2-hydroxyethyl)-piperazine-N'-2-ethanesulfonic acid
  • GPx activity was indirectly measured by a coupled reaction with GSH reductase.
  • Oxidized GSH produced upon reduction of H 2 O 2 by GPx, is recycled to its reduced state by glutathione reductase (GR) and nicotinamide adenine dinucleotide phosphate ( ⁇ ADPH).
  • the oxidation of ⁇ APDH is proportional to the GPx activity, and can be measured as a decrease in absorbance at 340 nm.
  • GPx activity was expressed as nmol/min/mg protein.
  • CAT activity was determined based on reaction of the enzyme with methanol in the presence of H 2 O 2 . The formaldehyde produced was measured spectrophotometrically at 540 nm using Purpald as the chromogen. CAT activity was expressed as nmol/rnin/mg protein.
  • Total SOD including all three types of SOD (Cu/Zn-, Mn- and Fe 7 SOD), was quantified using a tetrazolium salt to detect superoxide radicals generated by xanthine oxidase and hyposanthine.
  • One unit of SOD was defined as the amount of enzyme needed to exhibit 50% dismutation of the superoxide radical.
  • SOD activity was expressed as U/mg protein.
  • phase ⁇ anti-oxidant enzyme GST
  • the activity of phase ⁇ anti-oxidant enzyme, GST was assayed using a kit from Cayman Chemical. Protein samples were harvested in PBS buffer containing 1 mM of EDTA, and the protein concentration was determined by BCA assay. Total GST (cytosolic and microsomal) activity was assayed by measuring the conjugation of l-chloro-2,4-dinitrobenzene (CDNB) with reduced GSH 5 and monitored by an increase in absorbance at 340 nm. GST activity was expressed as nmol/min/mg protein.
  • CDNB l-chloro-2,4-dinitrobenzene
  • Primers and probes for rat GFAP, SMAA, collagen IaI, fibronectin, TGF- ⁇ l, TGF- ⁇ receptor I (TGF ⁇ RI) and TL-6 were ordered from Taqman's assay-on-demand database.
  • Rat ⁇ -actin was used as normalization gene, and its expression level was constant (Or difference was ⁇ ⁇ 0.5) throughout the experimental settings in this study.
  • Relative quantification of target mRNA was calculated using comparative threshold cycle method (AACi) as described in User Bulletin #2 (ABI Prism 7700 Sequence Detection System). Relative quantification was given by 2 " MCT to express the up-regulation or down regulation of the target gene under the treatments compared ' to the control.
  • Protein extracts were prepared by lysing a cell pellet in 10 volumes of boiled 1 % sodium dodecyl sulfate (SDS) (90-95 0 C) for 10 min for GFAP, or using NE-PER nuclear and cytoplasmic extraction reagents from Pierce (IL, USA) for nuclear proteins, or RIPA (Pierce, IL, USA) for SMAA, fibronectin, collagen ⁇ I(T), TGF- ⁇ l and TGF ⁇ RI. Protein content was quantified using a BCA kit (Pierce, IL, USA). 15 ⁇ g of total protein were resolved in 4-12% gradient SDS/polyacrylamide gel electrophoresis (PAGE) gel, and transferred to a nitrocellular membrane.
  • SDS sodium dodecyl sulfate
  • the membrane was then probed with primary antibodies against GFAP (Dako, Denmark), SMAA (Sigma, USA), procollagen ⁇ l(l), fibronectin, c-Jun, c-Fos, JunB, JunD, Fra-1, Fra-2 (Santa Cruz Biotechnology, CA, USA), NF-TCB P65, TGF- ⁇ l and TGF ⁇ RI (Abeam, UK), and detected by horseradish peroxidase-conjugated secondary antibody (Santa Cruz Biotechnology, CA, USA). For fibronectin and collal blotting, the protein extract was separated in 3-8% tris-acetate gel.
  • Protein bands were recorded on X-ray film by reacting with ECL chemiluminescence reagents (Amersham Biosciences, NJ, USA).
  • Alpha-tubulin (Abeam, UK) was used as the loading control for total protein
  • Tata binding protein (TBP) (Abeam, UK) was used as the loading control for nuclear protein.
  • Beta-galactosidase activity in extracts from T6/GFAP-fccZ cells treated by DBZIM or TDBZIM was measured using a chemiluminescent assay kit (Roche, Mannheim, Germany) in a 96-well plate format, according to the manufacturer's instructions. Briefly, cells were washed twice with ice-cold Ix PBS (pH 7.4), and lysed for 30 min at room temperature in the lysis buffer. The supernatant was collected for assays with a Tecan Safire ⁇ plate reader. Protein concentration was determined with a BCA protein assay Mt (Pierce, EL, USA). Specific ⁇ -galactosidase activity was obtained by normalizing against total cellular protein content.
  • IMSs DPIM and DBZIM were dissolved in saline, and administered to Friend virus.
  • B-Type mice mice (weighing ⁇ 25-30 g) intraperitoneally every other day for 2 weeks. The dosages were 0, 200, 300, 350, 400 and 500 mg/kg for DPIM, and 0, 10, 30, 35, 40 and 55 mg/kg for DBZIM. The injection volume was maintained at 100-160 ⁇ l. The mortality rate of mice was assessed after the treatment. The animal experimental protocol was approved by the Institutional Animal Care and Use Committee (IACUC) of Biological Resource Center (BRC) at Biopolis, Singapore.
  • IACUC Institutional Animal Care and Use Committee
  • TAA liver toxin thioacetamide
  • BDL bile duct ligation model
  • Ketamine 150 mg/kg
  • Xylazine 10 mg/kg
  • BDL bile duct ligation model
  • An abdominal incision at the midline is made to expose the common bile conduct, which was then ligated using 5-0 silk suture.
  • the bile duct is exposed, but not ligated.
  • DBZIM or DPIM was provided to the mice in drinking water at concentrations from 1 to 1000 mg/liter.
  • the collagen content in the fibrotic liver was assessed by the Sirius Red staining of the liver tissue sections.
  • DPIM liver fibrosis inducted by BDL.
  • DPIM was purchased from Sigma Chemicals (St. Louis, MO, USA).
  • mice Male FVB/N mice (8-10 weeks old) housed in a specific pathogen-free (SFP) facility on a 12 hour dark-light cycle and with free access to water and diet were used in the study.
  • SFP pathogen-free pathogen-free
  • the animal experiment protocol was approved by the institutional animal care and use committee (IACUC), the Biomedical Research Council (BMRC) of Singapore.
  • DPIM compound for treatment was supplied in the drinking water at three concentrations, 500 mg/1, 750 mg/1, lg/1, respectively.
  • the drinking water containing DPIM was changed weekly. It was observed that on average each mouse (normal or operated) drank approximately 3-4ml plain or DPM-containing water every day. Testing by NMR analysis was also conducted to confirm that DPIM is stable in water at room temperature condition for at least two weeks.
  • the compound treatment was started on the third day after animals recovered from the BDL procedure. After 4 weeks , livers were removed from mice for fixation 10% formalin, subsequently embedded in paraffin.
  • Serum was collected by retro-orbital method and serum alanine transaminase (ALT) activity was measured by automated procedures with Cobase c 111 (Roche diagnostic).
  • ALT serum alanine transaminase
  • DBZIM (1,3-bisbenzylimidazolium bromide) was synthesized based on a published method (Harlow et al. 1996).
  • Human heptocellular carcinoma cell lines BDLE (an undifferentiated cell line) and HepG2 (differentiated cell line) were purchased from JCRB (Japanese Collection of Research Bioresources).
  • Cell lines were routinely cultured in DMEM with 10% FBS in a humidified CO 2 incubator at 37 0 C, and split at 1:4 ratio by trypsinization (0.05% trypsin/0.53 mM ethylenediaminetetraacetic acid (EDTA) when they grew to confluence.
  • trypsinization 0.05% trypsin/0.53 mM ethylenediaminetetraacetic acid (EDTA) when they grew to confluence.
  • HLE cells grown in a 96-well plate were treated with compounds in 6 replicates for 24 hr. Cells were then fixed with 4% paraformaldehyde and stained with DAPI. Cells were analyzed and imaged on ArrayScan VTI HCS reader (Thermal Scientific, PA, USA) and D ⁇ A content (2 ⁇ vs. 4N) was determined based on DAPI staining.
  • HLE cells were grown, treated by compounds in 6 replicates for 48 hr in a 96-well plate. Cells were then fixed by using 4% paraformaldehyde and stained by DAPI and BrdU antibody using HitMt (Thermo Scientific, IL, USA). Cells were imaged and analyzed on ArrayScan VTI HCS reader (Thermal Scientific, PA, USA). Cell numbers were counted based on nuclear staining and BrdU incorporation was determined and expressed as percentage of positively stained cells.
  • Capspase 3/7 activity and lactate dehydrogenase (LDH) release were measured by Fluorometric assays (Promega Corp., WI, USA).
  • the HCC cells were cultured in 96-well plate and treated with DBZIM of various concentrations for 6 hr before assays.
  • the caspase 3/7 activity of sample cells with the compound treatrnent was expressed as relative fold change after normalizing to the control cells.
  • the LDH assay (indicative of membrane integrity) was performed using a Kit from Promega Corp according to manufactor's instructions.
  • Capspase 3/8/9 activities were determined using total protein extract as starting material (Biovision inc., CA, USA). HLE cells were cultured in T75 flask and treated with DBZM of various concentrations. Cytosolic protein extract was collected by pelleting and lysing the cells and centrifugation to remove the debris. Capspase 3, 8 and 9 activities were assayed using the same protein extract but adding the respective substrate. Absorbance readings were taken at 405 nm using Tecan Safire ⁇ plate reader. Results were expressed as relative caspase activity by normalizing to control cells without compound treatment.
  • HLE and HepG2 cells were treated with various concentrations of DBZIM or TRAIL
  • HLE cells cultured in 96-well plate were treated with DBZIM for 24 hr before staining and imaging. Cells were then fixed and stained with antibodies against apoptosis-inducing protein (ADP) and Surviyin (Lab Vision Corp. CA, USA) and buffer reagent from HitMt (Thermo Scientific, IL, USA). The cells were imaged and analyzed on ArrayScan VTI HCS reader (Thermal Scientific, PA, USA). Fluorescence intensity in the cytoplasmic and nuclear area was plotted for indication of cytoplasm to. nucleus translocation.
  • ADP apoptosis-inducing protein
  • Surviyin Label Vision Corp. CA, USA
  • HitMt Thermo Scientific, IL, USA
  • Protein extracts were prepared by lysing cell pellet in RJPA buffer (Pierce, IL, USA) for total protein extract, or in NE-PER reagent from Pierce (Pierce, IL, USA) for nuclear and cytoplasmic fractions, or in a Kit from BioVision (CA, USA) for mitochondria and cytosol fractions. Protein content was quantified using BCA Kit (Pierce, IL, USA). Fifteen micrograms of protein were resolved in 4-12% gradient SDS/polyacrylamide gel electrophoresis (PAGE) gel, and transferred to intracellular membrane.
  • the membrane was then probed with primary antibodies against BCI-X L , Bak, Bcl-2 ⁇ , Bax (Lab Vision Corp., CA, USA), survivin, xIAP, cIAP (R&D systems Inc., MN, USA), c-Jun, c-Fos, JunB, JunD, Fra-1, Fra-2 (Santa Craz Biotechnology, CA, USA) respectively, and detected by horseradish peroxidase-conjugated secondary antibody (Santa Cruz Biotechnology, CA, USA). Protein bands were recorded on X-ray film by reacting with ECL chemiluminescence reagents (Amersham Biosciences, NJ, USA).
  • Alpha-tubulin (Abeam, UK) was used as the loading control for total protein
  • Tata binding protein (TBP) (Abeam, UK) was used as the loading control for nuclear protein
  • Cox4 (Abeam, UK) was used as loading control for mitochondria protein.
  • Oxidative stress was measured using Cellomic HitKit (Thermo Scientific, IL, USA). HLE cells were treated with DBZIM for 24 hr, fixed and labeled by Dihydroethidium probe for quantitation of ROS generation and Hoechst dye for nuclear staining. The result was analyzed by Target Activation BioApplication software. The percentage of positive responding cells was plotted as the indication of ROS amount.
  • Thermal Scientific, PA, USA were used to exam the cytotoxicity and apoptosis profile of additional IMSs.
  • Cells were cultured and treated in 96-well plate for 24 hr before assaying. Cells were fixed and stained essentially following the ' manufacturer's instruction. The images were taken by ArrayScan VTI HCS reader (Thermal Scientific, PA, USA) and analyzed using Cell Viability and Cell Health Profiling BioApplication software respectively.
  • mice 5 ⁇ 6 weeks old, were inoculated with IxIO 7 Huh7 cells in 0.2 ml volume of Matrigel/DMEM mix. Since not every mouse developed a tumor after the inoculation of HCC cells, only those with visible tumors (about 8 weeks after inoculation) were used for subsequent experiment.
  • DBZM DBZBM, l,3,-Dibenzyl-2-memylir ⁇ lazolium bromide
  • Compound C), IBN- 2, 3, 4, 6, 8, 9 (Compound), 12, 13,15, 17, 18, 19, 20, 21, 24, 25, 26, 27, 28, 30, 31, 32 33, 34 were synthesized by mixing 1 mmol of substituted imidazole (like B 1 - B 4 ) and 1 mmol of substituted benzyl bromide(chloride) or alkyl bromide(chloride) in 1 ml ofDMF. The • mixture was stirred at 100 0 C for 16 hours. The reaction mixture was cooled down to room temperature and ether (10 ml) was added to precipitate product. The powder or gel precipitate was collected as imidazolium product and purified by re-crystalline or chromatograph method and characterized by NMR.
  • D3N-22, 23, 29 were made based on literature methods (Zhang et al. 2007; Chianese and Cratree 2005, Boydston et al. 2005).
  • Human Hepatocellular cancer cell lines HLE (p53 Mutant) and human gastric cancer cell line MKN 28 were obtained from Japan Health Sciences Foundation (Osaka, Japan).
  • the human breast cancer cell line MDAMB231 (mutant p53) and breast epithelial cells MCF-IOA (wild type p53) were obtained from ATCC. These cell lines were maintained in DMEM medium (RPMI 1640 for MDAMB231) supplemented with 10% fetal bovine serum and 1% of mixture Penicillin and Stretomycin incubated at 37 0 C in an atmosphere of 5% CO 2 .
  • the percentage of growth inhibition was determined by using a MTT assay to measure viable cells.
  • MTT assay 1 x 10 3 cells per well were cultured in 96-well plates and treated with 0 to 100 ⁇ M of various types of IMSs for 72 h. After incubation for specified times at 37 0 C in a humidified incubator, culture medium replaced with 10 ⁇ L of MTT reagent (5 mg/mL) contains 100 ⁇ L medium was added to each well and further incubated for 4 h. The reaction was stopped by adding 100 ⁇ L of DMSO. Absorbance was measured at 570 nm on a micro plate reader (SpectroMax 190, Molecular Devices). Data were presented from three separate experiments, and the percentage of IMSs induced cell growth inhibition was determined where DMSO-treated cells (control) were taken as 100%.
  • the cell pellets were resuspended in 1 ml of PBS and fixed by the addition of 70% ice-cold ethanol and stored at -2O 0 C. After incubation for 24hr, the cells were re- pelleted by centrifugation, the cells were washed once with PBS and resuspended in 0.1 mL of PBS containing 100 ⁇ g/ml of RNase and then incubated at 37°C for 15 min. Finally, the cells were stained with 0.5 ml of PI solution (100 ⁇ g/mL in PBS) for 30 min. Cell cycle distribution was detected by a BD PACS array flow cytometer (Becton Dickinson). Cells with sub-G 0 DNA were classified as apoptotic cells.
  • the cells were washed twice with cold PBS and then resuspend cell in 100 ⁇ lof x binding buffer containing a 10 ⁇ l of Annexin V-FITC (BD-Pharminagen) and 2 ⁇ l of PI from the stock of lOO ⁇ g/ml.
  • the cells were gently vortexed and incubated for 15 min at room temperature in the dark.
  • 400 ⁇ l of Ix binding buffer were added to each tube, and the samples were immediately analyzed by FACScan flow cytometer (Becton Dickinson).
  • the fluorescent signals of the Annexin-V conjugate and PI were detected at channels of fluorescence intensity FLl and FL2 (BD LSR ⁇ with software BD).
  • the HLE cells were seeded at a density of 4 X 10 5 in 10 cm dish. After 24 h incubation, the cells were replaced with fresh 10% DMEM medium and then treated with 60 ⁇ M of IBN-15, 90 ⁇ M of IBN-19, 90 ⁇ M of IBN-24, 30 ⁇ M of IBN-25 and 20 ⁇ M of IBN 32 and 0.045 % DMSO as control for 72h.
  • Whole cell lysates were prepared on day 3 with lysis buffer [125 mM Tris (pH 7.4), 2% Sodium dodecyl sulfate, 10% glycerol, 6M urea and a protease inhibitor cocktail (Sigma) and 0.02% Bromophenol blue].
  • the lysates were sonicated for 30 seconds and then heated for 5 mins at boiling temperature. The lysates were centrifuged at 13000 rpm for 10 mins and the protein concentration of each sample was estimated using the BCA protein assay. The lysates was then mixed with 5% 2-mercaptoethanol and stored at -80C. For Western blotting, samples containing 50 ⁇ g of total cell lysate were loaded onto a SDS-PAGE and subjected to electrophoresis. Proteins were transferred to a nitrocellulose membrane and then blocked with 5% non-fat dry milk in Tris buffer saline containing 0.1% Tween 20 for 60 min at 37 C.
  • the membranes were probed with a primary antibody overnight at 4 C in TBST containing 5% Bovine serum albumin. Then the membrane was incubated with horseradish peroxidase (HRP)-co ⁇ jugated anti-rabbit or anti-mouse secondary antibodies (GE Health care, Chalfont St Giles, UK). Detection was performed with enhanced chemiluminescence (ECL) reagent (Amersham Arlington Heights, IL) according to the manufacturer's protocol. Rabbit polyclonal Phospho-p53 (Serl5, 20, 46, 392), anti-caspase-9, anti-caspase-3, anti-PARP antibodies were purchased from Cell Signaling Technology, Inc. (Beverly, MA, USA).
  • HRP horseradish peroxidase
  • ECL enhanced chemiluminescence
  • Antibodies against p53 and p21 were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Equal loading of protein was demonstrated by probing the membranes with a mouse anti- ⁇ -actin monoclonal antibody (Sigma Chemical Co. (St. Louis, MO).
  • HLE cells were seeded at a density of 1 X 10 4 in 8 well chamber slide. After 24 h incubation, the cells were replaced with 200 ⁇ l fresh 10% DMEM medium and then treated with 60 ⁇ M of IBN-15, 90 ⁇ M of IBN-19, 90 ⁇ M of IBN-24, 30 ⁇ M of IBN-25 and 20 ⁇ M of IBN-32 and 0.045 % DMSO as control for 24 and 48 h. After washing, cells were fixed in 4% formaldehyde for 15 mins and permeabilized with 0.5% Triton X -100 in PBS for 10 mins.
  • the cells were then blocked for 60 min in PBS containing 3% bovine serum albumin and then incubated for 1 h with a mouse antibody against nucleolin (MS-3, Santa Cruz; 1:100 in blocking buffer). After washing extensively with PBS, the cells were then incubated for 1 h at room temperature in dark with a FITC-conjugated antibody against mouse IgG (1 :20 ilution). The slides were mounted with a VectaShield medium containing DAPI (Vector Laboratories, Burlingame, CA). Microscopy was performed with a Zeiss LSM 510 Meta (upright stand) confocal microscope.
  • RNA samples were seeded at a density of 4xlO 5 cells per dish in 10 cm dishes. Next day, the cells were exposed to 9OuM of IBN-19, 9OuM of IBN-24, 4OuM of IBN-25 and 0.045% of PMSO (Dimethyl Sulphoxide) as the control. After 48 hrs, total RNAs were extracted from the cells using Nucleospin RNA II kit as per the manufacturer's istructions. RNA quality and yield were evaluated after spectrophotometer measurements.
  • the human apoptosis PCR array (RT 2 Profiler) was used to analyze mRNA levels of 84 key genes involved in apoptosis, in a 96- well format, according to the manufacturer's instructions (Super Array Bioscience).
  • First-strand cDNA was synthesized with l ⁇ g of RNA by using a PCR array first strand-synthesis kit (C-03; Super array Bioscience). This kit uses reverse transcriptase (Power Script; Super array Bioscience) and a combination of random primers and oligo dT primers. The total volume of the reaction was 20 ⁇ L diluted to 100 ⁇ L.
  • PCR reactions were performed using real-time PCR (79s00HT 96- well block with RT 2 Real-Time SYBR Green PCR master mix PA-012; Applied Biosystems Instruments 7500). The total volume of the PCR reaction was 25 ⁇ l. An equivalent of l ⁇ g of RNA was applied to the PCR reaction. The thermocycler parameters were 95°C for 10 minutes, followed by 40 cycles of 95 0 C for 15 seconds and 6O 0 C for 1 minute. Relative changes in gene expression were calculated using the ⁇ C t (cycle threshold) method. An average of the number of cycles of the five housekeeping genes, GAPDH, Actin- ⁇ , ⁇ 2m, Hprtl, and Rpll3d, was used to normalize the expression between samples. The expression data are presented as fold regulation. GAPDH gene expression levels were utilized as an internal control to normalize the data. Reported fold changes in expression are the ratios of treatment over control values.
  • mice 5-6 weeks old, were inoculated with IxIO 7 Huh7 cells in 0.2 ml volume of Matrigel/DMEM mix. Since not every mouse developed a tumor after the inoculation of HCC cells, only those with visible tumors (about 8 weeks after inoculation) were used for subsequent experiment.
  • the compound treatment lasted for 3 weeks.
  • the tumor size was measured weekly, and the tumor volume was calculated using the formula: 0.52 x width 2 x length.
  • DBZM cytotoxicity of DBZM and TDBZM on cultured HSC-T6 cells using a 3-(4 3 5-dimethyM ⁇ azol-2-yl)-5-(3-carboxymethoxyp]ienyl)-2-(4-sulfoplienyl)-2H- tetrazolium (MTS) based proliferation assay kit (Promega, WI, USA).
  • Figs. Ia and Ib show the effect of DBZM (0-400 ⁇ M) and TDBZIM (0-200 ⁇ M) on the proliferation of T6 cells cultured in DMEM containing 10% of FBS.
  • DBZM inhibited cell proliferation by 3.5-8.9% with statistical significance.
  • TDBZM demonstrated a stronger inhibition of 13-19% as compared to the control.
  • TDBZM which is comprised of a trimer of DBZM, substantially suppressed cellular ROS level by 19% at 10 ⁇ M (P ⁇ 0.01) and by 36% at 100 ⁇ M (P ⁇ 0.00001) (Fig. 2b).
  • EGCG was able to attenuate ROS by 14% at 25 ⁇ M (P ⁇ 0.005), whilst NAC did not show apparent inhibition on ROS level even at 1 mM.
  • NAC (5 mM) did not show any effect on the total GSH, while EGCG (25 ⁇ M) induced the total GSH synthesis by 19% (P ⁇ 0.01), which was in agreement with an earlier report (Fu et al. 2006).
  • TDBZIM did not alter the total cellular GSH level at 10-100 ⁇ M (Fig. 3b).
  • GSH was oxidized to GSSG in the reaction to reduce H 2 O 2 to H 2 O. The amount of GSSG was also quantified as shown in Figs. 3c and 3d.
  • DBZIM depleted GSSG by 13% at 10 ⁇ M, 23% at 50 ⁇ M (P ⁇ 0.0005), 35% at 100 ⁇ M (P ⁇ 0.0001) and 85% at 300 ⁇ M (P ⁇ 0.0001).
  • EGCG reduced GSSG by 42% at 25 ⁇ M (P ⁇ 0.05) and NAC had no effect on GSSG at 5 mM.
  • TDBZIM also suppressed GSSG production by 12% at 10 ⁇ M (P ⁇ 0.05), by 35% at 50 ⁇ M (P ⁇ 0.05), and by 50% at 100 ⁇ M (P ⁇ 0.0001).
  • the GSH/GSSG ratio was enhanced by 8%, 15% (P ⁇ 0.001) and 23% (P ⁇ 0.0001) under the influence of DBZIM at 10, 50 and 100 ⁇ M, respectively (see Fig. 3e).
  • DBZIM 300 ⁇ M
  • the increase in GSH/GSSG ratio was more dramatic, reaching 7.4 fold of that of the control group.
  • NAC NAC
  • a 2-fold increase in the GSH/GSSG ratio was observed for EGCG at 25 ⁇ M (P ⁇ 0.0001).
  • FIG. 4a shows that DBZIM attenuated the GPx activity by 10% at 10 ⁇ M (P ⁇ 0.005) and by 10% at 50 ⁇ M (P ⁇ 0.01), but enhanced the GPx activity by 15% at a high concentration of 300 ⁇ M (P ⁇ 0.005), with a slight biphasic pattern.
  • GPx activity was suppressed by 18% at 10 ⁇ M (P ⁇ 0.05), by 25% at 50 ⁇ M (P ⁇ 0.01) and by 34% at 100 ⁇ M (P ⁇ 0.005) (Fig. 4b).
  • EGCG attenuated the GPx activity by 53% (P ⁇ 0.005) at ' 25 ⁇ M, while NAC did not show much effect at 5 mM (Fig. 4a).
  • FIG. 4g illustrates the GST activity of cells under the treatment of DBZIM (10-300 ⁇ M). GST activity was enhanced by 22% at 100 ⁇ M (P ⁇ 0.005) and by 32% at 300 ⁇ M (P ⁇ 0.0001). Ih contrast, EGCG and NAC did not have much affect on the GST activity.
  • TDBZM provided a more potent protective effect against DMSO's toxicity on primary HSCs.
  • DMSO-induced oxidative stress in cultured HSC T6 cells was quantitatively characterized by the depleted total cellular GSH level (74% of the control, P ⁇ 0.0001), the dramatically increased GSSG amount (4.4 fold of the control, P ⁇ 0.0001), the decreased GSH/GSSG ratio (17% of the control, P ⁇ 0.001), the induced GPx (2.62 times of the control, P ⁇ 0.0001) and CAT (1.7 fold of the control, P ⁇ 0.0001) anti-oxidant enzymes (see Figs. 5b,c,d,e,"f). DMSO also enhanced SOD activity slightly (10% increase), but the data were not statistically significant (Fig. 5g).
  • IMSs Suppressed HSC Activation Markers and Fibrogenic Endpoints in a Time- and
  • GFAP can be used as a biomarker along with SMAA for HSC activation in vitro ⁇ Zhang et. al 2006).
  • ECM including collagen and fibronectin
  • TDBZM Two MSs suppressed the mRNA level of both HSC activation markers (GFAP and SMAA) and fibrotic endpoint (collal and fibronectin) in a d ⁇ sage-dependent (Figs. 6a and 6b) and time- dependent (Figs. 7a-d) manner.
  • Fig. 6a and 6b Two MSs
  • DBZIM attenuated GFAP mRNA expression levels by 18% (P ⁇ 0.05) and 86% (P ⁇ 0.001) at 10 ⁇ M and 300 ⁇ M, respectively.
  • TDBZIM decreased GFAP mRNA by 32% at 1 ⁇ M (P ⁇ 0.01), by 20% at 10 ⁇ M (P ⁇ 0.01) and by 60% at 100 ⁇ M (P ⁇ 0.0001), SMAA by 27% only at a low dosage of 1 ⁇ M (P ⁇ 0.01), collal by 39% at 1 ⁇ M (P ⁇ 0.005), by 36% at 10 ⁇ M (P ⁇ 0.005) and by 53% at 100 ⁇ M (P ⁇ 0.001), and fibronectin by 32% at 1 ⁇ M (P ⁇ 0.0010), by 28% at 10 ⁇ M (P ⁇ 0.0005) and by 43% at 100 ⁇ M (P ⁇ 0.0001).
  • TGF- ⁇ 1 is the most important pro-fibrogenic cytokine.
  • quiescent HSCs respond to TGF- ⁇ 1 secreted by Kupffer cells and endothelial cells, and start to produce TGF- ⁇ l themselves via autocrine loops.
  • TGF- ⁇ l is over-expressed during HSC activation and liver fibrogenesis (Bachem et al. 1992).
  • the total TGF- ⁇ l synthesis under the treatment of MSs was measured.
  • DBZIM suppressed the TGF- ⁇ l mRNA by 28% (P ⁇ 0.05) and 33% (P ⁇ 0.005) at high concentrations of 100 ⁇ M and 300 ⁇ M, respectively.
  • TDBZM inhibited TGF- ⁇ l by ⁇ 23% regardless of dosage (1-100 ⁇ M) with statistical significance (P ⁇ 0.05) (Fig. 9b).
  • both DBZIM and TDBZM suppressed TGF- ⁇ l transcription by 40-50% (P ⁇ 0.05) (Fig. 9c).
  • the less apparent dosage- or time-dependency might be due to the auto-looping characteristic of TGF- ⁇ l.
  • IL-6 is regulated by NF- ⁇ B and involved in inflammatory responses. Being a pro- contractile and pro-inflammatory cytokine, IL-6 plays an important role in liver fibrogenesis.
  • IL-6 mRNA expression was quantified using real-time PCR. As shown in Figs. 10a, DBZIM inhibited IL-6 transcription in a dosage-dependent manner by 37% at 1 ⁇ M (P ⁇ 0.0005), by 46% at 10 ⁇ M (P ⁇ 0.0005), by 53% at 100 ⁇ M (P ⁇ 0.0001), and by 59% at 300 ⁇ M (P ⁇ 0.0001).
  • TDBZM demonstrated more potent inhibition of 37% at 1 ⁇ M (P ⁇ 0.01), 55% at 10 ⁇ M (P ⁇ 0.0001) and 80% at 100 ⁇ M (P ⁇ 0.0001) (Fig. 10b).
  • Time course study showed that 100 ⁇ M of DBZM significantly suppressed IL-6 mRNA expression by 53% at 48 h (P ⁇ 0.005).
  • Shorter treatment of DBZIM did not show statistically significant suppression on IL-6.
  • 100 ⁇ M of TDBZM demonstrated an inhibitory effect of 52% at 24 h (P ⁇ 0.005), and 80% at 48 h (P ⁇ 0.0001) (Fig. 10c).
  • NF- ⁇ B and AP-I are known to be sensitive to cellular redox states. NF-KB also plays an essential role in physiological conditions such as inflammation and tissue repair.
  • Protein expression of the NF- ⁇ B P65 and activation protein 1 (AP-I) family members (c-Fos, c-Jun, Jun B/D, Era- 1/2) were examined when the cells were treated with MSs.
  • nuclear NF- ⁇ B P65 in an active form was significantly attenuated by both DBZM and TDBZM. The amount of latent NF- ⁇ B in cytosol was not affected (data not shown).
  • AP-I, c-Fos, Jun D and Fra-1 were suppressed, while c-Jun, Jun B and Fra-2 were less affected.
  • IMSs of different structures were also tested in a preliminary structure-activity- relationship (SAR) study. Testing was performed on 3-Bisbenzylimidazolium bromide (DBZIM), l,3,5-tris(4-methyl-imidazolium)-linlced cyclophane-3Br (TDBZIM), 1,3-D ⁇ sopropylimidazolium tetrafluoroborate (DPIM), 1,3-Di-tert-butylirnidazolmumtetrafluoroborate (DBIM), 1,3-Bis(l- adamantyl)imidazolium tetrafluoroborate (AMTM), l,3-Bis(2,4,6-1rimemylphenyl)-imidazo]inium chloride (TMPHIM), l,3-Bis(2,6-diisopropyl-phenyl)-imidazolinium chloride (DPPHIM) and 1,3
  • those with smaller aliphatic groups such as 1,3-d ⁇ sopropylimidazolinium tetrafluoroborate (DPIM) and 1,3-di-fe ⁇ t-butylimidazolium tetrafluoroborate (DBIM)
  • DPIM 1,3-d ⁇ sopropylimidazolinium tetrafluoroborate
  • DBIM 1,3-di-fe ⁇ t-butylimidazolium tetrafluoroborate
  • TMPHTM 1,3-bis(2,4,6-trimethylphenyl)imidazolium chloride
  • DPPHIM 1,3-bis(2,4,6-trimethylphenyl)imidazolium chloride
  • DBZIM 1,3-bisbenzylimi dazolium bromide
  • DBZBIM 1,3- bisbenzylbenzimidazolium bromide
  • DPIM 3.1 mM
  • DBIM 1.7 mM
  • TDBZIM 1.7 mM
  • DBZBIM 310 ⁇ M
  • TMPHIM 110 ⁇ M
  • DPPHM 34 ⁇ M
  • Efficacy of compounds in suppressing fibrosis was also evaluated.
  • HSC T6 cells were treated with IMSs at concentrations that were below their respective IC50.
  • Compounds were either dissolved in DMSO or H 2 O depending on their solubility, and the intensities of the bands were compared to the respective control.
  • Fig. 13A proteins of SMAA, Collal, fibronectin, TGF- ⁇ l and TGF ⁇ RI were all suppressed with the treatment of MSs.
  • Semiquantitative data from measuring the gray scale of the band pixels is summarized in Fig. 13B .
  • GFAP mRNA was suppressed by 2 mM of DPIM by 78% (P ⁇ 0.0001), by 50 ⁇ M of AMIM by 69% (P ⁇ 0.0005), by 50 ⁇ M of TMPHIM by 66% (P ⁇ 0.001), and by 100 ⁇ M of DBZBIM by 50% (P ⁇ 0.005).
  • SMAA mRNA was suppressed by 1 mM of DBIM by 49% (P ⁇ 0.05), by 2 mM of DPIM by 78% (P ⁇ 0.0001), by 50 ⁇ M of AMIM by 23% (P ⁇ 0.05), and by 50 ⁇ M of TMPHIM by 49% (P ⁇ 0.05).
  • Fibronectin mRNA was suppressed by DPM by 48% (P ⁇ 0.05), by TMPHIM by 29% (P ⁇ 0.005), and by DBZIM by 26% (P ⁇ 0.005). Collal mRNA was suppressed by DBM by 58% (P ⁇ 0.0005), by DPM by 61% (P ⁇ 0.0001), by TMPHM by 62% (P ⁇ 0.0001), and by DPPHM by 47% (P ⁇ 0.05). EGCG did not show significant inhibition on HSC activation and fibrosis. This might due to the chemical instability of EGCG under the prolonged incubation of 48 h.
  • DBZIM and DPM were tested in two liver fibrosis models in mice induced by liver toxin thioacetamide (TAA) or by a surgical procedure called bile duct ligation (BDL), respectively.
  • TAA liver toxin thioacetamide
  • BDL bile duct ligation
  • DPM was chosen for further in vivo study, To ensure proficiency in BDL procedure, a number of exercises and tests were performed to eventually achieve over 90% success rate on the BDL surgery and over 4 weeks of survival time for the operated mice. The extent of fibrosis was assessed by the Sirius-Red collagen staining at 4 weeks post-BDL. An extensive collagen accumulation around the sinusoidal areas was observed in the BDL group (Fig. 16C), whereas only minimal collagen staining was seen in the sham operated group (Fig. 16A). Treatment of sham mice with lg/1 of DPM for 4 weeks did not induce any significant build-up of collagen (Fig. 16B).
  • the livers of different treated groups were weighed, 4 weeks after BDL, liver weight was increased by 83% compared with sham operated mice (1.50 ⁇ 0.19g for sham operated, vs. 2.74 + 0.5 Ig for BDL 4 weeks, P ⁇ 0.05).
  • DPIM compound treatments showed a significant liver weight reduction (P ⁇ 0.05, DPIM compound treatment vs. BDL 4 weeks): 1.97 ⁇ 0.44g for the 750mg/l treatment group, 1.65 ⁇ 0.2Og for the lg/1 treatment group (Fig. 18).
  • FIG. 19 H&E staining of the liver sections (Fig. 19) showed that DPIM treatment (Fig. 19C) reduced the number of proliferative cells (identified by the nuclear staining of Hamehexlin) around the sinusoidal area, when compared to the DBL group (Fig. 19B) and control sham group (Fig 19A). At the messenger level, DPM also reduced the collagen IaI mRNA (Fig. 20).
  • DBZM The effect of DBZM on the HCC cell proliferation and cell cycle was characterized on a high-content screening (HCS) platform. Under each concentration, at lease 1000 cells were analyzed. 5-bromo-2'-deoxyuridine (BrdU), which incorporates into replicating DNA, is used to identify cells which have progressed through S phase of cell cycle. DAPI staining was used for nuclear identification and DNA content determination. DBZM inhibited the HLE cell proliferation as indicated by less total cell number and less BrdU positive cells in a dose-dependant manner (Fig. 21A), and arrested HLE cells in the G0/G1 phase (Fig. 21B).
  • HCS high-content screening
  • - Cell-based caspase 3/7 activity and lactate dehydrogenase (LDH) release assay allow the differentiation of apoptosis and necrosis of cells under the influence of DBZEM.
  • caspase 3/7 activity indicative of apoptosis
  • LDH release indicative of cytotoxicity
  • DBZM used at the current treatment condition caused the HCC cell death mainly by apoptosis, rather than necrosis.
  • Apoptosis was further confirmed by annexin V staining.
  • PS membrane phosphatidylserine
  • the surface PS is readily detected by fluorescent conjugate of annexin V, which has a high affinity for PS and often used as an apoptosis marker.
  • DBZIM dose-dependently induced apoptosis in both HepG2 and HLE cells. It was noted that as a positive control, the TRAIL (TNF-Related Apoptosis Inducing Ligand) only induced apoptosis in HepG2, but not in HLE cells.
  • caspase 3, 8, 9 activities were assayed using colorimetric assays. As shown in Fig. 23 A, caspase 8 activity was not affected by DBZTM, whilst caspase 9 and 3 activities were dose-dependently enhanced by DBZTM, pointing a mitochondria-mediated apoptosis pathway. However, DBZIM was found to cause a minimal release of cytochrome C from the mitochondria to the cytosol (Fig. 23B).
  • the mitochondria-mediated intrinsic apoptosis pathway is mainly regulated- by the
  • Bcl-2 and the IAP Inhibitor of Apoptosis Protein
  • the anti-apoptotic protein BCI-X L and the pro-apoptotic protein Bak were slightly down- regulated and up-regulated by DBZIM respectively, whereas the other two members of the Bcl-2 . family, Bcl-2 ⁇ and B ax, were not affected by the treatment.
  • survivin was significantly suppressed by DBZIM, *whereas xIAP and cIAP were not affected (Fig. 24).
  • HCS analysis revealed that survivin was translocated from the nucleus to the cytoplasm under the influence of DBZIM (Fig. 25).
  • Apoptosis-inducing factor is normally localized in the mitochondria, and translocated to the nucleus upon induction of apoptosis. It induces mitochondria to release caspase 9. In nucleus, AIF also induces chromatin condensation and DNA fragmentation, a process known as caspase independent apoptosis pathway. DBZIM was found to induce the translocation of AIF to the nucleus by Western blotting (Fig. 26A). Immunocytostaining showed that AIF was translocated from the mitochondria/cytoplasm to the nucleus (Fig. 26B).
  • ROS Reactive oxygen species
  • a sustained and elevated level of transcription factor activation protein 1 (AP-I) complex is known to participate in apoptosis.
  • Western blotting (Fig. 28) of nuclear extracts from HLE cells showed that DBZIM significantly induced several key members of the AP-I complex, including C-Jun, Jun B, Jun D, and Fra-1, but not Fra-2.
  • DBZIM liver cell specific lung cells
  • DBZIM liver cell specific lung cells
  • IC50 value 50 ⁇ M for H1299 (lung cancer), 50 ⁇ M for MCF-7 (breast cancer), and 40 ⁇ M for AGS gastric cancer at day 3 (Table 3).
  • DBZIM showed much less cytotoxicity on the normal (noncancerous) lung cells (IMR90).
  • treatment of AGS and H1299 with DBZIM resulted in morphological (apoptotic look) changes and significant reduction in the number of cells (Fig 29A and 29B).
  • DBZIM significantly reduced the growth of two glioma cell lines, U87MG (human) and C6 (rat) (Fig. 30A and 30C), with IC50 (determined at day 2) for U87 MG and C6 being 2.1 mM and 1.0 mM, respectively.
  • Akt protein kinase B
  • DBZM is particularly efficacious in inducing apoptosis in AGS, H1299 and MCF-7 cells through a mechanism involving the reduction in the levels of me, phospho-AKT.
  • DBZIM reduced tumor growth in a xenograft HCC mouse model
  • HCC cell lines HepG2, Hep3B, Huh7, PLC, and HLE
  • Huh7 line was among the most efficient. Therefore a murine HCC model was established using the Huh7 cells for testing the antitumor efficacy of DBZM in vivo.
  • Fig. 33A Three weeks into the treatment, difference in tumor size was apparent between mice in the control and the treatment group (Fig. 33A). Measurement showed that the tumor volume in the DBZM-treated mice was significantly reduced by 40% (P ⁇ 0.01) when compared to the control mice (Fig. 33B). It was noted that the body weight (including a reduction in the tumor weight) of the DBZM-treated mice decreased by 17% (P ⁇ 0.01) after three weeks of treatment (Fig. 33C).
  • ⁇ 53 The expression level of ⁇ 53 in Hep 3B ( ⁇ 53null), HepG2 (p53 wild type), and HLE and PLC (p53 Mutant) hepatocarcinoma cells was determined by Western Blot. (Fig. 34). The expression of ⁇ 53 was high in cells bearing mutant p53, as compared to cells with the wild type P53.
  • IMSs EBN-15, IBN-19, D3N-24, IBN-25 and JBN-32
  • the cytotoxicity activity and proliferation inhibition of EMSs in tumor cells were measured by MTT.
  • the treatment of HLE liver cancer cells with IMSs resulted in a dose dependent inhibition of cell proliferation.
  • the control experiments witih DMSO alone (0.05% v/v) had no effect on cell proliferation.
  • the IC S0 value for IBN-15, IBN-19, IBN-24, IBN-25, and IBN-32 in HLE cells was estimated to be 60 ⁇ m, 90 ⁇ m, 90 ⁇ m, 30 ⁇ m, and 20 ⁇ m at 72 h.
  • Annexin V-PI dual-staining assay was performed in the same cell type under the same conditions to investigate whether the growth inhibition resulted from cell apoptosis.
  • Cells undergoing apoptosis would stain positive for Annexin V-FITC and negative for PI (quadrant 4, Q4).
  • Cells which stained positive for both Annexin V-FITC and PI (Q2) are either at the end stage of apoptosis or undergoing necrosis, and those which stained negative for both Annexin V-FITC and PI (Q3) were alive or undergoing undetectable apoptosis.
  • cell debris stained only for PI Ql).
  • IMSs IBN-15, IBN-19, EBN-24, IBN-25, and IBN-32 induced 33%, 29%, 29%, 21%, and 18% (Q2) cell apoptosis, respectively, whereas fewer apoptotic cells (7.2%) were observed for DMSO treated cells, confirming that the IMS-induced cell growth inhibition has a significant apoptotic component
  • IMSs increased the expression ofp53 and phosphorylated p53 (Serl5, 20, 46 and Ser
  • IMS-induced apoptosis analyses of p53 and its down stream effectors were performed in HLE cells, which carry the mutant p53.
  • Immunocytochemical analyses revealed that IMSs (IBN-19 and IBN-24) induced p53 expression and accumulation in the nucleus (Fig. 39).
  • IMSs induced the execution of apoptosis through activation of the mitochondrial pathway
  • the changes in the levels of pro-apoptotic protein Bax, and anti-apoptotic protein Bcl-2 were analyzed. Immunoblot analysis showed that treatment of HLE cells with various MSs increased Bax protein level and simultaneously decreased Bcl-2 level (Fig. 41).
  • HCC cell lines HepG2, Hep3B, Huh7, PLC, and HLE were tested for their ability to efficiently establish HCC in nude mice, and found that Huh7 line was among the most efficient. Therefore a murine HCC model was established using the Huh.7 line and one of the MSs (compound 9) was tested to assess its anti-tumor efficacy.
  • Fig 44A Three weeks into the treatment, significant difference in tumor size was apparent between mice in the control and the treatment group (Fig 44A). Measurement showed that the tumor volume in the C9-treated mice was significantly reduced by 43% and 62% (P ⁇ 0.01) at 0.6g/l and 1.5g/L respectively, when compared to the control mice (Fig. 44B). No change in body weight or gross pathology was noted for the C9- treated mice, when compared to the control mice (Fig. 44C), implying lack of significant general toxicity associated with the C9 under the current dosing scheme.
  • GSH plays a key role in cellular anti- oxidative processes by serving as an electron donor to GPx in the reduction of hydrogen peroxide to water, and as a nucleopMIic co-substrate to GST in the detoxification of xenobiotics.
  • Anti- ' oxidants exert their effect mainly through three different pathways: (1) neutralization of cellular ROS generated during metabolism and immune response, (2) induction of endogenous anti-oxidative enzymatic activity, and (3) chelation of iron or copper ions that catalyze the generation of hydroxyl radical.
  • GPx, CAT and SOD which constitute the first line of cellular anti-oxidative defense, are directly involved in the neutralization of ROS.
  • GPx enzyme reduces H 2 O 2 to H 2 O, while GSH functions as a cofactor and is consequently oxidized to GSSG.
  • CAT is a peroxisomal enzyme and converts H 2 O 2 to H 2 O.
  • SOD catalyzes the dismutation of superoxide. There are three forms of SOD: extracellular and intracellular copper/zinc (Cu/Zn) SODs, and a mitochondrial manganese (Mn) SOD.
  • GST is a phase ⁇ anti-oxidant enzyme. It has been reported that activation of HSCs is associated with the loss of GST activity in culture (Whalen et. al 1990).
  • IMSs might have mainly exerted their anti-oxidant effect through the direct neutralization of ROS, such as hydrogen, peroxide. Therefore, the burden on cellular anti-oxidative defense was alleviated by the exogenous synthetic anti-oxidative IMSs, resulting in a decrease in the expression of first-tier anti-oxidative enzymes.
  • the effect of IMSs on GSH synthesis and anti- oxidative enzymes showed a general pattern of attenuation at low dosages, less potent attenuation and even enhancement at high dosages. This suggested that DBZM worked through different mechanisms at low and high dosages.
  • IMSs at high dosage enhanced cellular defense activity against oxidative stress by synergizing neutralization of ROS and induction of endogenous anti-oxidant enzymes.
  • phase II enzymes such as total GST were enhanced in activity under the treatment of IMSs. It was known that GSTs protect the cells against oxidative toxicants by conjugating the xenobioti.es to GSH, thereby neutralizing their electrophilic sites and rendering the product water-soluble. The enhancement of GST activity indicated that the cells have better capability of getting rid of oxidants such as lipid peroxidation products, thereby minimizing the cellular oxidative injuries.
  • IMSs showed Anti-Inflammatory Effect Through Suppression of NF- KB and IL-6
  • Transcription factor NF- ⁇ B is found in the cytosol bound to inhibitory protein known as IKB.
  • IKB inhibitory protein
  • NF-KB is activated and translocated into the nucleus where it binds to DNA and regulates the transcription of gene-encoding proteins involved in immune or inflammatory responses (Vasiliou et al. 2000, AIi et al. 2004).
  • the activation of NF- ⁇ B is associated with phosphorylation and subsequent degradation of IKB.
  • the NF- ⁇ B -responsive site has been identified and characterized in the promoters and enhancers of many genes, including IL-6 and ICAM-I (AIi et al. 2004).
  • NF-KB is also known to be sensitive, to oxidative stress. Most agents activating NF-KB are either modulated by ROS or oxidant themselves. It has been reported that the treatment of antioxidant resveratrol (Chavez et al. 2007) or vitamin E (Liu et al. 1995) attenuated NF- ⁇ B elevation induced in carbon tetrachloride experimental f ⁇ brotic rodents.
  • HSCs are well-recognized cellular regulators in the development of hepatic fibrosis.
  • HSC activation associated with enhanced secretion of pro-fibrogenic, pro-inflammatory cytokines (such as TGF- ⁇ l and IL-6) is characterized by the over-expression of SMAA, and results in the over-deposition of ECM proteins including collal and fibronectin.
  • the anti-fibrotic effects of IMSs were evident by the suppression of HSC activation markers (GFAP and SMAA) and fibrotic endpoints (collal and fibronectin) in a dosage- and time-dependent manner.
  • a transiently induced SMAA expression was observed under the treatment of high-dosage TDBZIM (Fig.
  • TGF- ⁇ 1 is the most potent pro-fibrogenic- cytokine, which is over-expressed in activated HSCs.
  • TGF- ⁇ l signaling is initiated by the binding of the active form of TGF- ⁇ l to type ⁇ receptor, and subsequently leading to the phosphorylation of type I receptor (TGF ⁇ RI).
  • TGF ⁇ RI type I receptor
  • Activated TGF ⁇ RI recruits- Smads protein, and propagates the downstream signal to regulate the gene expression of matrix proteins (Fu et al. 2006).
  • IMSs' effect on treating liver fibrosis TGF- ⁇ l and TGF ⁇ RI in T6 cells under IMS treatment were measured.
  • Activation of HSC is also associated with elevated levels of NF- ⁇ B and NF- ⁇ B- responsive genes, including IL-6 (Mann et al. 2006).
  • the role of NF- ⁇ B is to maintain HSCs in the activated state and promote a chronic wound healing response.
  • NF-KB has been implicated in various aspects of liver disorders, including hepatic inflammation, fibrosis, and the development of hepatocellular carcinoma (Elsharkawy et al. 2007).
  • IMSs greatly suppressed active NF-KB p65 protein (Fig. 11), while not affecting the cytosolic NF- ⁇ B P65 level (data not shown).
  • AP-I is a homodimer or heterodimer composed of at least one Jun family protein (c-Jun, JunB and JunD) and another member from the Fos family (c-Fos, Fral and Fra2). It has been reported that JunD is functionally the most important AP-I factor in activated HSCs required for the induction of IL-6 andthe tissue inhibitor of metalloproteinase I (TIMP-I) gene transcription (Smart el al. 2001). It is known that HSC T6 cells express all members of Jun and Fos proteins (Zhang et al., 2006). Under the influence of IMSs, c-Fos, JunD and Fra-1 levels were attenuated (Fig. 11).
  • IMSs In line with the down-regulation of JunD, IL-6 gene transcription was suppressed (Figs. lOa-c).
  • IMSs might prevent hepatic fibrosis through the suppression of HSC activation, and by decreasing the deposition of ECM proteins (collal and fibronectin) and the production of pro-fibrogenic cytokines (TGF- ⁇ 1 , TGF ⁇ RI and IL-6) mediated through NF- ⁇ B and AP- 1 signaling.
  • NHC Precursors could Be Drug-Like Compounds
  • ROS has been implicated in many physiological conditions, including neurodegenerative diseases, cardiovascular diseases, stroke, heart attack, cancer, aging and fibrosis, etc. Therefore, anti-oxidation has been pursued as a therapeutic strategy for a number of diseases. Dietary anti-oxidants have been widely used to ameliorate excessive oxidative stress both in animal models and humans. For example, resveratrol has been shown to extend the lifespan of various species, and to be effective at improving the health and survival of mice on a high-calorie diet (Baur et al. 2006). Tea polyphenols (EGCG being the most abundant catechin) have been shown to inhibit carcinogen-induced DNA damage in animal models of skin, lung, colon, liver and pancreatic.
  • EGCG being the most abundant catechin
  • IMSs are precursors of NHCs, in which the electronic structure and stability, and thus, therapeutic safety and efficacy can be tuned by varying the N-substituents.
  • NHCs with bulky substituents favor a triplet ground state and exhibit radical-like reactivity, while those with smaller substituents adopt a singlet ground state and show both nucleophilic and electrophilic behavior (Bourissou et al. 2000).
  • IMSs demonstrated potent anti-oxidative, anti-inflammatory and anti- fibrotic properties in cultured HSCs.
  • IMSs showed relatively low and tunable toxicity with IC50 in the range of 35 ⁇ M to 3.1 mM. It is hypothesized that the anti-f ⁇ brogenic and anti-inflammatory effects of the IMS compounds are derived through the anti-oxidative property targeted at inhibiting HSC activation.
  • the radical scavenging property of the MSs is presumably originated from the product after a series of chemical reactions including:(l) the spontaneous conversion of IMSs to NHCs preferably under a basic condition, (2) the interaction of the NHCs at the carbon 2-position with free radicals such as ROS resulting in the formation of intermediate active radicals and the neutralization of the free radicals.
  • Hepatocellular carcinoma is known to develop over liver insults, which leads to hepatocyte inflammation and regeneration, matrix protein remodeling, fibrosis/cirrhosis and eventually HCC.
  • HCC is inherently chemotherapy-resistant and is know to express the drug- resistant gene MDR-I.
  • Advanced HCC also shows poor prognosis even after a successful surgical resection (Ariizumi et al.2004).
  • the data showed that 1 IMS compound DBZIM can induce apoptosis an in both differentiated (HepG2) and undifferentiated (HLE) HCC cell line without significant cytotoxicity (Fig. 22).
  • DBZM can also inhibit the proliferation of HLE (Fig. 21 A) via cell cycle arrest at G0/G1 phase (Fig. 21B).
  • Apoptosis can be triggered through either the extrinsic pathway or the intrinsic pathway.
  • the extrinsic pathway is initiated through the simulation of transmembrane death receptors such as Fas receptors and activation of caspase 8 which leads to caspase 3/7 cleavage.
  • the intrinsic pathway is initiated through the release of apoptosis activators from mitochondria and activation of caspase 9 is involved to cleave caspase 3/7 which leads to apoptosis.
  • enzyme activity assays showed that DBZIM induced the activities of caspase 9 and 3 but not caspase 8 (Fig. 23).
  • DBZIM was also shown to down-regulate survivin (Fig. 24) and induce cytoplasm location of survivin (Fig. 25).
  • Survivin is one member of IAPs protein family, expressed during human foetal development and in several neoplasms, but not in normal tissue except in thymus and placenta. It has been shown that nuclear translocation of surviving in HCC promotes the cancer cell growth. (Moon et al. 2003) Nuclear and cytoplasmic expression of survivin has also been related to prognosis of pancreatic cancer (Tonini et al. 2005).
  • DBZIM perturbated the regulatory mechanism by translocation of survivin and suggesting a possible new angle for searching for HCC chemotherapy.
  • the alteration of mitochondria function is associated with the early phases of the cell death. Meanwhile, observed inhibition of the mitochondrial respiratory chain causes the over production of ROS, which mediates the death signaling pathway. (Mignotte et al. 1998) The mode of cell death, either by apoptosis or necrosis is largely dependant on the level of intracellular ROS . Also it is difficult to differentiate whether ROS is the result or the cause of the killing process or both. In this study, it was interesting that MSs of different concentration had different physiological effect, in terms of ROS production.
  • DBZM was administered to a HCC xenograft mouse model (Huh7cells in nude mice) via drinking water, after the tumor growth was evident and at the log phase about 8 weeks after cell inoculation.
  • IMSs effectively inhibit tumor cell growth concomitant with induction of cell cycle arrest and apoptosis.
  • IMSs activate ATM system and subsequently the phosphorylation and stabilization of p53.
  • a correlation was drawn between the level of p53, phospho-p53 (Ser-15) and ATM in HLE liver cancer cells.
  • phospho-p53 Ser-15
  • ATM and ATR are activated by DNA damage. Activation of the ATM pathways has long been associated with the cell cycle checkpoint induced by several DNA damage/genotoxic stress. Once activated, ATM phosphorylates various downstream molecules such as p53, MDM2, Chkl, Chk2, resulting in cell cycle arrest or cell death.
  • Tumor suppressor gene p53 is a key element in the induction of cell cycle arrest and apoptosis following DNA damage or cellular stress in human cells and cell cycle arrest. (Saito et al. 2002, Hofseth et al, 2003) In this study, it has been shown that treatment of HLE cells with IMSs resulted in the accumulation of p53 and phospho-p53 (Serl5). It has also been found that the induction of ATM gene expression causes DNA damage andp53 activity, suggesting that p53 induced by DNA damage play an important role in IMSs-mediated cell cycle arrest.
  • Up-regulated p53 protein can activate the pro-apoptotic protein BAX and inhibits Bcl-2 to cause mitochondrial dysfunction and cytochrome c release, stimulating the mitochondrial apoptotic pathway (Melino et al., 2004).
  • IMSs inhibited BcI- 2 and activated. Bax in the HLE cancer cells. It was found that IMSs effectively inhibit tumor cell growth, concomitant with induction of cell cycle arrest and apoptosis. Furthermore, the fact that IMSs do not exhibit any significant toxicity in normal lung and breast cells suggests that IMSs possesses selectivity between normal and cancer cells.
  • AIi S Mann D A. Signal transduction via the NF- ⁇ B pathway: A targeted treatment modality for infection, inflammation and repair. Cell Biochem Funct 2004; 22:67-79.
  • Garlic compounds generate reactive oxygen species leading to activation of stress kinases and cysteine proteases for apoptosis in human glioblastoma T98G and U87MG cells. Cancer. 2007; 10(5)1083-1095.
  • Cirrhosis new research provides a basis for rational and targeted treatments.
  • MDR-I Multidrug resistance gene
  • ARE antioxidant response element
  • Vitamin E therapy of acute CCl 4 - induced hepatic injury in mice is associated with inhibition of nuclear factor-kappaB binding. Hepatology 1995;22:1474-1481.
  • Tonini G Vinvenzi B, Santhii D, Scarpa S, Vasaturo f, Malacrino C, Coppola R, magistrelli P, Borzomati D, Baldi A, Antinori A, Caricato M, Nuzzo G, Picciocchi A. Nuclear and cytoplasmic expression of suvivin in 67 sugically resected pancreatic cancer patients. Br J Cancer. 2005; 92:2225-2232. Tsukamoto H, Rippe R, Niemela O, Lin M. Roles of oxidative stress in activation of Kupffer and Ito cells. J Gastroenterol Hepatol 1995;10:s50-s53.
  • Valko M Izakovic M, Mazur M, Rhodes C, Tesler J Role of oxygen radical in DNA damage and cancer incidence. MoI. Cell. Biochem. 2004; 266; 37-56.

Abstract

There is presently provided methods for delivering an anti-fibrotic or anti-cancer agent to a cell. The methods comprise contacting a cell with an effective amount of imidazolium and imidazolinium compounds as described herein, including imidazolium and imidazolinium salts.

Description

METHOD FOR TREATING FIBROSIS AND CANCER WITH EVΠDAZOLIUM AND IMTOAZOLINIUM COMPOUNDS
CROSS-REFERENCE TO RELATED APPLICATION
[0001 ] This application claims benefit of, and priority from, U.S. provisional patent applications No. 61/006,769 filed on January 30, 2008, the contents of which are fully incorporated herein by reference.
FmLD OF THE INVENTION
[0002] The present invention relates to methods for delivering an anti-fibrotic or an anti-cancer agent to a cell, including methods for treating fibrotic disease or cancer.
BACKGROUND OF THE INVENTION
[0003] Hepatic stellate cells (HSCs) play a pivotal role in hepatic fϊbrogenesis and are considered a major cellular target for therapeutic intervention. However, no approved anti-fibrotic drug is currently on the market.
[0004] Hepatic fibrogenesis may be incurred as a result of insults arising from oxidative stress, chemical toxicity or viral infection. Thus fibrotic disease, including cell and tissue fibrosis, is among the physiological disorders that have been associated with oxidative stress. For example, oxidative stress resulting from the metabolic generation of ROS has been linked to HSC activation and liver fibrosis (Britton et al. 1994, Tsukamoto et al. 1995, Galli et al. 2005). It has been shown that products of lipid peroxidation lead to increased collagen synthesis by HSCs (Casird et al. 1997 andJParolaet al. 1996).
[0005] Reactive oxygen species (ROS) are oxygen derivatives that include free radicals and non-radical reactive molecules such as peroxides and oxygen derivatives. Oxidative stress occurs when there is an excess amount of ROS due to an imbalance between the generation and the elimination or neutralization of these molecules. Excess amounts of ROS may damage cell lipids, proteins and DNA resulting in the inhibition of normal cell function or the development of abnormal cellular behaviour which can in turn lead to a variety of diseases and conditions, depending on the cells affected. ROS have been implicated in many physiological disorders and in the onset and development of a number of diseases.
[0006] Some anti-oxidants have been explored as potential inhibitors of hepatic fibrosis (Kawada et al. 1998). (-)-Epigallocatechin gallate (EGCG), a major active component of tea catechin, has been shown to inhibit HSC activation in vitro via transforming growth factor-beta (TGF-β) signalling (Chen et al. 2002, Nakanuta et al. 2005, Fu et al. 2006). In clinical studies, a combination of vitamins E and C was shown to decrease the fibrosis score in non-alcoholic steatohepatitis patients, but did not affect hepatic inflammation (Harrison et al. 2003). N-acetyl-L- cysteine (NAC), a synthetic precursor of glutathione (GSH) that has been clinically used as an antioxidant, showed anti-fibrogenic properties through the suppression of TGF-β signalling transduction as well (Meurer et al. 2005).
[0007] Oxidative stress has also been associated with the onset and progression of cancer. It has been reported that oxidative stress as indicated by reduced anti-oxidant enzyme activities is associated with the deyelopment of primary carcinogenesis and metastasis in clinical patients (VaIi et al. 2008). ROS have been found to cause DNA damage increasing the risk of DNA mutation and thus the development of cancer (Hussain et al. 2005). Tea polyphenols are anti-oxidants and have been shown to inhibit carcinogen-induced DNA damage in animal models of skin, lung, colon, liver and pancreatic cancers (Frei et al. 2003).~In addition, anti-oxidants PBN and NXY-059 have demonstrated anti-cancer activity in hepatocellular carcinoma. (Floyd 2006). However, the effectiveness of the anti-oxidants investigated remains unclear (Valko et al, 2004).
[0008] The inflammatory response is the immune system's response to infection or injury and involves the activation of cells of the immune system which produce mediators, such as cytokines, that further activate other cells, leading to a cascade of immune reactions that fight off the infection or repair the injury. Like oxidative stress, inflammatory stresses have been associated with fibrotic disease and cancer (Rakoff-Nahoum 2006; Tsukamoto et al. 1999; Bachem et al. 1992; Vasiliou et al. 2000). For example, in the development of liver fibrosis, HSCs play a critical role. Responding to liver injury, HSCs undergo a process called "activation" and trans-differentiate to myofibroblast- like cells. This process is characterized by phenotypic changes including cell proliferation, over- expression of smooth muscle actin-α (SMAA), and deposition of extracellular matrix (ECM) proteins, including collagen type αl (I) (collal) and fibronectin.
[0009] Inflammatory cytokines represent major mediators for HSC activation. Among them, transforming growth factor-beta I (TGF-β 1) and interleukin 6 (TL-6) have been categorized as profibrogenic cytokines mainly responsible for the induction of ECM proteins (Tsukamoto 1999). HSCs respond to TGF-β 1 secreted from Kupffer cells and endothelial cells during liver injury, and themselves via autocrine action, resulting in HSC activation and liver fibrosis (Bachem et al. 1992). In addition, the activation of HSCs can also be attributed to chronic hepatic inflammation through the secretion of proinflammatory cytokine IL-6, leading to cirrhosis (Vasiliou et al. 2000) and hepatocellular carcinoma (Naugler et al, 2007). Transcription factor nuclear factor kappa B (NF-κB) is an important regulator for the secretion of inflammatory cytokines, and its subunit NF-κB p65 was reported to mediate liver fibrosis (Vasiliou et al. 2000).
[0010] Beyond sharing a common association with fibrotic diseases and cancer, the production and regulation of oxidative and inflammatory stress appear to be interconnected, as an inflammatory response can trigger oxidative stress and vice versa.
[0011 ] For example, in hepatic fibrogenesis, both ROS and pro-inflammatory cytokines have been shown to be involved in tiie activation of HSCs, the central event in the development of liver fibrogenesis. TGF-β is a major mediator for HSC activation and TGF-β signalling is affected by both oxidative stress and the induction of an inflammatory response (Tsukamoto et al.l999, Bachem et al. 1992, Chen et al. 2002, Nakanuta et al. 2005, Fu et al. 2006, Meurer et al. 2005). Another key molecule in the development of fibrosis, is NF-κB which is an important regulator of the secretion of inflammatory cytokines but is also known to be sensitive to oxidative stress. Most agents activating NF-κB are either modulated by ROS or oxidant themselves. It has been reported that treatment with anti-oxidant resveratrol (Chavez et al. 2007) or vitamin E (Liu et al. 1995) attenuated NF-icB elevation induced in carbon tetrachloride experimental fibrotic rodents.
[0012] Oxidative stress and inflammatory response have also been found to be interrelated in the pathogenesis of cancer. Oxidative stress can cause DNA mutations, some of which will result in the formation of cancer cells. However other mutations will result in cell death, stimulating an inflammatory response. In turn, an inflammatory response will not only provide survival and proliferative signals to cancer cells but may also induce the production of ROS (Rakoff-Nahoum 2006).
[0013] Dietary anti-oxidants have been widely used as a general approach to ameliorate excessive oxidative stress both in animal models and humans. For example, resveratrol has been shown to extend the lifespan of various species, and to be effective at improving the health and survival of mice on a high-calorie diet (Baur et al. 2006).
[0014] To date, the development of effective treatments for oxidative stress and certain diseases associated with oxidative stress using natural or synthetic anti-oxidants has been problematic. Stringent scientific proof for the efficacy of natural anti-oxidants has not been established (Droge et al. 2001). Some of the notable limitations for using natural anti-oxidants a.s therapeutics include low potency and fast turnover during metabolism. In contrast, the development of synthetic anti-oxidants has been inhibited by safety concerns. Nevertheless, some progress has been made in this direction. Modification of a natural anti-oxidant has been performed to enhance its potency (Keum et al. 2007). In addition, synthetic mimics of superoxide dismutase (SOD) and catalase have been shown to be effective in rodent models of ischemia and Parkinson's disease (Peng et al. 2005). Even more encouragingly, a class of nitron-free radical trap agents, alpha- phenyl-N-tert-butyl-nitron (PBN) and disodium 2,4-disulfophenyl-N-tert-butyhiitrone (NXY-059), have been shown to be potent neuroprotective agent (Maples et al. 2004), and have demonstrated anti-cancer activity in hepatocellular carcinoma through its anti-inflammatory property (Floyd 2006).
SUMMARY OF THE INVENTION
[0015] There is presently provided methods for delivering an anti-fibrotic or anti-cancer agent to a cell, the method comprising contacting a cell with an effective amount of imidazolium and imidazolinium compounds, including imidazolium and imidazoliαium salts, as described herein.
[0016] In one aspect, there is provided a method for delivering an anti-fibrotic or anti-cancer agent to a cell, the method comprising contacting the cell with an effective amount of a compound of general formula I or an oligomer or polymer thereof:
Figure imgf000006_0001
wherein:
the dashed line is absent or is present as a bond to form a second bond between the carbon to which R1 and R3 are attached and the carbon to which R2 and R4 are attached;
R1 and R2:
(i) are each independently H, straight or branched C1-C6 alkyl, straight or branched
C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C6-C1O aryl; (ii) together with their ring atoms form a 6- to 10-membered fused saturated, unsaturated or aromatic ring system; (iϋ) R1 and R5 together with their ring atoms, or R2 and R6 together with their ring atoms, form a 5- to 10-membered fused saturated, unsaturated or aromatic ring system and the other of R1 and R2 is as defined above in (i); or
. (iv) R1 and R5 together with their ring atoms and R2 and R6 together with their ring atoms each form a 5- to 10-membered fused saturated, unsaturated or aromatic ring system;
R3 and R4 are both H, or, when R1 and R2 together with their ring atoms form a 6- to 10-membered fused aromatic ring system or when the dashed line is present as a bond, R3 and R4 are absent;
R5 or R6:
(i) are as defined above for R1 and R2; or
(ii) are each independently straight or branched Ci-C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C18 cycloalkyl including fused cycloalkyl ring systems, C6-CiO aryl, C6-CiO aryl-Ci-C6 alkyl, C6-CiO 8TyI-C2-C6 alkenyl, or C6-Ci0 aryl-C2-C6 alkynyl, Ci-C6 alkyl-C6-Cio aryl, C2-C6 alkenyl-C6-Ci0 aryl, or C2-C6 alkynyl-C6-Cio aryl;
R7 is H, Ci-C6 alkyl, phenyl, substituted Ci-C6 alkyl or halo;
in which any of R1 to R7, where applicable, optionally has one or more carbon atoms replaced with" a heteroatom selected from N, O, S and P and is optionally substituted with one or more of straight or branched Ci-C6 alkyl, straight or branched C2-Cg alkenyl, straight or branched C2-C6 alkynyl, C3- Cis cycloalkyl including fused cycloalkyl ring systems, C6-CiO aryl, fluoro, tri-fluoro-methyl, . cyanato, isocyanato, carboxyl, Ci-C6 acyloxy, Ci-C6 acyl, carbonyl, amino, acetyl, acetoxy, oxo, nitro, hydroxy., Ci-C6 alkylcarboxy, Ci-C6 alkoxy, C2-C6 alkenoxy, C2-C6 alkynoxy; and
and in which one of the ring carbon atom to which R1 and R5 are attached and the ring carbon to R2 and R6 are attached is optionally replaced with a nitrogen atom;
or a pharmaceutically acceptable salt thereof.
[0017] In one embodiment, the compound is an imidazolium or an imidazolium. In another embodiment, the compound is an imidazolinium or an irnidazolinium salt.
[0018] In one embodiment, R5 is the same as R6. In another embodiment R5 and R6 are hydrocarbons.
[0019] In certain embodiments, the pharmaceutically acceptable salt of general formula I may be a chloride, bromide, tetrafluoroborate or hexafmorophosphate salt.
[0020] In various embodiments, the compound may be l-ethyl-3-methylimidazolium, 1,3- bisbenzylimidazolium, 1,3-dϋsopropylimidazolium, 1,3-di-tert-butylimidazolinium, l,3-bis(l- adamantyl)imidazolium, 1 ,3 -bis(2,4,6-1iimemylphenyl)-imidazolinium, 1 ,3-bis(2,6-diisopropyl- phenyl)-irnidazoHnium, 1,3-diallylimidazolium, l-benzyl-3-meώylimidazolium, l-butyl-3- methylimidazolium, l-(l-adamantyl)-3-(2,4,6-trimethylphenyl)-4,5-dihydroimidazolium, 2- benzyUmidazo[l,5-α]quJnolinium, l,3-bis(l-adamantyl)-benzimidazolium, 1,3- dicyclohexylbenzimidazolium, 1,3-dϋsopropyHmidazoMnium tetrafluoroborate, 1,3- diisopropyHmidazolium, 2-(2,6-diisopropylphenyl)-5-methylimidazo[l,5-fl]pyridinium, l-(2,6- diisopropylphenyl)-3-(2,4,6-1ximeib.ylphenyl)-imidazolinium, 2-mesityl-5- metfiylirnidazo[l,5- α]pyridinium, 2-mesityl-2,5,6,7-tetrahydropyrrolo[2,l-c][l,2,4]triazol-4-ium, l,3-bis(l- adamantyl)imidazoUnium, l-bu1yl-3-(2-pyridinyhnethyl)-lH-imidazoUum, 6,7-dihydro-2- pentafluorophneyl-5Η-pyrrolo [2, 1 -c] - 1 ,2,4-trizolium, 1 -methyl-3-(2-hydroxylethyl)-imidazolium, l-methyl-3-(4-isocynatobenzyl)-imidazolium, l-methyl-3-(4-carboxylbenzyl)-imidazolium, 1- methyl-3-(4-acetate-benzyl)-imidazolium, l-methyl-3-(2,2-dimethoxylethyl)-imidazolium, l-(2,4,6- trimethylphenyl)-3-(4-acetate-benzyl)-imidazolium, l,3-Dibenzyl-5-phenylimidazolium, 1-benzyl- 3-(4-carboxylbenzyl)-2-methyHmidazolium, l-benzyl-3-(3,4,5-trimethoxylbenzyl)-2- methylimidazolium, l-benzyl-3-(4-acetate-benzyl)-2-methyl-imidazolium, l-benzyl-3-(4- methylcarboxylatebenzyl)-2-methyl-imidazolium, l-benzyl-3-(2,2-dimethoxylethyl)-2-methyl- imidazolium, 2,6-di-(3-benzyl-imidazolium)-pyridine, 2,2' -di-(3-benzyl-imidazolium)-l,l'- binaphthalene, l-benzyl-3-(4-methylbenzyl)-imidazolium, l-benzyl-3-(2-trifluoromethylbenzyl)-2- methyUmidazolium, l-benzyl-3-(4-methylcarboxylatebenzyl)-5-phenyl-imidazolium, l-benzyl-3- (4-acetatebenzyl)-5-phenyl-imidazolium, l-benzyl-3-(4-methylbenzyl)-5-phenylimidazolium, (1 ,2- 4,5-diimidazolium)-N,N',N",NJ"-tetrabenzyl-benzene, l-benzyl-3-(2-propyn-l-yl)-imidazolium, 1- benzyl-3-(3-hydroxyl-propyl)-imidazolium, l,3-di(2-phenylethyl)-imidazolium, l-benzyl-3-(4- acetatebenzyl)-imidazolium, l-benzyl-3-(pyridin-2-yl)-imidazolium, l,3,5-tris-(4-methyl- imiazolium)-linked cyclophane, l,3-dibenzyl-2-(l,3-dibenzyl-lH-imidazol-2(3H)-ylidene)-2,3- dihydro-lH-imidazole, l-benzyl-3-methyl-imidazolium, l-(4-cyanatobenzyl)-3-methyl- imidazolium, l-(4-carboxybenzyl)-3-me1iιyl-imidazolium, l-methyl-3-(4-acetate-benzyl)- imidazolium, l-meΛyl-3-(2,2-dimethoxyethyl)-irnidazolium, or l-(2,4,6-trimethylphenyl)-3-(4- acetate-benzyl)-imidazolium, or a pharmaceutically acceptable salt thereof. [0021 ] In one embodiment, the compound is a dimer of a compound having a structure of general formula I. For example, the compound may be l,3-dibenzyl-2~(l,3~dibenzyl-lH-imidazol- 2(3H)-ylidene)-2,3-dihydro- lH-imidazole, (l,2-4,5-dϋimdazottum)-N,N' ,N" , N" ' -tetrabenzyl- benzene, 2,6-di-(3-benzyl-imidazolium)-pyridine or 2,2'-di-(3-benzyl-imidazolium)-l,r- binaphthalene, or a pharmaceutically acceptable salt thereof.
[0022] In another embodiment, the compound is a trimer of a compound having a structure of general formula I. For example, the compound is l,3,5-tris(4-me1hyl-imidazolium)-Mnked cyclophane or a pharmaceutically acceptable salt thereof.
[0023] In one embodiment of the present methods, the cell may be in vitro.
[0024] In another embodiment, the cell may be in vivo. For example, the method may comprise administering the agent to a subject for the treatment of fibrotic disease or the treatment of cancer. In one particular embodiment, the fibrotic disease may be hepatic fibrosis amd the compound may be for example, 3-dϋsopropylimidazolium or a pharmaceutically acceptable salt thereof. In other embodiments, the cancer may be hepatocellular carcinoma, lung cancer, breast cancer, stomach cancer or glioma or the compound may be for example, 3-Bisbenzylimidazolium, 3-dibenzyl-2- methylimidazolium, l-(2,4,6-trimethylphenyl)-3-(4-acetate-benzyl)-imidazo]ium, l-benzyl-3-(4- acetate-benzyl)-2-methyl-imidazolium, l-benzyl-3-(4-methylbenzyl)-imidazolium, l-benzyl-3-(2- trifluoromethιylbenzyl)-2-memylimidazolium or l,3-di(2-phenylethyl)-imidazolium, or a pharmaceutically acceptable salt thereof.
[0025] In another aspect, there is provided use of a compound having a structure of general formula I, or an oligomer or polymer thereof, for delivering an anti-fibrotic or anti-cancer agent to a cell in vivo:
[0026] In another aspect, there is provided use of a compound having a structure of general formula I, or an oh'gomer or polymer thereof, in the manufacture of a medicament for delivering an anti-fibrotic or anti-cancer agent to a cell in vivo. '
[0027] In one embodiment of the uses described herein, the compound is an imidazolium or an . imidazolium salt. In another embodiment, the compound is an imidazolinium or an imidazolinium salt.
[0028] In other embodiments, R5 is the same as R6. In another embodiment R5 and R6 are hydrocarbons. [0029] In certain embodiments, the pharmaceutically acceptable salt of general formula I may be a chloride, bromide, tetrafluoroborate or hexafluorophosphate salt.
[0030] In various embodiments of the present uses, the compound may be l-ethyl-3- methylimidazolium, 1,3-bisbenzylimidazolium, 1,3-diisopropylimidazolium, 1,3-di-tert- butylimidazolinium, l,3-bis(l-adamantyl)hnidazolium, l,3-bis(2,4,6-trimethylphenyl)- imidazolinium, l,3-bis(2,6-dϋsopropyl-phenyl)-imidazolinium, 1,3-diallylimidazolium, l-benzyl-3- methylimidazolium, l-butyl-3-methylimidazolium, l-(l-adamantyl)-3-(2,4,6-trimethylphenyl)-4,5- dihydroimidazolium, 2-benzylimidazo[l,5-α]quinolinium, l,3-bis(l-adamantyl)-benzimidazolium, 1,3-dicyclohexylbenzimidazohum, 1,3-dusopropylimidazoliπium tetrafluoroborate, 1,3- diisopropylimidazolium, 2-(2,6-diisopropylphenyl)-5-methyHmidazo[l ,5-σ3pyridinium, l-(2,6- diisopropylphenyl)-3-(2,4,6-trimethylphenyl)-imidazolinium, 2-mesityl-5- methylimidazo[l ,5- α]pyridinium, 2-mesityl-2,5,6,7-tetxahydropyrrolo[2,l-c][l,2,4]triazol-4-ium, l,3-bis(l- adamantyl)imidazolimum, l-butyl-3-(2-pyridinylmethyl)-lH-imidazolium, 6,7-dihydro-2- pentafluorophneyl-5Η-pyrrolo[2,l-c]-l,2,4-trizolium, l-methyl-3-(2-hydroxylethyl)-imidazolium, l-methyl-3-(4-isocynatobenzyl)-imidazolium, l-methyl-3-(4-carboxylbenzyl)-imidazolium, 1- methyl-3-(4-acetate-benzyl)-imidazolium, l-methyl-3-(2,2-dimethoxylethyl)-imidazolium, l-(2,4,6- 1τimethylphenyl)-3-(4-acetate-berizyl)-imidazoliurn, l,3-Dibenzyl-5-phenylimidazolium, 1-benzyl- 3-(4-carboxylbenzyl)-2-methyKmidazolium, l-benzyl-3-(3,4,5-trimethoxylbenzyl)-2- methylimidazolium, l-benzyl-3-(4-acetate-benzyl)-2-methyl-imidazolium, l-benzyl-3-(4- methylcarboxylatebenzyl)-2-methyl-imidazolium, l-benzyl-3-(2,2-dimethoxylethyl)-2-methyl- imidazolium, 2,6-di-(3-benzyl-imidazolium)-pyridine, 2,2'-di-(3-benzyl-imidazolium)-l,r- binaphthalene, l-benzyl-3-(4-methylbenzyl)-imidazolium, l-benzyl-3-(2-trifluoromethylbenzyl)-2- methylimidazoliurn, l-benzyl-3-(4-methylcarboxylatebenzyl)-5-phenyl-imidazolium, l-benzyl-3- (4-acetatebenzyl)-5-phenyl-imidazolium, l-benzyl-3-(4-methylbenzyl)-5-phenylimidazolium, (1,2- 4,5-dϋmidazolium)-N,N',N",N'"-tetrabenzyl-benzene, l-benzyl-3-(2-propyn-l-yl)-imidazolium, 1- benzyl-3-(3-hydroxyl-propyl)-imidazolium, l,3-di(2-phenylethyl)-imidazolium, l-benzyl-3-(4- acetatebenzyl)-imidazolium, l-benzyl-3-(pyridin-2-yl)-imidazolium, l,3,5-tris-(4-methyl- imiazolium)-linked cyclophane, l,3-dibenzyl-2-(l,3-dibenzyl-lH-imidazol-2(3H)-ylidene)-2,3- dihydro-lH-imidazole, l-benzyl-3-methyl-imidazolium, l-(4-cyanatobenzyl)-3-methyl- imidazolium, l-(4-carboxybenzyl)-3-methyl-imidazoILum, l-methyl-3-(4-acetate-benzyl)- imidazolium, l-methyl-3-(2,2-dimethoxyethyl)-imidazolium, or l-(2,4»6-trimethylphenyl)-3-(4- acetate-benzyl)-imidazolium, or a pharmaceutically acceptable salt thereof.
[0031 ] In one embodiment, the compound is a dimer of a compound having a structure of general formula I. For example, the compound may be l,3-dibenzyl-2-(l,3-dibenzyl-lH-imidazol- 2(3H)-yπdene)-2,3-daydro-lH-imidazole, (l,2-4,5-diimidazolium)-N,N',N'', N'''-tetrabenzyl- benzene, 2,6-di-(3-benzyl-imidazolium)-pyridine or 2,2'-di-(3-benzyl-imidazolium)-l,r~ binaphthalene, or a pharmaceutically acceptable salt thereof.
[0032] In another embodiment, the compound is a trimer of a compound having a structure of general formula I. For example, the compound is l,3,5-tris(4-methyl-imidazolium)-linked cyclophane or a pharmaceutically acceptable salt thereof.
[0033] In one embodiment of the present uses, the anti-fibrotic agent is delivered for the treatment of fibrotic disease. In a particular embodiment, the fibrotic disease may be hepatic fibrosis amd the compound may be for example, 3-diisopropylimidazolium or a pharmaceutically acceptable salt thereof.
[0034] In another embodiment of the present uses, the anti-cancer agent is delivered for the treatment of cancer. In various embodiments the cancer s hepatocellular carcinoma, lung cancer, breast cancer, stomach cancer or glioma and the compounds may be, for example, 3- Bisbenzylimidazolium, 3-Dibenzyl-2-methylimidazolium, l-(2,4,6-trimethylphenyl)-3-(4-acetate- benzyl)-imidazolium, l-benzyl-3-(4-acetate-benzyl)-2-methyl-imidazόlium, l-benzyl-3-(4- methylbenzyl)-imidazolium, l-benzyl-3-(2-trifluoromethylbenzyl)-2-methylimidazolium or 1,3- di(2-phenylethyl)-imidazolium, or a pharmaceutically acceptable salt thereof.
[0035] In another aspect there is provided, a compound that is an imidazolium or imidazolinium that is l-ethyl-3-methylimidazolium, l-methyl-3-(2-hydroxylethyl)-imidazolium, l-methyl-3-(4- isocynatobenzyl)-imidazolium, l-methyl-3-(4-carboxylbenzyl)-imidazolium, l-methyl-3-(4-acetate- benzyl)-imidazolium, l-methyl-3-(2,2-dimethoxylethyl)-imidazolium, l-(2,4,6-trimethylphenyl)-3- (4-acetate-benzyl)-imidazolium, l,3-Dibenzyl-5-phenylimidazolium, l-benzyl-3-(4- carboxylbenzyl)-2-methylimidazolium, l-benzyl-3-(3,4,5-trimethoxylbenzyl)-2- methylimidazolium, l-benzyl-3-(4-acetate-benzyl)-2-methyl-imidazolium, l-benzyl-3-(4- methylcarboxylatebenzyl)-2-methyl-imidazolium, l-benzyl-3-(2,2-dimethoxylethyl)-2-methyl- imidazolium, 2,6-di-(3-benzyl-imidazolium)-pyridine, 2,2'-di-(3-benzyl-imidazolium)-l,r- binaphthalene, l-benzyl-3-(4-methylbenzyl)-imidazolium, l-benzyl-3-(2-trifluoromethylbenzyl)-2- methylimidazolium, 1 -benzyl-3 -(4-me1hylcarboxylatebenzyl)-5-phenyl-imidazolium, 1 -benzyl-3 - (4-acetatebenzyl)-5-phenyl-imidazolium, 1 -benzyl-3 -(4-methylbenzyl)-5-plienylimidazolium, (1 ,2- 4,5-diimidazolium)-N,N',N",N'"-tetrabenzyl-benzene, l-benzyl-3-(2-propyn-l-yl)-imidazolium, 1- benzyl-3-(3-hydroxyl-propyl)-imidazoIium, l,3-di(2-phenylethyl)-imidazolium, l-benzyl-3-(4- acetatebenzyl)-imidazolium, l-benzyl-3-(pyridin-2-yl)-imidazolium, l,3,5-tris-(4-methyl- imiazoh'um)-liiLked cyclophane, l,3-dibenzyl-2-(l,3-dibenzyl-lH-irnidazol-2(3H)-ylidene)-2,3- dihydro-lH-imidazole, l-benzyl-3-methyl-imidazolium, l-(4-cyanatobenzyl)-3-methyl- imidazolium, 1 -(4-carboxybenzyl)-3 -methyl-imidazolium, 1 -methyl-3-(4-acetate-benzyl)- imidazolinm, l-methyl-3-(2,2-dimethoxyethyl)-imidazolium, or l-(2,4,6-trimethylphenyl)-3-(4- acetate-benzyl)-imidazolium, or a pharmaceutically acceptable salt thereof.
[0036] Other aspects and features of the present invention will become apparent to those of ordinary skill in the art upon review of the following description of specific embodiments of the invention in conjunction with the accompanying figures.
BRIEF DESCRIPTION OF THE DRAWINGS
[0037] In the figures, which illustrate, by way of example only, embodiments of the present invention:
[0038] Figure. 1. Cytotoxicity of (a) DBZIM and (b) TDBZIM. HSC T6 cells were seeded at a density of 5000 cells/well in 96-well plate, and cultured for 18-24 h in 10% FBS DMEM before the addition of various compounds (XM-OO μM) in 10% FBS DMEM for 48 h of proliferation before assaying. The signal was normalized against a vehicle control treatment (0 μM of compound). The data were obtained from four independent experiments, and presented as mean and standard error or mean (SEM), *aP < 0.05, *bP < 0.01 and *CP < 0.005.
[0039] Figure.2. (a) DBZIM and (b) TDBZIM attenuated cellular ROS level. HSC T6 cells were incubated with DBZIM (10, 50, 100 and 300 μM), TDBZIM (10, 50 and 100 μM), NAC (1 mM) and EGCG (25 μM) for 48 h before assaying for cellular ROS levels. The data were presented as relative values after normalizing against a vehicle control. The data were presented as mean and SEM, N = 6, *bP < 0.01, *CP < 0.005, and *dP < 0.0005 when compared to the vehicle control.
[0040] Figure. 3. Effect of DBZIM and TDBZIM on amount of GSH (a) DBZM attenuated total cellular GSH amount, and (b) TDBZIM did not change the total GSH. (c) DBZM and (d) TDBZM both suppressed cellular GSSG (the oxidation product of GSH). (e) DBZM and (f) TDBZM both enhanced the GSH/GSSG ratio. HSC-T6 cells were incubated with compounds of various concentrations for 48 h, and assayed for the total GSH and GSSG amounts. The GSU/GSSG ratio was calculated as (GSH - GSSG)/GSSG. The GSH and GSSG amounts were normalized against the total protein. Note that DBZEM was dissolved in DMSO, and TDBZM was dissolved in H2O. The data were presented as mean and SEM, N = 6, *aP < 0.05, *bP < 0.01, *CP < 0.005, and *dP < 0.0005, when compared to the vehicle control. [0041 ] Figure. 4. Effect of DBZIM and TDBZIM on GPx, CAT5 SOD and GST. DBZM affected (a) GPx activity and (c) CAT activity in a dosage-dependent manner. TDBZIM suppressed (b) GPx activity and (d) CAT activity, (e) DBZIM and (f) TDBZIM had little effect on SOD activity, (g) DBZIM induced GST activity. HSC T6 cells were treated with compounds of various concentrations for 48 h, and homogenized by sonication (60% frequency for 30 s) in PBS (pH 7.4, 1 mM of EDTA) for GPx, GST and CAT assays, or in 20 mM of HEPES buffer containing 1 mM of EGTA, 210 mM of mannitor and 70 mM of sucrose (pH 7.2) for SOD assay. Enzyme activity was normalized against total protein. DBZIM was dissolved in DMSO, and TDBZIM was dissolved in H2O. The data were presented as mean and SEM, N = 6, *aP < 0.05, *bP < 0.01, *CP < 0.005, and *dP < 0.0005, when compared to the vehicle control.
[0042] Figure 5. DBZIM and TDBZIM protected primary HSC cells against oxidative stress induced by 0.2% (v/v) of DMSO. (a) Optical images of cells treated with IMSs for 26 and 93 h. Scale bar = 60 μm. 0.2% (v/v) of DMSO induced oxidative stress by depleting the total GSH level (b), inducing the GSSG level (c), reducing the GSH/GSSG ratio (d), and inducing GPX (e), CAT (f) and SOD (g) activity levels. The data were presented as mean and SEM, N = 6, *CP < 0.005, and *dP < 0.0005, when compared to the untreated sample.
[0043] Figure. 6. (a) DBZIM and (b) TDBZIM suppressed the mRNA expression of HSC activation markers (GFAP and SMAA) and fibrotic endpoints (collal and fibronectin) in a dosage-dependent manner. HSC T6 cells were treated with DBZIM (1, 10, 100 and 300 μM) and TDBZIM (1, 10 and 100 μM) for 48 h. β-actin was used as the normalization gene; its expression level was constant under the experimental conditions. The ΔΔCt method was used for relative quantification with vehicle control sample being the calibrator. The data were presented as mean and SEM, N = 6, *aP < 0.05, *b P < 0.01, *CP < 0.005, and *dP < 0.0005, when compared to the vehicle control.
[0044] Figure. 7. DBZIM (100 μM) and TDBZIM (100 μM) suppressed (a) GFAP, (b) SMAA, (c) collal and (d) fibronectin mRNA expression in a time-dependent manner. HSC T6 cells were treated with DBZIM and TDBZIM, and assayed after 8, 24 and 48 h. A transient elevation of SMAA expression was observed for TDBZIM at 24 h. The mRNA expression was calculated using the ΔΔCt method by normalizing to β-actin house-keeping gene and vehicle control sample, and presented as relative quantification data with mean and SEM, N = 6, *aP < 0.05, *bP < 0.01, *CP < 0.005 , and *dP < 0.0005, when compared to the vehicle control.
[0045] Figure. 8. Western blotting of collal, fibronectin, GFAP and SMAA protein expression in HSC T6 cells treated with DBZIM (100 μM) and TDBZIM (50 μM) for 48 h. 15 μg of total proteins were resolved in 3-8% of tris-acetate gel for collal and fibronectin, or 4-12% gradient PAGE-SDS gel for GFAP and SMAA. The target proteins were recognized by the respective antibodies, and visualized by the ECL method. The membrane was stripped and re- probed for α-tubulin as the loading control.
[0046] Figure. 9. DBZIM and TDBZIM suppressed TGF-βl mRNA. HSC T6 cells were incubated with (a) DBZM (1, 10, 100 and 300 μM) and (b) TDBZIM (1, 10 and 100 μM) for 48 h for the dosage-dependent study, and (c) with DBZIM (100 μM) and TDBZM (100 μM) for 8, 24 and 48 h for the time-dependent study. The expression level of TGF-βl mRNA was quantified with real-time RT-PCR. The data were presented as mean and SEM, N = 6, *aP < 0.05, *bP < 0.01, *°P < 0.005, when compared to vehicle control.
[0047] Figure. 10. DBZIM and TDBZIM suppressed JL-6 mRNA. HSC T6 cells were incubated with (a) DBZM (1, 10, 100 and 300 μM) and (b) TDBZM (1, 10 and 100 μM) for 48 h for the dosage-dependent study, and (c) with DBZM (100 μM) and TDBZIM (100 μM) for 8, 24 and 48 h for the time-dependent study. The expression level of IL-6 mRNA was quantified with real-time RT-PCR. The data were presented as mean and SEM, N = 6, *bP < 0.01, *CP < 0.005, and *dP < 0.0005, when compared to the vehicle control.
[0048] Figure. 11. Western blotting of NF-κB and AP-I (c-Fos, JunD and Fra-1) protein expressions in HSC T6 cells treated with DBZIM (100 μM) and TDBZIM (50 μM) for 48 h. 15 μg of nuclear protein was resolved in 4-12% gradient PAGE-SDS gel. The target proteins were recognized by the respective antibodies, and visualized by the ECL method. The membrane was stripped and probed for the Tata binding protein TBP used as the loading control.
[0049] Figure. 12. (a) DBZIM and (b) TDBZIM attenuated GFAP-£αcZ transgene as reported by β-galactosidase activity. Stable transfected T6 GFAP-LαcZ cells were treated with compounds of 1-300 μM concentrations, and assayed after 24 h and 48 h. The β-galactosidase activity was normalized'to the total protein, and presented as relative value after comparing to the vehicle control. The data were presented as mean and SEM, N = 6, *CP < 0.005, and *dP < 0.0005, when compared to the vehicle control.
[0050] Figure. 13. Western blotting of SMAA, collal, fibronectin, TGF-βl, TGFβ RI protein expressions in HSC T6 cells treated with various IMSs for 48 h. (A)DBIM (1 mM), AMM (50 μM), TMPHM (50 μM), DPPHM (10 μM) and DBZBM (100 μM) were dissolved in DMSO with a final DMSO concentration of 0.2% (v/v). DPM (2 mM), EGCG (25 μM) and DBZtM (100 μM) were dissolved in H2O. Alpha-tubulin was used as the loading control. Densitometric measurement of protein band intensity was normalized against the respective vehicle control. (B) Relative densitometric quantification of protein band intensity. The vehicle treated sample was normalized to 1.
[0051 ] Figure. 14. Real-time PCR quantification of (a) GFAP, (b) SMAA, (c) fibronectin and (d) collal mRNA expressions in HSC T6 cells treated with various IMSs for 48 h. DBIM (1 mM), AM-M (50 μM), TMPHIM (50 μM), DPPHIM (10 μM) and DBZBIM (100 μM) were dissolved in DMSO with a final DMSO concentration of 0.2% (v/v). DPIM (2 mM), EGCG (25 μM) and DBZIM (100 μM) were dissolved in H2O. β-actin was used as the normalization gene; its expression was constant under the experimental conditions. Relative quantification was normalized against the respective vehicle control. The data were presented as mean and SEM, N = 6, *aP < 0.05, *CP < 0.005, and *dP < 0.0005, when compared to the vehicle control.
[0052] Figure. 15 Effect of DBZEM and DPIM on liver fibrosis in mice. DBZEM was tested on mice induced by (a) liver toxin thioacetamide (TAA) or by (b)bile duct ligation (BDL) . DPIM was also tested on mice induced by TAA (data not shown)) or by (c) bile duct ligation (BDL). DBZIM attenuated fibrosis in the TAA model (at 500mg/L, 12 weeks) and the BDL model (at 10mg/L, 4weeks), whereas the DPIM displayed its effect in the BDL model at lg/L (4 wks).
[0053] Figure. 16. Representative of images of Sirius red staining in liver sections from different DPIM compound treated group mice. Each treated group had 6-8 mice, except for 500mg/I treated group, which had only one mouse in the group. Red staining areas represent collagen deposits. (A) sham operated, (B) sham operated + DPIM lg/1 for 4 weeks, (C) BDL for 4 weeks, (D) BDL 4 weeks + 500mg/l DPIM treatment, (E) BDL 4 weeks + 750mg/l DPIM treatment, and (F) BDL 4weeks + lg/1 DPIM treatment.
[0054] Figure. 17. Percentage of Sirius red staining areas. Sirius red staining areas from different treatment group mice were quantified with Image J software, at least 6 different areas from left and middle lobes of liver of each mouse were covered. * P<0.05, BDL vs. DPIM treated groups, # P<0.05, 750mg/l vs. lg/1 treated groups.
[0055] Figure 18. Liver weights of different treatment groups. Each group consisted of 6-8 mice which were 10-12 weeks old at the start of surgery procedure. *P<0.05, sham operated control group vs. BDL 4weeks, ** P<0.05, DPIM compound treated groups vs-. BDL weeks group, # P
<0.05, 750mg/l DPM treated group vs. lg/1 DPM treated group. 10056] Figure 19. Representative images of H&E staining of liver sections. (A) control group; (B) BDL 4 weeks group; and (C) BDL 4 weeks +DPIMlg/l treatment group.
[0057] Figure 20. Quantitative real-time PCR for collagen IaI mRNA. DPIM reduced collagen IaI mRNA.
[0058] Figure 21. DBZIM inhibited cell proliferation and disrupted cell cycle in HLE cells
(A). DBZM inhibited HCC cell proliferation. HLE cells were seeded at a density of 5000 cells/well in 96-well plate, and cultured for 18-24 h in 10% EBS DMEM before the addition of various compounds (0-1.25 mM) in 10% FBS DMEM for 48 h of proliferation before assaying. Cell count was calculated based on Hoechst dye staining and BrdU incorporation was based on the total fluorescence intensity of antibody staining. (B). DBZM arrested HCC cells at G0/G1 phase. HLE cells were incubated with DBZIM (0, 1, 3 mM) for 24 hr before fixation and DAPI staining for DNA content (2N vs. 4N) determination.
[0059] Figure.22. DBZIM induced caspase 3/7 activity in HLE cells (A) and HepG2 (C) and did not cause LDH releasing in HLE (B) and HepG2 (D). HLE or HepG2 cells were incubated with DBZEVI of various concentrations for 6 hr, and assayed for caspase 3/7 activity and LDH releasing (expressed as % toxicity). DBZIM induced apoptosis as characterized by annexin V staining using flow cytometer in HLE (E) and HepG2 (F). Scatter plot and histogram gave the representative information on the cell population distribution while bar graphs quantified the percentage of apoptotic cells from the histogram. HLE and HepG2 cells were treated by DBZIM of various concentrations for 24 hr before harvesting for annexin "V staining and flow cytometric analysis.
[0060] Figure 23. (A). DBZIM did riot change caspase 8 activity and induced caspase 9 and 3 activity in HLE cells. HLE cells were grown in a T75 flask and treated with DBZM for 24 hr. The cells were then lysed and cytoplasmic protein was collected for assaying caspase 8, 9 arid 3 using kits from Biovision. The data was normalized against the vehicle control. (B). DBZM did not cause cytochrome c release from mitochondria to cytosol. HLE cells were treated by DBZM of 1.5 mM for 24 hr. Cytosolic and mitochondria protein fractions were collected using a kit from Biovision. 15.μg of each protein was resolved in 4-12% gradient PAGE-SDS gel. The target proteins were recognized by antibody of cytochrome C, and visualized by the ECL method. The membrane was stripped and probed for the α-tubulin as the loading control for cytosolic fraction and Cox4 for mitochondria fraction.
[0061 ] Figure 24. DBZIM affected Bcl-2 and IAP protein expression. HeρG2 cells were treated with DBZIM (0, 0.5, 1.0, 1.5 and 2.0 mM) for 24 hr. Total protein was collected using RIPA buffer. 15 μg of total protein was resolved in 4-12% gradient PAGE-SDS gel. The target proteins were recognized by respective antibodies, and visualized by the ECL method. The membrane was stripped and probed for the α-tubulin as the loading control.
[0062] Figure 25. DBZIM induced nucleus to cytoplasm translocation of survivin protein.
(A) Representative immunocytostaining images of HLE cells treated by DBZIM (0 and 1.0 mM) for 24 hr. Arrows indicate the accumulation and the disappearance of survivin in the nucleus of the control and the DBZIM-treated cells, respectively. The images were acquired and analyzed using ArrayScan VTI HCS reader (Thermal Scientific, PA, USA) and Target Activation BioApplication software. (B) Quantitation of translocation event. Cells were grown in 96-well plate and treated by DBZIM of various concentrations for 24 hr before fixation and immunocytostaining for survivin. Lower value in "Cytoplasmic-Nuclear Difference" indicated a lower amount of cytoplasmic survivin.
[0063] Figure 26. DBZIM induced cytoplasm-to-nucleus translocation of AIF. (A). DBZlM translocated ALF from cytoplasm to nucleus. HepG2 cells were treated by DBZIM for 24 hr. Cytoplasmic and nuclear proteins were fractionated using a kit from Pierce. 15 μg of proteins were resolved in 4—12% gradient PAGE-SDS gel. The target proteins were recognized by the antibody of AIF, and visualized by the ECL method. The membrane was stripped and re-probed for α-tubulin as the loading control for cytoplasmic fraction and Tata binding protein TBP for nuclear protein. (B). HLE cells were grown in 96-well plate and treated by DBZIM for 24 hr. The cells were then fixed and stained by antibody of AIF. The images were acquired and analyzed using ArrayScan VTI HCS reader (Thermal Scientific, PA, USA) and Target Activation BioApplication software. The arrows indicate the cytoplasmic and the nuclear location of the ATF staining in the control and the DBZIM, respectively. (C). Quantification of AIF staining in different subcellular compartments. Higher value in "Cytoplasmic-Nuclear . '
Difference" indicated higher nuclear AIF.
[0064] Figure 27. High dose DBZIM induced ROS production in HLE cells. HLE cells were grown in 96-well plate and treated by DBZIM for 24 hr. The cells were then fixed and stained by DHE for quantification of ROS generation. The images were acquired and analyzed using ArrayScan VTI HCS reader (Thermal Scientific, PA5 USA) and Target Activation BioApplication software.
[0065] Figure. 28. DBZIM induced up-regulation of AP-I expression in HepG2 cells. HepG2 cells were treated withDBZM for 24 hr. Cytoplasmic and nuclear proteins were fractionated using a kit from Pierce. 15 μg of nuclear proteins were resolved in 4-12% gradient PAGE-SDS gel. The target proteins were recognized by their respective antibodies, and visualized by the ECL method. The membrane was stripped and re-probed for Tata binding protein TBP as loading control.
[0066] .Figure 29. DMZIM induced apoptosis. Pictures from phase-contrast microscopy show the apoptotic morphology of gastric cancer cell AGS (A) and lung cancer cell H1299 (B) after exposure to DBZM for 72 h, respectively. Extending the length of exposure caused increased loss of confluence with apoptotic cells in cell cultures.
[0067] Figure 30. DBZIM inhibited cell proliferation in glioma cell line C6 (A) and U87 MG (C) and induced caspase 3/7 activity in C6 (B) and U87 MG CD). C6 or U87 cells were incubated with DBZlM of various concentrations for 48 hr for proliferation assay and 6 hr for caspase 3/7 activation assay. Proliferation was measured using MTS Mt from Promega and expressed as relative cell proliferation with control group was normalized to 1. Caspase 3/7 activity was quantified using a kit from Promega and also expressed as relative caspase 3/7 activity of treated groups over control group.
[0068] Figure 31. DBZIM induces cleavage of caspase-3, caspase-9 and PARP as indication of apoptosis. Western blot analysis for the effect of DBZIM, on the cleavage of caspase-3, Caspase-9 and PARP A) in p53 wild type gastric cancer cell AGS and B) in p53 wild type breast cancer cell line MCF-7. Western blot analysis conducted on cells after 72 h exposure to 50 μM of DBZIM.
[0069] Figure 32. DBZIM inhibits phosphorylation of Akt. When AGS, MKN28, H1299 and MCF-7 cells were treated with DBZIM at their corresponding IC50 concentration, phosphorylation at serine 473 of Akt was downregulated in p53 wild type cancer cell line AGS (A), in p53 mutant gastric cancer cell MKN 28 (B), in p53 null Lung cancer cell H1299 (C), and in p53 wild type breast cancer cell MCF-7 (D) . Time course inhibition of phosphorylated-AKT was shown by Western blot, β-actin was used as an internal loading control.
[0070] Figure 33. DBZIM reduced HCC tumor in vivo. Tumors in the HCC-xenografted mice in the control group and in the treatment group, 3 weeks post inoculation, are shown in (A). The tumor growth curve and body weight chart are depicted in (B) and (C), respectively. * P < 0.01.
[0071 ] Figure 34. Endogenous expression of p53. Cell lysates from Hep3B (p53 null), Hep G2 (wild type), HIe and PIc (p53 mutant) were subjected to Western blotting using p53 (DO-I) and actin antibodies.
[0072] Figure 35. Phase-contrast microscopy of cells exposed to IMSs. Pictures from phase-contrast microscopy show cell cultures after exposure to MSs (IBN 15, IBn 19, IBN 24, IBN 25 and IBN 32) for 72 h, respectively. Extending the length of exposure caused increased loss of confiuency in cell cultures.
[0073] Figure 36. IMSs induce apoptosis in Hepatocarcinoma cells. A) Cells were treated with vehicle and IBN 15, IBN 19, IBN 24, IBN 25 and IBN 32 for 72 h, and apoptosis was quantified using Annexin V-FTTC/PI staining followed by flow cytometric analysis. 10,000 cells were detected for every sample. Living cells are in the bottom left quadrants (JhIl1C- and PI-), early apoptotic cells in the bottom right quadrants (FTTC+ and PI-), and late apoptotic cells in the upper right quadrants (apoptotic Jt1Il1C+ and PI+). B) The percentage of apoptotic cells with respect to different MSs.
[0074] Figure 37. IMSs arrest Hepatocarcinoma cells. Cells were treated witih vehicle and IBN 15, IBN 19, IBN 24, IBN 25 and IBN 32 for 72 h, and cell cycle distribution was assessed by flow cytometry. A) Example FACS profiles of propidium iodide-stained control cells, and cells treated with IMSs respectively. B) % of cells in cell cycle phase C) % of cells in subGO population. Each value is the mean + S.D. of three determinations.
[0075] Figure 38. IMSs induce cleavage of caspase-3, caspase-9 and PARP. Western blot analysis for the effect of IBN 15, IBN 19, IBN 24, IBN 25, IBN 32 on the cleavage of caspase-3, Caspase-9 and PARP in HLE cells after 72 h exposure to indicated doses of IMSs.
[0076] Figure 39. IMSs-induced apoptosis was accompanied by accumulation of p53 in the nucleus. HLE cells were treated with the indicated dose of IBN 19 and IBN 24 for 48 h. The treated cells were fixed with 4% formaldehyde, immunostained with anti.-p53 (green fluorescence) and DAPI (blue fluorescence), and analysed by confocal microscopy.
[0077] Figure 40. IMSs-induced accumulation of p53 and p53 phosphorylation on serines 15, 20, 46, and 392 in p53mutant HLE cells. (A) Hepatocarcinoma cancer cell HLE were treated with indicated concentration of IBN 15, IBN 19, IBN 24, IBN 25 and IBN 32 for 72h. The levels of phosphorylation of p53 protein and Ser-15, Ser-20, Ser-46 and Ser-392 were determined by Western blotting, β-actin was used as a loading control.
[0078] Figure 41. IMSs-induced apoptosis through the initiation of the mitochondrial pathway. Cells were treated with various IMSs and the levels of Bcl-2 and BAX were assessed by Western blot assay.
[0079] Figure 42. Real-Time PCR array analysis of p53 signaling pathway-associated gene expression. (A) Anti-apoptotic genes. (B) Growth inhibition gene SESN2. (C) Cell cycle check point genes. (D) Cell proliferation genes. (E) DNA repair gene ATM. The relative level of gene expression was normalized with housekeeping gene GAPDH. Values are expressed as mean ± standard deviation.
[0080] Figure 43. Effect of Compound C (DBZMIM) and compound 9 (MABZMDM) on lung cancer and gastric cancer cells. Compound C (DBZMIM) and compound 9 (MABZM3M) induced morphological change and apoptosis in lung cancer cells MDAMB 231 (A) and in gastric cancer cells MKN 28 (B), 120 h after the treatment.
[0081] Figure 44. Effect of compound 9on HCC-xenografted mice. Tumors in the HCC- xenografted mice in the control group and in the treatment group, 3 weeks post inoculation, were shown in (A). The tumor growth curve and body weight chart was depicted in (B) and (C), respectively. * P < 0.01.
[0082] Table 1. Names and Structures of IMSs.
[0083] Table 2. IC50 values of IMSs.
[0084] Table 3. IC50 values of DBZIM in various cancer cell lines.
[0085] Table 4. IC50 values of IBN-15, 19, 24, 25 and 32 in HLE cells.
[0086] Table 5. IC50 values of DBZMTM and Compound 9 in gastric cancer cells.
[0087] Table 6. IC50 values of DBZMTM and Compound 9 in breast cancer cells.
[0088] Table 7. IC50 values of DBZMTM and Compound 9 in normal breast cells.
DETAILED DESCRIPTION
[0089] The methods described herein relate to the discovery that the imidazolium and imidazolinium compounds (collectively "IMSs") as described herein, including in the form of imidazolium and imidazolinium salts, may be used as anti-fibrotic and anti-cancer agents or to treat fibrotic disease or cancer. The inventors have synthesized novel MSs that may be used in the methods described herein. [0090] Both imidazolium and imidazoliniums are based on an imidazole ring and both are N,N' -substituted; imidazoliums are N5N' -substituted imidazoles, while imidazoliniums are N,N'- substituted imidazolines and do not have the carbon-carbon double bond between positions C4 and C5 that is present in imidazole. Imidazole itself is incorporated in many biological molecules, and synthetic C-substituted imidazoles have become an important part of many pharmaceuticals (Olmos et al. 1999) (Casanovas et al. 2000).
[0091 ] One attractive feature of using MSs in synthetic chemistry is the structural versatility they provide. The electronic structure and stability, and thus the therapeutic safety and efficacy of IMSs, can be fine-tuned by varying the N-substituents (substituents on the nitrogen atoms of the central ring) and the central ring of the molecule. It will be understood that the "central ring" of MSs refers to the five membered ring containing 2 nitrogen atoms (either the imidazole or imidazoline ring) to which various substituents may be bound to create different IMS molecules. IMSs provide inexpensive and chemically tunable building blocks for the development of novel therapeutics.
[0092] Some IMSs are precursors of N-heterocyclic carbenes (NHCs). NHCs can be easily generated from IMSs having a hydrogen atom at the C2 position of the central ring and substituents on both nitrogen atoms of the central ring i.e. two N-substituents. NHCs are generated from these IMSs by the deprotonation of the IMS under the appropriate conditions. Deprotonation of IMSs may be carried out under basic conditions or in a diluted solution under neutral conditions.
[0093] It is difficult to generate NHCs from IMSs with substituents other than a hydrogen atom at the C2 position of the central ring. However radical species may be formed from these IMSs by cleaving the N-substituents.
[0094] The inventors have discovered that the IMSs as described herein may be used as anti- fibrotic or anti-cancer agents or to treat fibrotic disease or cancer. Without being limited to any particular theory, the IMSs as described herein may exhibit anti-oxidative and anti-inflammatory properties. Both fibrotic disease and cancer have been linked to oxidative stress response. In turn, in both fibrotic disease and cancer, oxidative stress has been shown to be closely linked with inflammatory response.
[0095] IMSs that may be used as anti-fibrotic agents or as anti-cancer agents are compounds having a structure of the general formula I:
Figure imgf000022_0001
[0096] In formula I, the dashed line is absent or is present as a bond to form a second bond between the carbon to which R1 and R3 are attached and the carbon to which R2 and R are attached. Thus, the carbon to which R1 and R3 are attached and the carbon to which R2 and R4 are attached may be connected by either a single bond or a double bond.
[0097] R1 and R2 (i) are each independently H, straight or branched Cj-C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C6-C10 aryl; or (ϋ) together with their ring atoms form a 6- to 10-membered fused saturated, unsaturated or aromatic ring system; or (ϋi) R1 and R5 together with their ring atoms or R2 and R6 together with their ring atoms, form a 5- to 10-membered fused saturated, unsaturated or aromatic ring system and the other of R1 and R2 is as defined above in (i); or (iv) R1 and R5 together with their ring atoms and R2 and R6 together with their ring atoms form 5- to 10-membered fused saturated, unsaturated or aromatic ring system.
[0098] R3 and R4 are both H, or, when R1 and R2 together with their ring atoms form a 6- to 10- mmeemmbbeeired fused aromatic ring system or when the dashed line is present as a bond, R and R are absent.
[0099] When R5 or R6 is not fused together with R1 or R2, respectively, as set out above, R5 and R6 are each independently straight or branched Ci-C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C1S cycloalkyl including fused cycloalkyl ring systems, C6- C10 aryl, C6-C10 aryl-Ci-C6 alkyl, C6-C10 aryl-C2-C6 alkenyl, or C6-Ci0 aryl-C2-C6 alkynyl, C1-C6 alkyl-C6-C10 aryl, C2-C6 alkenyl-C6-Cio aryl, or C2-C6 alkynyl-C6-C10 aryl.
[00100] R7 may be H, C1-C6 alkyl, phenyl, substituted C1-C6 alkyl or halo.
[00101 ] Any of the above substituents R1 to R7, where applicable, may optionally have one or more carbon atoms replaced with a heteroatom selected from N, O, S and P. As well, any of the above substituents R1 to R7, where applicable, may optionally be substituted with one or more of straight or branched C1-C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C1S cycloalkyl including fused cycloalkyl ring systems, C6-C10 aryl, fluoro, tri-fluoro- methyl, cyanato, isocyanato, carboxyl, Ci-C6 acyloxy, C1-C6 acyl, carbonyl, amino, acetyl, acetoxy, OXO, nitro, hydroxyl, C1-C6 alkylcarboxy, C1-C6 alkoxy, C2-C6 alkenoxy, C2-C6 alkynoxy.
[00102] One of the ring carbon atom to which R1 and R5 are attached and the ring carbon to which R2 and R6 are attached may be optionally replaced with a nitrogen atom.
[00103] The above compounds may be present in salt form. Thus, MSs that may be used in the present methods include pharmaceutically acceptable salts of compounds of formula I and oligomers and polymers of such salts. Such salts have a structure of the general formula II:
Figure imgf000023_0001
[00104] General formula II is as defined above for general formula I, with the further feature of counterion X. X is a pharmaceutically acceptable anion, including but not limited to chloride, bromide, tetrafluoroborate, hexafluorophosphate.
[00105] The term "fused" as used herein in reference to ring structures refers to the sharing of at least two atoms between ring structures. When two ring atoms (either of which may be C or N) are included in a fused ring system, the ring system is fused to the central imidazolium or imidazolinium ring.
[00106] In particular embodiments, the dashed line may be absent or is present as a bond to form a second bond between the carbon to which R1 and R3 are attached and the carbon to which R2 and R4 are attached; R1 and R2 are both H or together with their ring atoms form a 6- to 10- membered fused aromatic ring system; R3 and R4 are both H or nothing; R5 and R6 are each independently straight or branched C1-C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C18 cycloalkyl including fused cycloalkyl ring systems, C6-C1O aryl, C6- C10 aryl-CrC6 alkyl, C6-C1O aryl-C2-C6 alkenyl or C6-C10 3TyI-C2-C6 alkynyl; R7 is H; any of which substituents R1 to R7, where applicable may have one or more carbon atoms replaced with a heteroatom selected from N, O, S and P and any of which may optionally be substituted with one or more of straight or branched C1-C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C18 cycloalkyl including fused cycloalkyl ring systems, C6-C1O aryl, fluoro, tri- fϊuoro-methyl, cyanato, carboxyl, carbonyl, amino, acetyl, oxo, nitro, Ci-C6 alkoxy, C2-C6 alkenoxy, C2-C6 alkynoxy. [00107] For example, in various embodiments, the compound may be an imidazolium salt, with a double bond between the two ring atoms to which R1 and R2 are attached. In various other embodiments, the compound may be an imidazolinium salt, wherein R3 and R4 are hydrogens and there is only a single bond between their ring carbons. In yet other embodiments, R3 and R4 are nothing and R1 and R2 together with their ring atoms form a 6- to 10-membered fused aromatic ring system.
[00108] The substituents comprising R5 and R6 may also vary. For example, in some embodiments, the IMS of the present method may be a compound wherein R5 and R6 are the same. In different embodiments, R5 or R6 may be aralkyls, branched alkyls or cycloalkyls including fused ring systems. Ih other embodiments R5 or R6 may comprise a phenalkyl group. In other embodiments, one or both of R5 and R6 may comprise an adamantanyl group.
[00109] IMSs that may be used in the present method also include oligomers or polymers formed from compounds of general formula I. Thus, "oligomer" as used herein, refers to, for example, 2 or more, 3 or more, 4 or more, 5 or more, 10 or more, 15 or more, 20 or more, 25 or more, 50 or more, 100 or more, 150 or more compounds of general formula I connected together in the manner described below. Thus, oligomer includes dimers and trimers. "Polymers" as used herein refers to, for example, 100 or more, 150 or more, 200 or more, 250 or more, 500 or more, 1000 or more compounds of general formula I connected together in the manner described below.
[00110] In oligomers or polymers as used herein, compounds of general formula I are connected to each other to form a macromolecular structure, which may be cyclised.
[00111] For example, trimers as described herein include 1 ,3 ,5-tris(4-methyl-imidazolium)- linked cyclophane or a pharmaceutically acceptable salt thereof (TDBZIM).
[00112] In the macromolecular structure, two compounds of general formula I may be connected together so that one or more R1, R2, R5, R6 or R7 substituent is bivalent and is thus shared between the two compounds, rather than have a relevant substituent present in the oligomer or polymer for every compound of general formula I present. For example, in such a dimer, two compounds may be connected by a shared bivalent R1 substituent, and thus in the complete dimer, there is only one occurrence of an R1 substituent for the two compounds of general formula I.
[00113] Thus, the compounds of general formula I may be j oined to each other via a bivalent substituent such as R1, R2, R5, R6, or R7. For example, an R5 substituent on one compound of general formula I may also be R5 on a second compound of general formula I such that the two compounds are linked via a shared R5 substituent. Similarly, oligmers or polymers may be formed by linking compounds via shared R1, R2, R5, R6, or R7 substituents. For example, dimers of this description include 2,6-di-(3-benzyl-imidazolium bromide)-pyridine or a pharmaceutically acceptable salt thereof (IBN-22).
[00114] Sharing of substituents as described above can include sharing of two substituents that together with their ring carbons form a fused ring system. Thus, for example, if R and R together with their ring carbons form a ring system on a first compound of formula I, additional carbons within the ring will be ring carbons from a second compound of general formula I. For example, dimers of this description include benzo(l,2-4,5-diimidazolium)-N,N',N'', N"'- tetrabenzyl-di-bromide or a pharmaceutically acceptable salt thereof (IBN-29).
[00115] Alternatively, a substituent such as one or more of R1, R2, R5, R6 or R7 may be bonded to another such substituent on a second compound in order to connect two compounds together to form an oligomer or polymer, and thus there will be one occurrence of the relevant Unking substituent for each compound of general formula I in the oligomer or polymer. Multiple compounds falling within general formula I may be joined together (oligomerised) in this way, by one or more shared bivalent R1, R2, R5, R6 or R7 substituents. For example, such dimers include 2,2' -di-(3 -benzyl-imidazoHum bromide)- 1,1' -binaphthalene or a pharmaceutically acceptable salt thereof (IBN-23).
[00116] Additionally, two compounds of general formula I may be attached by a single or double bond between respective carbon atoms to which R7 is attached in general formula I to form a dimer. These molecules include for example, l,3-dibenzyl-2-(l,3-dibenzyl-lH-imidazol-2(3H)- ylidene)-2,3-dib.ydro-lH-imidazole or a pharmaceutically acceptable salt thereof (compound H).
[00117] The oligomerised or polymerised compounds may be the same compound, such that all substituents in general formula I are the same in each oligomerised or polymerised compound, or may be different, with one or more substituents differing between compounds, with the exception that shared bivalent substituent or substituents will obviously be the same in the compounds in which the bivalent substituent is shared.
[00118] Table 1 contains the names and structures of particular examples of the IMSs described herein.
[00119] In particular embodiments, the compounds of the present invention may be 1-ethyl- 3-methylimidazolium, 1,3-bisbenzylimidazolium, 1,3-dϋsopropylimidazolium, 1,3-di-tert- butylimidazolinium, l,3-bis(l-adamantyl)imidazolium, l,3-bis(2,4,6-trimethylphenyl)- imidazolinium, l,3-bis(2,6-dusopropyl-phenyl)-imidazolMum, 1,3-diallylimidazolium, l-benzyl-3- methylimidazolium, l-butyl-3-methylimidazolium, l-(l-adamantyl)-3-(2,4,6-trimethylphenyl)-4,5- dihydroimidazolium, 2-benzylimidazo[l,5-α3quinoIinium, l,3-bis(l-adamantyl)-benzimidazolium, l^-dicyclohexylbenzimidazolium, 1,3-diisopropylimidazoliniuni tetrafluoroborate, 1,3- diisopropylimidazolium, 2-(2,6-dusopropylphenyl)-5-methylimidazo[l ,5-α]pyridinium, l-(2,6- diisopropylphenyl)-3 -(2,4,6-trimetih.ylphenyl)-imidazolinium, 2-mesityl-5- methylimidazo[l,5- σ]pyridinium, 2-mesityl-2,5,6,7-tetrahydropyrrolo[2,l-c][l,2,4]triazol-4-ium, 1,3-Ms(I- adamantyl)imidazolinium, l-butyl-3-(2-pyridinylmethyl)-lH-imidazolium, 6,7-dlhydro-2- pentafluorophneyl-5Η-pyrrolo[2,l-c]-l52,4-trizolium, or any pharmaceutically acceptable salt thereof.
[00120] Ih particular embodiments, the compounds of the present invention may be 1-ethyl- 3-methylimidazolium bromide, 1,3-bisbenzylimidazolium bromide (DBZIM), 1,3- dϋsopropylimidazolium tetrafluoroborate (DPIM), 1,3-di-tert-butylimidazoliniumtetrafluoroborate (DBIM), l,3-bis(l-adamantyl)imidazolium tetrafluoroborate (AMIM), l,3-bis(2,4,6- ttimemylphenyl)-imidazolimum chloride (TMPHIM), l,3-bis(2,6-diisopropyl-phenyl)- imidazolinium chloride (DPPHIM), 1,3-diallylimidazolium bromide (Compound D), l-benzyl-3- methylimidazoKum bromide (Compound E), l-butyl-3-methylimidazolium chloride (Compound G), l-(l-adamantyl)-3-(2,4,6-tiimemylphenyl)-4,5-dihydroimidazolium chloride (Compound Sl), 2- benzylimidazo[l,5-α]quinolimum chloride (Compound S2), l,3-bis(l-adamantyl)-benzimidazolium chloride (Compound S3), l.S-dicyclohexylbenzimidazolium chloride (Compound S6), 1,3- dϋsopropylimidazolinium tetrafluoroborate (Compound S7), 1,3-diisopropylimidazolium chloride (Compound S8), 2-(2,6-dusopropylpheneyl)-5-metiιyuinidazo[l,5-α]pyridinium hexafluorophosphate (Compound S9), l-(2,6-diisopropylphenyl)-3-(2,4,6-trimethylphenyl)- imidazolinium chloride (Compound SlO), 2-mesityl-5- methylimidazo[l,5-α]pyridinium chloride (Compound Sl 1), 2-mesityl-2,5,6,7-tetrahydropyrrolo[2,l-c][l,2,4]triazol-4-ium chloride (Compound S12), l,3-bis(l-adamantyl)imidazolinium tetrafluoroborate (Compound S13), 1-butyl- 3-(2-pyridinyhnethyl)-lH-imidazolium hexafluorophosphate (Compound S 14), 6,7-dihydro-2- pentafluorophneyl-5Η-pyrrolo[2,l-c]-l,2,4-trizolium tetrafluroborate (Compound S15).
[00121 ] The compounds of general formula I include novel compounds, including l-ethyl-3- methylimidazolium, l-methyl-3-(2-hydroxylethyl)-imidazolium,. l-methyl-3-(4-isocynatobenzyl)- imidazolium, l-methyl-3-(4-carboxylbenzyl)-miidazolium, l-methyl-3-(4-acetate-benzyl)- imidazolium, l-methyl-3-(2,2-dimethoxylethyl)-imidazolium, l-(2,4,6-trimethylphenyl)-3-(4- acetate-benzyl)-iπudazolium, l,3-Dibenzyl-5-phenylimidazolium, l-benzyl-3-(4-carboxylbenzyl)-2- methylimidazolium, l-benzyl-3-(3,4,5-trime1hoxylbenzyl)-2-methylimidazolium, l-benzyl-3-(4- acetate-benzyl)-2-methyl-imidazolium, l-benzyl-3-(4-memylcarboxylatebenzyl)-2-methyl- imidazolium, l-benzyl-3-(2,2-dimethoxylethyl)-2-meth.yl-imidazoliιιm, 2,6-di-(3-benzyl- imidazolium)-pyridine, 2,2'-di-(3-benzyl-imidazolium)-l,r-binaphliialene, l-benzyl-3-(4- methylbenzyl)-imidazolium, l-benzyl-3-(2-trifluorometiiylbenzyl)-2-methylinτidazolium, 1-benzyl- 3-(4-methylcarboxylatebenzyl)-5-ρhenyl-imidazolinm, l-benzyl-3-(4-acetatebenzyl)-5-phenyl- imidazolium, l-benzyl-3-(4-methylbenzyl)-5-phenylimidazolium, (1,2-4,5-diimidazolinm)- N,N' ,N",N'"-tetrabenzyl-benzene, l-benzyl-3-(2-propyn-l-yl)-imidazoh"um, l-benzyl-3-(3- hydroxyl-propyl)-imidazolium, 1 ,3-di(2-phenylethyl)-imidazolium, l-benzyl-3-(4-acetatebenzyl)- imidazolium, l-benzyl-3-(pyridin-2-yl)-imidazolium, l,3,5-Ms-(4-methyl-imiazolium)-linked cyclophane, l,3-dibenzyl-2-(l,3-dibenzyl-lH-imidazol-2(3H)-ylidene)-2,3-dihydro-lH-iinidazole, l-benzyl-3-methyl-imidazolium, l-(4-cyanatobenzyl)-3-metb.yl-iroidazolium, l-(4-carboxybenzyl)- 3-methyl-imidazo]ium, l-methyl-3-(4-acetate-benzyl)-imidazolium, l-methyl-3-(2,2- ditnethoxyetb.yl)-imidazolium, and l-(2,4,6-trimetiιylphenyl)-3-(4-acetate-benzyl)-iinidazolium, or any pharmaceutically acceptable salt thereof.
[00122] The compounds of general formula I include novel compounds, including l-ethyl-3- methylimidazolium bromide, l-methyl-3-(2-hydroxylethyl)-rmidazolium bromide (IBN-2), 1- methyl-3-(4-isocynatobenzyl)-imidazolium chloride (IBN-3), l-methyl-3-(4-carboxylbenzyl)- imidazolium bromide (IBN-4), l-metiiyl-3-(4-acetate-benzyl)-imidazolium chloride (IBN-6), 1- methyl-3-(2,2-dimethoxylethyl)-imidazolium bromide (IBN-8), l-(2,4,6-trimethylphenyl)-3-(4- acetate-benzyl)-imidazolium chloride (IBN-9), l,3-Dibenzyl-5-phenylimidazolium bromide (IBN- 15), l-benzyl-3-(4-carboxylbenzyl)-2-methylimidazolium chloride (IBN-17), l-benzyl-3-(3,4,5- trimethoxylbenzyl)-2-methylimidazolium chloride (IBN-18), l-benzyl-3-(4-acetate-benzyl)-2- methyl-imidazolium chloride (IBN-19), l-benzyl-3-(4-methylcarboxylatebenzyl>2-methyl- imidazolium chloride (EBN-20), l-benzyl-3-(2,2-dimethoxylethyl)-2-methyl-imidazolium bromide (IBN-21), 2,6-di-(3-benzyl-imidazolium bromide)-pyridine (IBN-22), 2,2'-di-(3-benzyl- imidazolium bromide)- l,l'-binaphthalene (IBN-23), l-benzyl-3-(4-methylbenzyl)-imidazolium chloride (IBN-24), l-benzyl-3-(2-trifluoromethylbenzyl)-2-methylimidazolium chloride (IBN-25), l-benzyl-3-(4-methylcarboxylatebenzyl)-5-phenyl-imidazolium bromide (IBN-26), l-benzyl-3-(4- acetatebenzyl)-5-phenyl-imidazolium bromide (TBN-27), l-benzyl-3-(4-methylbenzyl)-5- phenylimidazolium chloride (IBN-28), Benzo(l,2-4,5-dϋmidazolium)-N,N',N",N'"-tetrabenzyl-,di- bromide (IBN-29), l-benzyl-3-(2-propyn-l-yl)-imidazolium bromide (ffiN-30), l-benzyl-3-(3- hydroxyl-ρropyl)-imidazolium bromide (TBN-31), l,3-di(2-phenylethyl)-imidazolium bromide O-BN-32), l-benzyl-3-(4-acetatebenzyl)-imidazolium chloride (IBN-33), l-benzyl-3-(pyridin-2-yl)- imidazolium bromide (IBN-34), l,3,5-tris-(4-metliyl-iiniazolium)-linked cyclophane'3Br (TDBZIM), l,3-dibenzyl-2-(l,3-dibenzyl-lH-imidazol-2(3H)-ylidene)-2,3-dihydro-lH-imidazole (compound BQ, l-benzyl-3-methyl-imidazolium bromide (compound 12), l-(4-cyanatobenzyl)-3- methyl-imidazolium chloride, l-(4-carboxybenzyl)-3-methyl-imidazolium bromide, l-methyl-3-(4- acetate-benzyl)-imidazolium chloride, l-methyl-3-(2,2-dimethoxyethyl)-imidazolium bromide, and l-(2,4,6-fiimeraylphenyl)-3-(4-acetate-benzyl)-irnidazolium chloride.
[00123] The compounds of general formula I and oligomers and polymers thereof are commercially available or may be synthesized using routine chemistry, as described in the Examples set out below. Methods of synthesis have also been described in Harlow et al. 1996, Zhang et al. 2007; Chianese and Cratree 2005 and Boydston et al. 2005.
[00124] Thus, there is presently provided a method for delivering an anti-fibrotic or anticancer agent to a cell, the method comprising contacting the cell with an effective amount of a compound of general formula I, a pharmaceutically acceptable salt thereof or an oligomer or polymer thereof.
[00125] As used herein, "delivering" an anti-fibrotic or anti-cancer agent to a cell refers to providing the agent in sufficiently close proximity to the cell such that the agent can exert its anti- fibrotic or anti-cancer effects on the cell. In vitro, for example, the agent may be delivered to the cell by adding the agent to the cell culture media. In vivo, for example, the agent may be delivered by administering the agent to a subject as a pharmaceutical composition.
[00126] As used herein, "contacting" a cell refers to direct and indirect contact with the cell.
Direct contact refers to a direct interaction between the agent and the cell. In contrast, indirect contact involves "contacting the cell" via interactions with other molecules or compounds. For example, the agent may interact with a molecule, affecting a change in that molecule that causes it to interact with the cell or interact with another molecule which will then interact with the cell to exert an effect. Indirect contact may involve a series of molecular interactions resulting in an effect on the cell.
[00127] An anti-fibrotic agent as used herein refers to a compound that alters, reduces or inhibits molecules, mechanisms or effects associated with fibrotic disease. For example, the agent may be an anti-inflammatory or an anti-oxidative, induce apoptosis, alter, reduce or inhibit the activity of fibrogenic mediators such as TGF-β, NF-κB or TL-6 or neutralize proliferative, fibrogenic, contractile or pro-inflammatory responses of cells. Without being limited to any particular theory, the anti-fibrotic agent may reduce or inhibit fibrotic disease by: (a) reducing inflammation to avoid stimulating activation of cells involved in fibrotic disease such as stellate cells, (b) directly down-regulating activation of cells involved in fibrotic disease, (c) neutralizing proliferative, fibrogenic, contractile or pro-inflammatory responses of cells involved in fibrotic disease, (d) inducing apoptosis such as, for example, Bcl-xL or Fas; or (e) inducing ECM degradation.
[00128] An anti-cancer agent as used herein refers to a compound that alters, reduces or inhibits molecules, mechanisms or effects associated with cancer. For example, the agent may be an anti-inflammatory or an anti-oxidative, induce apoptosis, inhibit abnormal cell growth, inhibit cell proliferation, inhibit DNA mutation or modify the activity or level of activity of molecules associated with cancer such as survivin, caspase 9, caspase 3, BCI-XL, Bak, BAX, ATM, p53, Cdc25A, Cdc2, SESN2 and AP-I .
[00129] The cell to which the anti-fibrotic or anti-cancer agent is to be provided may be any cell, including an in vitro cell, a cell in culture, or an in vivo cell within a subject. The term "cell" as used herein refers to and includes a single cell, a plurality of cells or a population of cells where context permits, unless otherwise specified. The cell may be an in vitro cell including a cell explanted from a subject or it may be an in vivo cell in a subject. Similarly, reference to "cells" also includes reference to a single cell where context permits, unless otherwise specified.
[00130] The cell may be derived from any organism, for example an insect, a microorganism including a bacterium, or an animal including a mammal including a human.
[00131] The cell of the present method may be within a subject having fibrotic disease or cancer, a subject requiring treatment for fibrotic disease or cancer or a subject in which prevention of fibrotic disease or cancer is desired. In some embodiments, the subject is a human subject.
[00132] Fibrotic disease will be understood by those skilled in the art to refer to a condition which may be characterized or caused by the development of excess fibrous tissue or over production of extracellular matrix in an- organ or tissue and may include for example liver fibrosis, kidney fibrosis, lung fibrosis, bone fibrosis., systemic sclerosis, mixed connective tissue. Father examples of fibrotic disease are provided in US 2007/0043016 which is herein incoporated by reference.
[00133] In one embodiment of the present methods, an anti-fibrotic agent may be delivered to a cell in a subject having hepatic fibrosis. The compound may be for example 1,3- diisopropylimidazolium or a pharmaceutically acceptable salt thereof.
[00134] A skilled person will understand cancer to encompass a class of diseases in which cells exhibit abnormal cell growth and the potential to invade nearby tissues. In some forms of cancer, the abnormal cells may also spread to other locations in the body. Different types of cancer include for example, breast cancer, colorectal cancer, brain cancer, prostate cancer, cervical cancer, ovarian cancer, bone cancer, skin cancer, lung cancer, pancreatic cancer, bladder cancer, gallbladder cancer, kidney cancer, esophageal cancer, Hodgkin lymphoma, Non-Hodgkin lymphoma, laryngeal cancer, leukemia, multiple myeloma, oral cancer, pleural mesothelioma, small intestine cancer, testicular cancer, uterine cancer, thyroid cancer and stomach cancer.
[00135] In one embodiment of the present methods, an anti-cancer agent may be deliverd to a cell in a subject having hepatocellular carcinoma, lung cancer, breast cancer, stomach cancer or glioma. The compound may be for example 1,3-Bisbenzylimidazolium, 1, 3,-Dibenzyl-2- methylimidazoHum,l-(2,4,6-trimethylphenyl)-3-(4-acetate-benzyl)-mαidazoHum, l,3-Dibenzyl-5- phenylimidazolium , l-benzyl-3-(4-acetate-benzyl)-2-methyl-imidazolium, l-benzyl-3-(4- methylbenzyl)-imidazolium, l-benzyl-3-(2-trifluoromemylbenzyl)-2-methylimidazolium or 1,3- di(2-phenylethyl)-imidazolium, or a pharmaceutically acceptable salt thereof.
[00136] -The term "effective amount" as used herein means an amount effective, at dosages and periods of time necessary to achieve the desired result, for example to treat fibrotic disease or cancer. The total amount of IMS to be administered will vary, depending on several factors, including the severity and type of the disorder, the mode of administration, and the age and health of the subject. Methods for determining an effective amount of a particular IMS for treating fibrotic disease or cancer will be readily apparent to a person skilled in the art.
[00137] "Treating" fibrotic disease or cancer refers to an approach for obtaining beneficial or desired results, including clinical results. Beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disorder or disease, stabilization of the state of disease, prevention of development of disorder or disease, prevention of spread of disorder or disease, delay or slowing of disorder or disease progression, delay or slowing of disorder or disease onset, amelioration or palliation of the disorder or disease state, and remission (whether partial or total). "Treating" can also mean prolonging survival of a subject beyond that expected in the absence of treatment. "Treating" can also mean inhibiting the progression of disorder or disease, slowing the progression of disorder or disease temporarily, although more preferably, it involves halting the progression of title disorder or disease permanently.
[00138] To aid in administration, the IMS may be formulated as an ingredient in a pharmaceutical composition. The compositions may contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives and various compatible carriers or diluents.
[00139] The proportion and identity of the pharmaceutically acceptable carrier is dependant on a variety of factors including the chosen route of administration, compatibility with the IMS molecule and standard pharmaceutical practice. Generally, the pharmaceutical composition will be formulated with components that will not significantly impair the biological properties of the IMS.
[00140] Suitable vehicles and diluents are described, for example, in Remington's
Pharmaceutical Sciences (Remington, The Science and Practice of Pharmacy, 21st edition, Lippincott Williams & Wilkins, Philadelphia, PA., 2006). It would be known to a person skilled in the art how to prepare a suitable pharmaceutical composition.
[00141 ] The pharmaceutical composition may be administered to a subject in a variety of forms depending on the selected route of administration, as will be understood by those skilled in the art. The composition of the invention may be administered for example, by oral administration, surgically or by injection to the desired site.
[00142] In different embodiments, the composition is administered by injection (subcutaneously, intravenously, intramuscularly, etc.) directly at a desired site, for example in the vicinity of fibrotic disease or cancer that is to be treated.
[00143] The dose of the pharmaceutical composition that is to be used depends on the particular fibrotic disease or cancer disorder being treated, the severity of the condition, individual patient parameters including age, physical condition, size and weight, the duration of the treatment, the nature of concurrent therapy (if any), the specific route of administration and other similar factors that are within the knowledge and expertise of the health practitioner. These factors are known to those of skill in the art and can be addressed with minimal routine experimentation.
[00144] It will be understood that pharmaceutical compositions may be provided in a variety of dosage forms and thus, in different embodiments, IMSs may be administered in different dosage forms including for example pills, tablets, capsules, solutions, suspensions, powder and injections. Conventional procedures and ingredients for preparing and administering the different dosage forms would be known to a skilled person and are described for example, in Remington's Pharmaceutical Sciences (Remington, The Science and Practice of Pharmacy, 21st edition, Lippincott Williams & Wilkins, Philadelphia, PA., 2006).
[00145] Uses of the IMSs as described herein, including compounds having a structure of general formula I, a pharmaceutically acceptable salt thereof or an oligomer or polymer thereof for delivering an anti-fibrotic or anti-cancer agent to a cell in vivo and in the preparation of a medicament for delivering an anti-fibrotic or anti-cancer agent to a cell in vivo are also contemplated.
[00146] The IMSs of the present method may exert their anti-fibrotic and anti-cancer effect as a result of anti-oxidative properties, which may function via a multitude of molecular mechanisms. Anti-oxidarits generally exert their effect mainly through three different pathways: (1) neutralization of cellular free radical ROS generated during metabolism and immune response, (2) induction of endogenous anti-oxidative enzymatic activity, and (3) chelation of iron or copper ions that catalyze the generation of hydroxyl radical. Without being limited to any particular theory, it appears that the anti-oxidative properties of the IMSs may result, at least in part, due to their activity as a radical scavenger. It appears that the neutralization of free radicals by IMSs may occur after a- series of chemical reactions including (1) the spontaneous conversion of MSs to NHCs preferably under a basic condition, (2) the interaction of the NHCs at the carbon 2-position with free radicals such as ROS resulting in the formation of intermediate active radicals and the neutralization of the free radicals.
[00147] The present inventors have discovered that the IMSs described herein also exhibit anti-inflammatory properties. Without being limited to any particular theory, the dual anti-oxidative and anti-inflammatory properties of the IMSs may be attributable, at least in part, to the interplay between oxidative stress and inflammation. For example, it is hypothesized that in the treatment of liver fibrosis, the anti-inflammatory effects of the IMSs compounds defined herein may be attributable to the IMSs' anti-oxidative properties that inhibit HSC activation and thus the resulting . production of pro-inflammatory cytokines. Similarly, without being limited to any particular theory, the inhibition of inflammatory- mediator IL-6 by an IMS may be partially attributable to the anti- oxidative properties of the IMS which suppress the activation of transcription factor NF-κB, an important regulator of the secretion of inflammatory cytokines.
[00148] In addition to their anti-oxidative and anti-inflammatory properties, the IMSs may exhibit other effects that are useful in treating fibrotic disease and cancer. Anti-oxidants have been found to also have apoptosis-inducing properties. For example, EGCG, a green tea extract has shown to induce apoptosis of human hepatocellular carcinoma in cultured cell lines and a xenograft model (Nishikawa et al. 2006) while garlic extract, a natural anti-oxidant, has also been shown to induce apoptosis in human glioblastoma cells (Das et al. 2007) and colon carcinoma cells (Jakubilova et al. 2006) by inducing oxidative stress.
[00149] Without being limited to any particular theory, it appears that the MSs may be effective in treating cancer due to inducement of apoptosis and inhibition of cell proliferation through cell cycle arrest. For example, IMSs may trigger apoptosis and cell cycle arrest by increasing or decreasing the activity and expression of molecules involved in apoptosis and cell cycling including for example survivin, caspase 9, caspase 3, BCI-XL, Bak, BAX, ATM, p53, Cdc25A, Cdc2, SESN2 and AP-I.
[0015OJ The present methods and compounds are further exemplified by way of the following non-limited examples.
[00151] EXAMPLES
[00152] Materials and Methods
[00153] Anti-oxidative, anti-inflammatory and anti-fibrotic properties of IMSs
[00154] Synthesis and Characterization of IMSs
[00155] DBZM, TDBZM and DBZBM were synthesized in-house. DPM, DBM, AMIM, TMPHM and DPPHIM were purchased from Sigma Chemicals (USA). The chemical structures of DBZM (1,3-bisbenzylimidazolium bromide), DBZBM (1,3-bisbenzyl-benzimidazolium bromide) and TDBZM (l,3,5-tris(4-methyl-imidazolium)-Unked cyclophane-3Br) are illustrated in Table 1. DBZM and DBZBM were synthesized using a method disclosed in the literature (Harlow et al. 1996). TDBZIM was synthesized by mixing 4-methylimidazole (123 mg, 1.5 mmol) and 2,4,6- tris(bromomethyl)mesitylene (400 mg, 1 mmol) in 200 ml of N,N'-dimethylformamide (DMF) in a reaction vial. The reaction mixture was heated to 1000C for 2 days. Colorless crystals of TDBZIM were precipitated, and collected in 40% yield (160 mg). Nuclear magnetic resonance was conducted with the following results: 1H nuclear magnetic resonance (NMR) (400 MHz, CD3OD): δ 7.77 (s, IH), 5.53 (s, 2H), 5.41 (s, 2H), 4.60 (s, IH), 2.55 (s, 3H), 2.19 (s, 6H); 13C NMR (100 MHz, CD3OD): δ 143.49, 136.27, 131.73, 131.54, 123.45, 67.06, 16.35, 9.57. Elemental analysis of TDBZlM (C36H45Br3N6) provided the following results: C, 53.21; H, 5.88; N, 10.12 (calc. C, 53.95; H, 5.66; N, 10.49). [00156] Primary HSC Isolation and HSC-T6 Line
[00157] The isolation of primary HSCs from Wistar rat liver was performed primarily following the method provided in a previous report (Weiskirchen et al. 2005). Briefly, the supernatant of cell suspension after hepatocyte removal was washed and re-suspended in 9.5 ml of Gey's balance salt solution (GBSS), and mixed with 8 ml of 28.7% (w/v) Histodenz in GBSS. The gradient was prepared by laying the cell suspension underneath 6 ml of GBSS, and centrifuged in Histodenz gradient at 1400xg for 20 mm. The stellate cells were separated into a fuzzy band just above title interface of the Histodenz solution and the aqueous buffer. The stellate cell band was harvested, washed, and IxIO6 cells/well were seeded in a 6-well culture plate in Dulbecco modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS). The viability of the HSCs was > 95% as determined by trypan blue exclusion staining, and the purity of the HSCs was > 90% as assessed by glial fibrillary acidic protein (GFAP) positive staining in > 200 cells counted. Primary HSCs were routinely cultured in DMEM with 10% FBS in a humidified CO2 incubator at 37°C, and split at 1:4 ratio by trypsinization (0.05% trypsin/0.53 mM ethylenediaminetetraacetic acid (EDTA)) when they grew to confluency. The HSC-T6 cell line was kindly provided by Dr. Scott Friedman of Mount Sinai School of Medicine, New York. A stable T6/GFAP-LαcZ clonal cell line was established as previously reported (Maubach et al.2006). HSC-T6 and T6/GFAP-Z,αcZ cells were cultured in the same conditions as the primary HSCs.
[00158] Cell Treatment with IMSs
[00159] Stock solutions of IMSs were prepared either in dimethylsulfoxide (DMSO) or H2O depending on their solubility. The final DMSO concentration in the culture medium was kept below 0.2% (v/v). NAC was obtained from Merck KGaA (Germany), and EGCG was purchased from Sigma and used directly without further purification. Cells were initially seeded in cell culture plates or flasks in DMEM supplemented with 10% FBS for 18-24 h before the addition of compounds of various concentrations for different time periods. Details of the each treatment can be found in the figure legends.
[00160] Cellular ROS Determination
[00161 ] The ROS level of treated HSC-T6 cells was determined using the dichlorofluorescein (DCF) labeling method (Molecular Probes Inc., OR, USA). Cultured cells were harvested by trypsinization, and resuspended in phenol red-free DMEM Qhvitrogen). Cells were then incubated with 10 μg/ml of 2' ,7'-dichloro-fluorescein diacetate (DCFH-DA) in DMEM for 15 min before resuspension in DMEM for assaying. Cell solution was dispensed into a black-wall 96-well pate in triplicate for fluorescence readout at Ex = 485 nm and Em = 530 nm with a Tecan Safire 31 plate reader. The fluorescence unit was normalized against viable cell numbers, which were determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT assay, Promega, WI, USA). Cellular ROS level was expressed relative to the control sample.
[00162] Glutathione (GSH) and GSH/Disulflde Dimer of Glutathione (GSSG) Assays
[00163] Total cellular GSH and GSSG levels were determined by using assay kits from Cayman Chemical (Ann Arbor, MI, USA) following the manufacturer's instructions. Briefly, HSC- T6 cells treated with DBZIM, TDBZIM, NAC, or EGCG were scalped and homogenized in phosphate buffered saline (PBS) by sonication at 60% frequency for 30 s, and the supernatant was collected after centrifugation at 16,000xg for 10 min at 40C. Protein content of the samples was determined by bicinchonicic acid(BCA) assay.
[00164] For GSH assays, the protein sample was deproteinated by metaphosphoric acid (Aldrich, Catalog No. 23927-5), and the pH was adjusted by triethanolamine (Aldrich, Catalog No. T5830-0) according to the manual. The sample was ready for assaying total GSH including both the reduced and oxidized forms. Quantification of GSSG5 exclusive of GSH, was performed by first derivatizing GSH with 2-vinylpyridine and assaying separately. Colorimetric signal was recorded at 405 nm with a Tecan Safire π plate reader. The levels of GSH or GSSG were determined as nmol/μg protein.
[00165] Glutathione Peroxidase (GPx), Catalase (CAT) and Superoxide Dimutase (SOD)
Assays
[00166] Activity levels of three anti-oxidant enzymes, GPx, CAT and SOD, were determined using assay kits from Cayman Chemical. To assay GPx and CAT activities, protein samples were harvested in ice-cold PBS containing 1 mM of EDTA. To assay SOD activity, protein samples were prepared in 20 mM of N-(2-hydroxyethyl)-piperazine-N'-2-ethanesulfonic acid (HEPES) buffer containing 1 mM of ethylene glycol-bis(2-aminoethylether)-N,N,N',N'-tetraacetic acid (EGTA), 210 mM of mannitol and 70 mM of sucrose. Protein content was determined by BCA assay.
[00167] GPx activity was indirectly measured by a coupled reaction with GSH reductase. Oxidized GSH, produced upon reduction of H2O2 by GPx, is recycled to its reduced state by glutathione reductase (GR) and nicotinamide adenine dinucleotide phosphate (ΝADPH). The oxidation of ΝAPDH is proportional to the GPx activity, and can be measured as a decrease in absorbance at 340 nm. GPx activity was expressed as nmol/min/mg protein. |O0168] CAT activity was determined based on reaction of the enzyme with methanol in the presence of H2O2. The formaldehyde produced was measured spectrophotometrically at 540 nm using Purpald as the chromogen. CAT activity was expressed as nmol/rnin/mg protein.
[00169] Total SOD, including all three types of SOD (Cu/Zn-, Mn- and Fe7SOD), was quantified using a tetrazolium salt to detect superoxide radicals generated by xanthine oxidase and hyposanthine. One unit of SOD was defined as the amount of enzyme needed to exhibit 50% dismutation of the superoxide radical. SOD activity was expressed as U/mg protein.
[00170] Glutathione s-Transferase (GST) Assay
[00171 ] The activity of phase π anti-oxidant enzyme, GST, was assayed using a kit from Cayman Chemical. Protein samples were harvested in PBS buffer containing 1 mM of EDTA, and the protein concentration was determined by BCA assay. Total GST (cytosolic and microsomal) activity was assayed by measuring the conjugation of l-chloro-2,4-dinitrobenzene (CDNB) with reduced GSH5 and monitored by an increase in absorbance at 340 nm. GST activity was expressed as nmol/min/mg protein.
[00172] Total RNA Isolation and Real-Time RT-PCR
[00173] Total RNA was isolated using NucleoSpin RNAII isolation kit (Nacherey & Nagel, Germany), and quantified by ND-100 spectrophotometer (Nanodrop Technologies, DE, USA). The details of real-time RT-PCR were reported elsewhere (Zhang et al. 2006). Briefly, it was a two-step real-time RT-PCR using Taqman chemistry. Total RNA was first reverse transcribed to cDNA, and real-time PCR was run and detected in ABI 7500 Fast Real-Time PCR System (Applied Biosystems, CA, USA). Primers and probes for rat GFAP, SMAA, collagen IaI, fibronectin, TGF- βl, TGF-β receptor I (TGFβ RI) and TL-6 were ordered from Taqman's assay-on-demand database. Rat β-actin was used as normalization gene, and its expression level was constant (Or difference was < ±0.5) throughout the experimental settings in this study. Relative quantification of target mRNA was calculated using comparative threshold cycle method (AACi) as described in User Bulletin #2 (ABI Prism 7700 Sequence Detection System). Relative quantification was given by 2" MCT to express the up-regulation or down regulation of the target gene under the treatments compared'to the control.
[00174] Western Blotting
[00175] Protein extracts were prepared by lysing a cell pellet in 10 volumes of boiled 1 % sodium dodecyl sulfate (SDS) (90-950C) for 10 min for GFAP, or using NE-PER nuclear and cytoplasmic extraction reagents from Pierce (IL, USA) for nuclear proteins, or RIPA (Pierce, IL, USA) for SMAA, fibronectin, collagen αI(T), TGF-βl and TGFβ RI. Protein content was quantified using a BCA kit (Pierce, IL, USA). 15 μg of total protein were resolved in 4-12% gradient SDS/polyacrylamide gel electrophoresis (PAGE) gel, and transferred to a nitrocellular membrane. The membrane was then probed with primary antibodies against GFAP (Dako, Denmark), SMAA (Sigma, USA), procollagen αl(l), fibronectin, c-Jun, c-Fos, JunB, JunD, Fra-1, Fra-2 (Santa Cruz Biotechnology, CA, USA), NF-TCB P65, TGF-βl and TGFβ RI (Abeam, UK), and detected by horseradish peroxidase-conjugated secondary antibody (Santa Cruz Biotechnology, CA, USA). For fibronectin and collal blotting, the protein extract was separated in 3-8% tris-acetate gel. Protein bands were recorded on X-ray film by reacting with ECL chemiluminescence reagents (Amersham Biosciences, NJ, USA). Alpha-tubulin (Abeam, UK) was used as the loading control for total protein, and Tata binding protein (TBP) (Abeam, UK) was used as the loading control for nuclear protein.
[00176] Quantification of β-Galactosidase Activity
[00177] Beta-galactosidase activity in extracts from T6/GFAP-fccZ cells treated by DBZIM or TDBZIM was measured using a chemiluminescent assay kit (Roche, Mannheim, Germany) in a 96-well plate format, according to the manufacturer's instructions. Briefly, cells were washed twice with ice-cold Ix PBS (pH 7.4), and lysed for 30 min at room temperature in the lysis buffer. The supernatant was collected for assays with a Tecan Safire π plate reader. Protein concentration was determined with a BCA protein assay Mt (Pierce, EL, USA). Specific β-galactosidase activity was obtained by normalizing against total cellular protein content.
[00178] General Toxicity of IMSs in Mice
[00179] IMSs (DPIM and DBZIM) were dissolved in saline, and administered to Friend virus. B-Type (FVB) mice (weighing ~ 25-30 g) intraperitoneally every other day for 2 weeks. The dosages were 0, 200, 300, 350, 400 and 500 mg/kg for DPIM, and 0, 10, 30, 35, 40 and 55 mg/kg for DBZIM. The injection volume was maintained at 100-160 μl. The mortality rate of mice was assessed after the treatment. The animal experimental protocol was approved by the Institutional Animal Care and Use Committee (IACUC) of Biological Resource Center (BRC) at Biopolis, Singapore.
[00180] Statistical Analysis [00181 ] All quantitative results were presented as mean and standard error of mean (SEM). Experimental data were analyzed using two-tailed Student's t-test assuming unequal variances. A P- value of < 0.05 was considered statistically significant.
[00182] Effect IMSs on liver fibrosis in mice
[00183] For the liver toxin thioacetamide (TAA) model, TAA was given by i.p. injection at 200mg/kg body weight, 3 times per week, for 12 weeks, to induce hepatic fibrosis in mice. For the bile duct ligation model (BDL), adult mice were anesthetized with Ketamine (150 mg/kg)/Xylazine (10 mg/kg) via i.p. injection. An abdominal incision at the midline is made to expose the common bile conduct, which was then ligated using 5-0 silk suture. In the control animal, the bile duct is exposed, but not ligated. DBZIM or DPIM was provided to the mice in drinking water at concentrations from 1 to 1000 mg/liter. The collagen content in the fibrotic liver was assessed by the Sirius Red staining of the liver tissue sections.
[00184] Additional studies were carried out on the effect of DPIM on liver fibrosis inducted by BDL. DPIM was purchased from Sigma Chemicals (St. Louis, MO, USA).
[00185] Mice and compound dosing
[00186] Male FVB/N mice (8-10 weeks old) housed in a specific pathogen-free (SFP) facility on a 12 hour dark-light cycle and with free access to water and diet were used in the study. The animal experiment protocol was approved by the institutional animal care and use committee (IACUC), the Biomedical Research Council (BMRC) of Singapore.
[00187] Cholestatic fibrosis was induced by BDL, briefly, mice were anesthetized with intraperitoneal injection of ketamine (150mg/kg) and xylazine (10mg/kg), after midline laparotomy, the common bile duct was ligated twice with 5-0 silk suture. The sham operation was performed similarly except that bile duct was not double ligated. The experimental mice were divided into four groups: 1) control only (sham operated, but no BDL); 2) control + DPIM; 3) BDL only; 4) BDL + DPIM. Each group consists of 6-8 mice. DPIM compound for treatment was supplied in the drinking water at three concentrations, 500 mg/1, 750 mg/1, lg/1, respectively. The drinking water containing DPIM was changed weekly. It was observed that on average each mouse (normal or operated) drank approximately 3-4ml plain or DPM-containing water every day. Testing by NMR analysis was also conducted to confirm that DPIM is stable in water at room temperature condition for at least two weeks. The compound treatment was started on the third day after animals recovered from the BDL procedure. After 4 weeks , livers were removed from mice for fixation 10% formalin, subsequently embedded in paraffin.
[00188] Measurement of hepatic enzymes
[00189] Serum was collected by retro-orbital method and serum alanine transaminase (ALT) activity was measured by automated procedures with Cobase c 111 (Roche diagnostic).
[00190] Collagen staining by Sirius Red
[00191 ] For consistency, paraffin-sections prepared from the left and median lobes were stained with Sirius-Red to visualize the collagen content. Briefly, after de-wax, re-hydration, air- drying, the sections were stained in 0.1% Sirius Red solution for one hour, washed twice with 0.5% acetic acid, then dehydrated in three changes of ethanol, the sections were finally cleared in xylene and mounted with Histomount (National diagnostics, Georgia, USA). The collagen stained areas were measured in at least 6 independent fields under low magnification (4x) for each left and median lobe, respectively, and quantified with Image J software.
[00192] Statistical analysis
[00193] AU images displayed are representative of 6-8 mice, unless stated otherwise. All quantitative data were expressed as mean ± SE, differences among different treatment groups were analysed by two-tailed unpaired Student t-test, P value less than 0.05 were considered statistically significant.
[00194] Effect of DBZIM on Hepatocellular Carcinoma and Other Tumour Cells
[00195] DBZIM (1,3-bisbenzylimidazolium bromide) was synthesized based on a published method (Harlow et al. 1996).
[00196] Cell culture and compound treatment
[00197] Human heptocellular carcinoma cell lines: BDLE (an undifferentiated cell line) and HepG2 (differentiated cell line) were purchased from JCRB (Japanese Collection of Research Bioresources). Human gastric cancer cell lines AGS and MKN28, lung cancer cell line H1299, breast cancer cell line MCF-7, glioma cell line U87 MG, and C6 were all purchase from ATCC. Cell lines were routinely cultured in DMEM with 10% FBS in a humidified CO2 incubator at 370C, and split at 1:4 ratio by trypsinization (0.05% trypsin/0.53 mM ethylenediaminetetraacetic acid (EDTA) when they grew to confluence. [00198] Stock solution of DBZIM was prepared in water. Cells were initially seeded in cell culture plates or flasks in DMEM supplemented with 10% FBS for 18-24 h before the addition of compound of various concentrations for different time periods. Details of the each treatment can be found in the figure legends.
[00199] DNA content analysis
[00200] HLE cells grown in a 96-well plate were treated with compounds in 6 replicates for 24 hr. Cells were then fixed with 4% paraformaldehyde and stained with DAPI. Cells were analyzed and imaged on ArrayScan VTI HCS reader (Thermal Scientific, PA, USA) and DΝA content (2Ν vs. 4N) was determined based on DAPI staining.
[00201 ] Cell proliferation assay
[00202] HLE cells were grown, treated by compounds in 6 replicates for 48 hr in a 96-well plate. Cells were then fixed by using 4% paraformaldehyde and stained by DAPI and BrdU antibody using HitMt (Thermo Scientific, IL, USA). Cells were imaged and analyzed on ArrayScan VTI HCS reader (Thermal Scientific, PA, USA). Cell numbers were counted based on nuclear staining and BrdU incorporation was determined and expressed as percentage of positively stained cells.
[00203] Proliferation of Glioma cell lines: U87 MG and C6 under the treatment of DBZIM was examined using 3-(4,5-dimethyllhiazol-2-yl)-5-(3-carboxyinethoxyphenyl)-2-(4-sulfoplienyl)- 2H-tetrazolium (MTS) based proliferation assay kit (Promega, WI, USA). MTS reagent was added to the control and treated cells (after 48 hr) directly and the absorbance signal was read at 490 nm using a Tecan S afire II plate reader. Proliferation was similarly performed for gastric cancer cell line AGS, lung cancer cell line H1299, and breast cancer cell line MCF-7, but the IC50 value was determined at 72 h, instead of 48 h.
[00204] Cell based caspase 3/7 and LDH assays
[00205] Capspase 3/7 activity and lactate dehydrogenase (LDH) release were measured by Fluorometric assays (Promega Corp., WI, USA). The HCC cells were cultured in 96-well plate and treated with DBZIM of various concentrations for 6 hr before assays. For caspase 3/7 assay, medium was removed and substrate buffer was added to lyse the cell and react with caspase 3/7 to give rise to fluorescence signal which was read at Ex = 485 nm and Em = 530 nm on a Tecan Safire π plate reader after 1 hr incubation. The caspase 3/7 activity of sample cells with the compound treatrnent was expressed as relative fold change after normalizing to the control cells. The LDH assay (indicative of membrane integrity) was performed using a Kit from Promega Corp according to manufactor's instructions.
[00206] Protein extract based caspase 3/8/9 colorimetric assays-
[00207] Capspase 3/8/9 activities were determined using total protein extract as starting material (Biovision inc., CA, USA). HLE cells were cultured in T75 flask and treated with DBZM of various concentrations. Cytosolic protein extract was collected by pelleting and lysing the cells and centrifugation to remove the debris. Capspase 3, 8 and 9 activities were assayed using the same protein extract but adding the respective substrate. Absorbance readings were taken at 405 nm using Tecan Safire π plate reader. Results were expressed as relative caspase activity by normalizing to control cells without compound treatment.
[00208] Annexin V staining and analysis by cytometry
[00209] HLE and HepG2 cells were treated with various concentrations of DBZIM or TRAIL
(100 ng/ml) for 24 hr, harvested and labeled with Annexin V-FITC (BioVision, CA, USA). The labeled cells were analysed by flow cytometry FACSCalibur (Beckman-Dickson, NJ, USA) and CellQuest software. Cells showing green staining in the plasma membrane were considered as apoptotic cells.
[00210] AIF/Survivin translocation assay
[00211 ] HLE cells cultured in 96-well plate were treated with DBZIM for 24 hr before staining and imaging. Cells were then fixed and stained with antibodies against apoptosis-inducing protein (ADP) and Surviyin (Lab Vision Corp. CA, USA) and buffer reagent from HitMt (Thermo Scientific, IL, USA). The cells were imaged and analyzed on ArrayScan VTI HCS reader (Thermal Scientific, PA, USA). Fluorescence intensity in the cytoplasmic and nuclear area was plotted for indication of cytoplasm to. nucleus translocation.
[00212] SDS-PAGE and Western blot
[00213] Protein extracts were prepared by lysing cell pellet in RJPA buffer (Pierce, IL, USA) for total protein extract, or in NE-PER reagent from Pierce (Pierce, IL, USA) for nuclear and cytoplasmic fractions, or in a Kit from BioVision (CA, USA) for mitochondria and cytosol fractions. Protein content was quantified using BCA Kit (Pierce, IL, USA). Fifteen micrograms of protein were resolved in 4-12% gradient SDS/polyacrylamide gel electrophoresis (PAGE) gel, and transferred to intracellular membrane. The membrane was then probed with primary antibodies against BCI-XL, Bak, Bcl-2α, Bax (Lab Vision Corp., CA, USA), survivin, xIAP, cIAP (R&D systems Inc., MN, USA), c-Jun, c-Fos, JunB, JunD, Fra-1, Fra-2 (Santa Craz Biotechnology, CA, USA) respectively, and detected by horseradish peroxidase-conjugated secondary antibody (Santa Cruz Biotechnology, CA, USA). Protein bands were recorded on X-ray film by reacting with ECL chemiluminescence reagents (Amersham Biosciences, NJ, USA). Alpha-tubulin (Abeam, UK) was used as the loading control for total protein, Tata binding protein (TBP) (Abeam, UK) was used as the loading control for nuclear protein, and Cox4 (Abeam, UK) was used as loading control for mitochondria protein.
[00214] ROS determination
[00215] Oxidative stress was measured using Cellomic HitKit (Thermo Scientific, IL, USA). HLE cells were treated with DBZIM for 24 hr, fixed and labeled by Dihydroethidium probe for quantitation of ROS generation and Hoechst dye for nuclear staining. The result was analyzed by Target Activation BioApplication software. The percentage of positive responding cells was plotted as the indication of ROS amount.
[00216] High-content analysis for cytotoxicity and apoptosis
[00217] HitMt: Multiparameter Cytotoxicity 1 and Multiparameter Apoptosis 1 assay kits
(Thermal Scientific, PA, USA) were used to exam the cytotoxicity and apoptosis profile of additional IMSs. Cells were cultured and treated in 96-well plate for 24 hr before assaying. Cells were fixed and stained essentially following the' manufacturer's instruction. The images were taken by ArrayScan VTI HCS reader (Thermal Scientific, PA, USA) and analyzed using Cell Viability and Cell Health Profiling BioApplication software respectively.
[00218] HCC xenograft model and dosing
[00219] Balb/c Nude mice, 5τ6 weeks old, were inoculated with IxIO7 Huh7 cells in 0.2 ml volume of Matrigel/DMEM mix. Since not every mouse developed a tumor after the inoculation of HCC cells, only those with visible tumors (about 8 weeks after inoculation) were used for subsequent experiment. The tumor-bearing mice were randomly divided to the control and the treatment group- (n=3), respectively. Mice in the treatment group had free access to drinking water containing DBZM (2 grams/liter) only, while mice in the control group had free access to water only. The compound treatment lasted for 3 weeks. The tumor size was measured weekly, and the tumor volume was calculated using the formula: 0.52 x width2 x length.
[00220] Anticancer activities of additional IMSs [00221] Synthesis and characterization of IMSs
[00222] All solvents and chemicals were used as obtained from commercial suppliers, unless otherwise indicated. Centrifugation was performed on Eppendorf Centrifuge 5810R (4000 rpm, 10 min). 1H and 13C NMR spectra were recorded on Bruker AV-400 (400 MHz) instrument. Data for 1H and 13C NMR were reported in chemical shift (δ ppm) and multiplicity (s = singlet, d = doublet, t = triplet, q = quartet, m = multiplet). GC-MS was performed on Shimadzu GCMS QP2010. Gas liquid chromatography (GLC) was performed on Agilent 6890N gas chromatographs equipped with split-mode capillary injection system and flame ionization detector.
[00223] To prepare substituted imidazoles, NaH (60% in oil, 420 mg, 10.5 mmol) was added to a DMF solution of imidazole (A1) (680 mg, 10 mmol) at 0 0C, and the resulting suspension was stirred at room temperature for 2 h. Benzylbromide (1.71 g, 10 mmol) was added to the residue. The resulting solution was stirred at room temperature for another 4 h. The solvent was removed under vacuum. The product was extracted with dichloromethane (DCM), and B1 was obtained in quantitative yield after removing the solvent. B2 to B4 were synthesized in the same protocol with A2 to A4 as imidazole starting materials. The products were confirmed by MS and NMR.
Figure imgf000043_0001
[00224] DBZM , DBZBM, l,3,-Dibenzyl-2-memylirώlazolium bromide (DBZMM,
Compound C), IBN- 2, 3, 4, 6, 8, 9 (Compound), 12, 13,15, 17, 18, 19, 20, 21, 24, 25, 26, 27, 28, 30, 31, 32 33, 34 were synthesized by mixing 1 mmol of substituted imidazole (like B1- B4) and 1 mmol of substituted benzyl bromide(chloride) or alkyl bromide(chloride) in 1 ml ofDMF. The mixture was stirred at 1000C for 16 hours. The reaction mixture was cooled down to room temperature and ether (10 ml) was added to precipitate product. The powder or gel precipitate was collected as imidazolium product and purified by re-crystalline or chromatograph method and characterized by NMR.
[00225] D3N-22, 23, 29 were made based on literature methods (Zhang et al. 2007; Chianese and Cratree 2005, Boydston et al. 2005).
[00226] The remaining IMSs listed in Table 2 were purchased from Sigma.
[00227] Typically 100 mM of stock solution of IMSs was prepared in DMSO. Aliquots of stock solution were stored until use at -20 C.
[00228] Cell Lines and cell culture
[00229] Human Hepatocellular cancer cell lines HLE (p53 Mutant) and human gastric cancer cell line MKN 28 were obtained from Japan Health Sciences Foundation (Osaka, Japan). The human breast cancer cell line MDAMB231 (mutant p53) and breast epithelial cells MCF-IOA (wild type p53) were obtained from ATCC. These cell lines were maintained in DMEM medium (RPMI 1640 for MDAMB231) supplemented with 10% fetal bovine serum and 1% of mixture Penicillin and Stretomycin incubated at 37 0C in an atmosphere of 5% CO2.
[00230] Growth inhibition assay
[00231 ] The percentage of growth inhibition was determined by using a MTT assay to measure viable cells. For the MTT assay, 1 x 103 cells per well were cultured in 96-well plates and treated with 0 to 100 μM of various types of IMSs for 72 h. After incubation for specified times at 370C in a humidified incubator, culture medium replaced with 10 μL of MTT reagent (5 mg/mL) contains 100 μL medium was added to each well and further incubated for 4 h. The reaction was stopped by adding 100 μL of DMSO. Absorbance was measured at 570 nm on a micro plate reader (SpectroMax 190, Molecular Devices). Data were presented from three separate experiments, and the percentage of IMSs induced cell growth inhibition was determined where DMSO-treated cells (control) were taken as 100%.
[00232] Cell cycle analysis
[00233] To perform cell cycle distribution analysis, 4 x 105 cells were plated in 10 cm dish. The cells (70% confluence) were serum-starved for 24 h to synchronize them in the Go phase of the cell cycle. After 24 h incubation, the cells were replaced with fresh 10% DMEM medium and then treated 60 μM of IBN-15, 90 μM of IBN-19, 90 μM of IBN-24, 30 μM of IBN-24 and 20 μM of IBN-32 ) and 0.045 % DMSO as control for 72 h. The floating and trypsinized adherent cells were collected and washed with PBS. The cell pellets were resuspended in 1 ml of PBS and fixed by the addition of 70% ice-cold ethanol and stored at -2O0C. After incubation for 24hr, the cells were re- pelleted by centrifugation, the cells were washed once with PBS and resuspended in 0.1 mL of PBS containing 100 μg/ml of RNase and then incubated at 37°C for 15 min. Finally, the cells were stained with 0.5 ml of PI solution (100 μg/mL in PBS) for 30 min. Cell cycle distribution was detected by a BD PACS array flow cytometer (Becton Dickinson). Cells with sub-G0 DNA were classified as apoptotic cells.
[00234] Apoptosis assay
[00235] To determine apoptosis analysis, 4 x 105 cells were plated in 10 cm dish. The cells were then treated with 60 μM of IBN-15, 90 μM of IBN-19, 90 μM of IBN-24, 30 μM of EBN-25 and 20 μM of IBN-32 ) and 0.045 % DMSO as control for 72 h. The floating and trypsinized adherent cells were collected and the apoptosis was quantified using Annexin V-FITC reagent (BD Biosciences Pharmingen) and PI (Invitrogen) following the manufacturer's protocol. Briefly, the cells were washed twice with cold PBS and then resuspend cell in 100 μlof x binding buffer containing a 10 μl of Annexin V-FITC (BD-Pharminagen) and 2 μl of PI from the stock of lOOμg/ml. The cells were gently vortexed and incubated for 15 min at room temperature in the dark. Subsequently 400 μl of Ix binding buffer were added to each tube, and the samples were immediately analyzed by FACScan flow cytometer (Becton Dickinson). The fluorescent signals of the Annexin-V conjugate and PI were detected at channels of fluorescence intensity FLl and FL2 (BD LSR π with software BD).
[00236] SDS-PAGE and Western blotting
[00237] The HLE cells were seeded at a density of 4 X 105 in 10 cm dish. After 24 h incubation, the cells were replaced with fresh 10% DMEM medium and then treated with 60 μM of IBN-15, 90 μM of IBN-19, 90 μM of IBN-24, 30 μM of IBN-25 and 20 μM of IBN 32 and 0.045 % DMSO as control for 72h. Whole cell lysates were prepared on day 3 with lysis buffer [125 mM Tris (pH 7.4), 2% Sodium dodecyl sulfate, 10% glycerol, 6M urea and a protease inhibitor cocktail (Sigma) and 0.02% Bromophenol blue]. The lysates were sonicated for 30 seconds and then heated for 5 mins at boiling temperature. The lysates were centrifuged at 13000 rpm for 10 mins and the protein concentration of each sample was estimated using the BCA protein assay. The lysates was then mixed with 5% 2-mercaptoethanol and stored at -80C. For Western blotting, samples containing 50 μg of total cell lysate were loaded onto a SDS-PAGE and subjected to electrophoresis. Proteins were transferred to a nitrocellulose membrane and then blocked with 5% non-fat dry milk in Tris buffer saline containing 0.1% Tween 20 for 60 min at 37 C. The membranes were probed with a primary antibody overnight at 4 C in TBST containing 5% Bovine serum albumin. Then the membrane was incubated with horseradish peroxidase (HRP)-coηjugated anti-rabbit or anti-mouse secondary antibodies (GE Health care, Chalfont St Giles, UK). Detection was performed with enhanced chemiluminescence (ECL) reagent (Amersham Arlington Heights, IL) according to the manufacturer's protocol. Rabbit polyclonal Phospho-p53 (Serl5, 20, 46, 392), anti-caspase-9, anti-caspase-3, anti-PARP antibodies were purchased from Cell Signaling Technology, Inc. (Beverly, MA, USA). Antibodies against p53 and p21 were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Equal loading of protein was demonstrated by probing the membranes with a mouse anti-β-actin monoclonal antibody (Sigma Chemical Co. (St. Louis, MO).
[00238] Immunostaining and confocal microscopy
[00239] For immunostaining, HLE cells were seeded at a density of 1 X 104 in 8 well chamber slide. After 24 h incubation, the cells were replaced with 200 μl fresh 10% DMEM medium and then treated with 60 μM of IBN-15, 90 μM of IBN-19, 90 μM of IBN-24, 30 μM of IBN-25 and 20 μM of IBN-32 and 0.045 % DMSO as control for 24 and 48 h. After washing, cells were fixed in 4% formaldehyde for 15 mins and permeabilized with 0.5% Triton X -100 in PBS for 10 mins. The cells were then blocked for 60 min in PBS containing 3% bovine serum albumin and then incubated for 1 h with a mouse antibody against nucleolin (MS-3, Santa Cruz; 1:100 in blocking buffer). After washing extensively with PBS, the cells were then incubated for 1 h at room temperature in dark with a FITC-conjugated antibody against mouse IgG (1 :20 ilution). The slides were mounted with a VectaShield medium containing DAPI (Vector Laboratories, Burlingame, CA). Microscopy was performed with a Zeiss LSM 510 Meta (upright stand) confocal microscope.
[00240] Real-time quantitative RT-PCR analysis
[00241 ] Cells were seeded at a density of 4xlO5 cells per dish in 10 cm dishes. Next day, the cells were exposed to 9OuM of IBN-19, 9OuM of IBN-24, 4OuM of IBN-25 and 0.045% of PMSO (Dimethyl Sulphoxide) as the control. After 48 hrs, total RNAs were extracted from the cells using Nucleospin RNA II kit as per the manufacturer's istructions. RNA quality and yield were evaluated after spectrophotometer measurements. [00242] The human apoptosis PCR array (RT2 Profiler) was used to analyze mRNA levels of 84 key genes involved in apoptosis, in a 96- well format, according to the manufacturer's instructions (Super Array Bioscience). First-strand cDNA was synthesized with lμg of RNA by using a PCR array first strand-synthesis kit (C-03; Super array Bioscience). This kit uses reverse transcriptase (Power Script; Super array Bioscience) and a combination of random primers and oligo dT primers. The total volume of the reaction was 20 μL diluted to 100 μL. PCR reactions were performed using real-time PCR (79s00HT 96- well block with RT2 Real-Time SYBR Green PCR master mix PA-012; Applied Biosystems Instruments 7500). The total volume of the PCR reaction was 25 μl. An equivalent of lμg of RNA was applied to the PCR reaction. The thermocycler parameters were 95°C for 10 minutes, followed by 40 cycles of 950C for 15 seconds and 6O0C for 1 minute. Relative changes in gene expression were calculated using the ΔΔCt (cycle threshold) method. An average of the number of cycles of the five housekeeping genes, GAPDH, Actin-β, β2m, Hprtl, and Rpll3d, was used to normalize the expression between samples. The expression data are presented as fold regulation. GAPDH gene expression levels were utilized as an internal control to normalize the data. Reported fold changes in expression are the ratios of treatment over control values.
[00243] Treatment of HCC mice with compound 9
[00244] Balb/c Nude mice, 5-6 weeks old, were inoculated with IxIO7 Huh7 cells in 0.2 ml volume of Matrigel/DMEM mix. Since not every mouse developed a tumor after the inoculation of HCC cells, only those with visible tumors (about 8 weeks after inoculation) were used for subsequent experiment. The tumor-bearing mice were randomly divided to the control and the treatment group (n=3), respectively. Mice in the treatment group had free access to drinking water containing compound 9 (IBN-9) at 0.6g/l or 1.5g/l only, while mice in the control group had free access to water only. The compound treatment lasted for 3 weeks. The tumor size was measured weekly, and the tumor volume was calculated using the formula: 0.52 x width2 x length.
[00245] Statistical analysis
[00246] All data are presented as mean ± SD. Significant differences between the groups were determined using the unpaired Student's t-test. Levels of significance were defined as follows: p < 0.05 (*), p < 0.01 (**) and p < 0.001 (***).
[00247] Results
[00248] Anti-oxidative, anti-inflammatory and anti-fibrotic properties of IMSs [00249] Determination of Toxicity of IMSs on Cultured HSC T6 Cells and FVB Mice
[00250] Firstly we assessed the cytotoxicity of DBZM and TDBZM on cultured HSC-T6 cells using a 3-(435-dimethyMπazol-2-yl)-5-(3-carboxymethoxyp]ienyl)-2-(4-sulfoplienyl)-2H- tetrazolium (MTS) based proliferation assay kit (Promega, WI, USA). Figs. Ia and Ib show the effect of DBZM (0-400 μM) and TDBZIM (0-200 μM) on the proliferation of T6 cells cultured in DMEM containing 10% of FBS. At high concentrations of 250-400 μM, DBZM inhibited cell proliferation by 3.5-8.9% with statistical significance. At high concentrations of 125-200 μM, TDBZM demonstrated a stronger inhibition of 13-19% as compared to the control.
[00251 ] In the preliminary toxicity studies, two forms of IMSs were selected for in vivo testing with FVB inbred mice. For DBZM, the lethal dosage was 50 mg/kg and LD50 was estimated to be 30^0 mg/kg. For DPM, the lethal dosage was 500 mg/kg and LD50 was ~ 300- 400 mg/kg.
[00252] DBZIM and TDBZIM Attenuated Cellular ROS Level and Enhanced GSH/GSSG Ratio
[00253] To measure the cellular oxidative stress level of HSC T6 cells treated with MSs, cells cultured with DBZM (10, 50, 100 and 300 μM) or TDBZIM (10, 50 and 100 μM) for 48 h in full serum medium were assayed for ROS, GSH and GSSG. Cells treated with NAC (1 or 5 mM), and EGCG (25 μM) were included as references. MSs treatment led to significantly attenuated cellular ROS level. As shown in Fig. 2a, ROS was suppressed by DBZM in a dosage-dependent manner (by 25% at 50 μM (P < 0.005) and by 34% at 300 μM (P < 0.0005)). TDBZM, which is comprised of a trimer of DBZM, substantially suppressed cellular ROS level by 19% at 10 μM (P < 0.01) and by 36% at 100 μM (P < 0.00001) (Fig. 2b). In comparison, EGCG was able to attenuate ROS by 14% at 25 μM (P < 0.005), whilst NAC did not show apparent inhibition on ROS level even at 1 mM.
[00254] The effect of the compounds on the level of an important endogenous antioxidant, GSH, was also investigated. As indicated in Figures 3a and 3b, the total cellular GSH amount was quantified including the reduced glutathione (GSH) and oxidized glutathione (GSSG). As shown in Fig. 3a, DBZM attenuated the total GSH level by 7%, 12% (P < 0.005) and 20% (P < 0.0001) at 10, 50 and 100 μM, respectively. At a high dosage of 300 μM of DBZM, slight enhancement by 8% was observed, but without statistical significance. NAC (5 mM) did not show any effect on the total GSH, while EGCG (25 μM) induced the total GSH synthesis by 19% (P < 0.01), which was in agreement with an earlier report (Fu et al. 2006). In contrast to its monomer, TDBZIM did not alter the total cellular GSH level at 10-100 μM (Fig. 3b). GSH was oxidized to GSSG in the reaction to reduce H2O2 to H2O. The amount of GSSG was also quantified as shown in Figs. 3c and 3d. DBZIM depleted GSSG by 13% at 10 μM, 23% at 50 μM (P < 0.0005), 35% at 100 μM (P < 0.0001) and 85% at 300 μM (P < 0.0001). In contrast, EGCG reduced GSSG by 42% at 25 μM (P < 0.05) and NAC had no effect on GSSG at 5 mM. TDBZIM also suppressed GSSG production by 12% at 10 μM (P < 0.05), by 35% at 50 μM (P < 0.05), and by 50% at 100 μM (P < 0.0001). The GSH/GSSG ratio was enhanced by 8%, 15% (P < 0.001) and 23% (P < 0.0001) under the influence of DBZIM at 10, 50 and 100 μM, respectively (see Fig. 3e). At a high dosage of DBZIM (300 μM), the increase in GSH/GSSG ratio was more dramatic, reaching 7.4 fold of that of the control group. There was no change in the GSH/GSSG ratio for NAC at 5 mM, whereas a 2-fold increase in the GSH/GSSG ratio was observed for EGCG at 25 μM (P < 0.0001). For TDBZIM, the GSH/GSSG ratio was enhanced by 14% at 10 μM, by 65% at 50 μM (P < 0.05) and by 106% at 100 μM (P < 0.001) (Fig. 3f). These results suggested that these two forms of IMSs significantly enhanced the GSH/GSSG ratio mainly through attenuating the production of GSSG from the oxidation of GSH, resulting in improved,anti-oxidation.
[00255] DBZIM and TDBZIM Attenuated GPx and CAT Activities, and DBZIM enhanced GST Activity.
[00256] To further investigate the anti-oxidant properties of the IMSs, the activities of antioxidant enzymes: GPx, CAT, SOD. and GST were assayed. Figure 4a shows that DBZIM attenuated the GPx activity by 10% at 10 μM (P < 0.005) and by 10% at 50 μM (P < 0.01), but enhanced the GPx activity by 15% at a high concentration of 300 μM (P < 0.005), with a slight biphasic pattern. For TDBZIM, GPx activity was suppressed by 18% at 10 μM (P < 0.05), by 25% at 50 μM (P < 0.01) and by 34% at 100 μM (P < 0.005) (Fig. 4b). In comparison, EGCG attenuated the GPx activity by 53% (P < 0.005) at'25 μM, while NAC did not show much effect at 5 mM (Fig. 4a).
[00257] A similar pattern was observed for DBZlM' s effect on CAT activity. DBZIM was able to attenuate CAT activity by 19%, 16% and 12%, respectively at 10 μM, 50 μM and 100 μM, respectively (P<0.05) (Fig. 4c). When the concentration was increased to 300 μM, the attenuating effect decreased to 8% without statistical significance. TDBZTM inhibited the CAT activity by 25% at 10 μM (P < 0.005), 31% at 50 μM (P < 0.0005) and 20% at 100 μM (P < 0.005), with a less potent effect above 50 μM (Fig. 4d). EGCG showed 13% suppression on CAT activity, but the data were not significant, whereas NAC demonstrated 35% inhibition (P < 0.01) (Fig. 4c). [00258] As shown in Figs. 4e and 4f, DBZIM, TDBZM, EGCG and NAC all did not show much effect on the SOD activity, suggesting that these compounds did not act on superoxides. Figure 4g illustrates the GST activity of cells under the treatment of DBZIM (10-300 μM). GST activity was enhanced by 22% at 100 μM (P < 0.005) and by 32% at 300 μM (P < 0.0001). Ih contrast, EGCG and NAC did not have much affect on the GST activity.
[00259] DBZIM and TDBZIM Protected Primary HSCs from DMSO Cytotoxicity DMSO is a common solvent used in cell-based assays (at concentrations of < 0.5% (Wv)) and cell cryopreservation (at a concentration of ~ 10% (v/v). For the primary HSCs, however, DMSO imposed significant toxicity at a concentration of 0.2% (v/v) (Fig. 5a). Control cells incubated with 0.2% (v/v) of DMSO died after 26 h. In contrast, the cells survived in the co-presence of 100 μM of DBZIM or 50 μM of TDBZIM and 0.2% (v/v) of DMSO. When they were cultivated for over 93 h, the cells remained viable and proliferated to confluency in the presence of TDBZIM. However, most cells died in the presence of DBZM after 93 h. This suggested that TDBZM provided a more potent protective effect against DMSO's toxicity on primary HSCs.
[00260] DMSO-induced oxidative stress in cultured HSC T6 cells was quantitatively characterized by the depleted total cellular GSH level (74% of the control, P < 0.0001), the dramatically increased GSSG amount (4.4 fold of the control, P < 0.0001), the decreased GSH/GSSG ratio (17% of the control, P < 0.001), the induced GPx (2.62 times of the control, P < 0.0001) and CAT (1.7 fold of the control, P < 0.0001) anti-oxidant enzymes (see Figs. 5b,c,d,e,"f). DMSO also enhanced SOD activity slightly (10% increase), but the data were not statistically significant (Fig. 5g). This finding suggested that the protective effect of MSs against DMSO cytotoxicty on primary HSCs was through the attenuation of oxidative stress. A similar observation was reported in another study that found that quercetin, a flavonoid (0.1 μM), can effectively protect human lens epithelial cells from dying from 1% (v/v) of DMSO, presumably through the attenuation of oxidative stress (Cao et al. 2007).
[00261 ] IMSs Suppressed HSC Activation Markers and Fibrogenic Endpoints in a Time- and
Dosage-Dependent Manner
[00262] It has earlier been shown that GFAP can be used as a biomarker along with SMAA for HSC activation in vitro {Zhang et. al 2006). Deposition of ECM (including collagen and fibronectin) was the endpoint used to assess the degree of fibrosis. Two MSs (DBZM and TDBZM) suppressed the mRNA level of both HSC activation markers (GFAP and SMAA) and fibrotic endpoint (collal and fibronectin) in a dόsage-dependent (Figs. 6a and 6b) and time- dependent (Figs. 7a-d) manner. As shown in Fig. 6a, DBZIM attenuated GFAP mRNA expression levels by 18% (P < 0.05) and 86% (P < 0.001) at 10 μM and 300 μM, respectively. It attenuated SMAA by 21% at 100 μM (P < 0.05) and by 58% at 300 μM (P < 0.001), collal by 21% at 100 μM (P < 0.05) and by 44% at 300 μM (P < 0.001), and fibronectin by 19% (P < 0.005) to 40% (P < 0.0001) with dosages of 1 μM to 300 μM.
[00263] The dosage dependency of TDBZIM was less prominent compared to its monomer counterpart (Fig. 6b). TDBZM decreased GFAP mRNA by 32% at 1 μM (P < 0.01), by 20% at 10 μM (P < 0.01) and by 60% at 100 μM (P < 0.0001), SMAA by 27% only at a low dosage of 1 μM (P < 0.01), collal by 39% at 1 μM (P < 0.005), by 36% at 10 μM (P < 0.005) and by 53% at 100 μM (P < 0.001), and fibronectin by 32% at 1 μM (P < 0.0010), by 28% at 10 μM (P < 0.0005) and by 43% at 100 μM (P < 0.0001).
[00264] Time course study showed that 100 μM of DBZIM and TDBZIM, respectively, suppressed GFAP by 28% (P < 0.05) and 30% (P < 0.05) at 8 h, by 40% (P < 0.005) and 36% (P < 0.005) at 24 h, and by 56% (P < 0.0001) and 60% (P < 0.0001) at 48 h. They suppressed collal by 22% and 22% (P < 0.01) at 8 h, by 29% (P < 0.005) and 40% (P < 0.0001) at 24 h, and by 21% (P < 0.05) and 53% (P < 0.001) at 48 h. They also suppressed fibronectin by 15% (P < 0.05) and 25% (P
< 0.05) at 8 h, by 17% (P < 0.05) and 29% (P < 0.05) at 24 h, and by 25% (P < 0.0005) and 43% (P
< 0.0001) at 48 h. It was generally true that the IMSs suppressed almost all HSC activation markers and endpoint molecules in a time- and dosage-dependent manner, except that SMAA was transiently induced by up to 79% by 100 μM of TDBZIM at 24 h, while no change was observed with both compounds at 8 h, and 21% suppression was noted for DBZIM (P < 0.05) at 48 h. SMAA expression was particularly induced by TDBZIM at a high dosage. Western blotting of GFAP, SMAA, collal and fibronectin was also performed to evaluate the influence of compounds on the protein expression level. As shown in Fig. 8, both DBZIM (100 μM) and TDBZM (50 μM) attenuated procollagen αl(l) and fibronectin significantly, and GFAP expression to a less degree, when the cells were treated for 48 h. DBZM suppressed SMAA protein, but TDBZM enhanced SMAA, which agreed with the real-time PCR data. Other time points (8 h and 24 h) were also examined, but no significant change was observed (data were not shown).
[00265] IMSs Suppressed Transcription ofTGF-βl and TGFβRI
[00266] TGF-β 1 is the most important pro-fibrogenic cytokine. When the liver is insulted, quiescent HSCs respond to TGF-β 1 secreted by Kupffer cells and endothelial cells, and start to produce TGF-βl themselves via autocrine loops. TGF-βl is over-expressed during HSC activation and liver fibrogenesis (Bachem et al. 1992). The total TGF-βl synthesis under the treatment of MSs was measured. As shown in Fig. 9a, DBZIM suppressed the TGF-βl mRNA by 28% (P < 0.05) and 33% (P < 0.005) at high concentrations of 100 μM and 300 μM, respectively. TDBZM inhibited TGF-βl by ~ 23% regardless of dosage (1-100 μM) with statistical significance (P < 0.05) (Fig. 9b). For 8-24 h treatment, both DBZIM and TDBZM suppressed TGF-βl transcription by 40-50% (P < 0.05) (Fig. 9c). The less apparent dosage- or time-dependency might be due to the auto-looping characteristic of TGF-βl.
[00267] DBZIM and TDBZIM Inhibited Pro-Inflammatory and Pro-Fϊbrogenic Cytokine IL-6
[00268] IL-6 is regulated by NF-κB and involved in inflammatory responses. Being a pro- contractile and pro-inflammatory cytokine, IL-6 plays an important role in liver fibrogenesis. To study the influence of MSs on IL-6, IL-6 mRNA expression was quantified using real-time PCR. As shown in Figs. 10a, DBZIM inhibited IL-6 transcription in a dosage-dependent manner by 37% at 1 μM (P < 0.0005), by 46% at 10 μM (P < 0.0005), by 53% at 100 μM (P < 0.0001), and by 59% at 300 μM (P < 0.0001). TDBZM demonstrated more potent inhibition of 37% at 1 μM (P < 0.01), 55% at 10 μM (P < 0.0001) and 80% at 100 μM (P < 0.0001) (Fig. 10b). Time course study showed that 100 μM of DBZM significantly suppressed IL-6 mRNA expression by 53% at 48 h (P < 0.005). Shorter treatment of DBZIM did not show statistically significant suppression on IL-6. 100 μM of TDBZM demonstrated an inhibitory effect of 52% at 24 h (P < 0.005), and 80% at 48 h (P < 0.0001) (Fig. 10c).
[00269] DBZIM and TDBZIM Mediated through NF- KB and AP-I Transcription Factors
[00270] Transcription factors NF-κB and AP-I are known to be sensitive to cellular redox states. NF-KB also plays an essential role in physiological conditions such as inflammation and tissue repair. Protein expression of the NF-κB P65 and activation protein 1 (AP-I) family members (c-Fos, c-Jun, Jun B/D, Era- 1/2) were examined when the cells were treated with MSs. As shown in Fig. 11, nuclear NF-κB P65 in an active form was significantly attenuated by both DBZM and TDBZM. The amount of latent NF-κB in cytosol was not affected (data not shown). AP-I, c-Fos, Jun D and Fra-1 were suppressed, while c-Jun, Jun B and Fra-2 were less affected.
[00271 ] IMSs Regulated the Expression of a HSC-Specific GFAP-Reporter Surrogate
It has been shown earlier that the GFAP-lacZ transgene reporter could be used to screen anti- fibrotic compounds (Maubach et. al 2006, Zhang et. al 2006). In the current experiment, the β- galactosidase activity under the treatment of DBZIM and TDBZM at different dosages (0-300 μM) and time points (24 h and 48 h) was assayed. Significant suppression on β-galactosidase activity was observed for DBZM: by 25% at 100 μM (P < 0.001) and by 34% at 300 μM (P < 0.001) (Fig. 12a). For TDBZIM, a smaller and transient suppression (20%) was observed at 100 μM at 24 h (Fig. 12b).
[00272] Safety and Efficacy of Additional IMSs with Different Substituents
[00273] IMSs of different structures were also tested in a preliminary structure-activity- relationship (SAR) study. Testing was performed on 3-Bisbenzylimidazolium bromide (DBZIM), l,3,5-tris(4-methyl-imidazolium)-linlced cyclophane-3Br (TDBZIM), 1,3-Dϋsopropylimidazolium tetrafluoroborate (DPIM), 1,3-Di-tert-butylirnidazolmumtetrafluoroborate (DBIM), 1,3-Bis(l- adamantyl)imidazolium tetrafluoroborate (AMTM), l,3-Bis(2,4,6-1rimemylphenyl)-imidazo]inium chloride (TMPHIM), l,3-Bis(2,6-diisopropyl-phenyl)-imidazolinium chloride (DPPHIM) and 1,3- Bisbenzyl-benzimida-zolium bromide (DBZBIM). In general, those with smaller aliphatic groups, such as 1,3-dϋsopropylimidazolinium tetrafluoroborate (DPIM) and 1,3-di-feτt-butylimidazolium tetrafluoroborate (DBIM), showed less toxicity on T6 cells, compared to those with bulky aromatic groups, such as l,3-bis(2,4,6-trimethylphenyl)imidazolium chloride (TMPHTM) and l,3-bis(2,6- dϋsopropylphenyl)imidazolium chloride (DPPHIM). Those with a methyl spacer between the imidazolium and aromatic ring, such as 1 ,3-bisbenzylimi dazolium bromide (DBZIM) and 1,3- bisbenzylbenzimidazolium bromide (DBZBIM), showed moderate toxicity. The toxicity of compounds was ranked based on IC50 as follows: DPIM (IC50 = 3.1 mM) < DBIM (IC50 was undetermined due to limited solubility in DMSO, but estimated to be 2-3 mM) < DBZTM (IC50 = 1.7 mM) < TDBZIM (IC50 ~ 500 μM) < DBZBIM (IC50 = 310 μM) < AMIM (TC50 = 166 μM) < TMPHIM (IC50 = 110 μM) < DPPHM (IC50 = 34 μM).
[00274] Efficacy of compounds in suppressing fibrosis was also evaluated. HSC T6 cells were treated with IMSs at concentrations that were below their respective IC50. Compounds were either dissolved in DMSO or H2O depending on their solubility, and the intensities of the bands were compared to the respective control. As shown in Fig. 13A, proteins of SMAA, Collal, fibronectin, TGF-βl and TGFβ RI were all suppressed with the treatment of MSs. Semiquantitative data from measuring the gray scale of the band pixels is summarized in Fig. 13B . If all 5 parameters are assigned with equal importance, the efficacy of MSs could be roughly ranked as DPM > TMPHM > AMM > DBM > DBZBM > DBZM > DPPfflM > TDBZM. [00275] GFAP, SMAA, fibronectin and collal transcripts were also quantified by real-time PCR. As shown in Figs. 14a,b,c,d, GFAP mRNA was suppressed by 2 mM of DPIM by 78% (P < 0.0001), by 50 μM of AMIM by 69% (P < 0.0005), by 50 μM of TMPHIM by 66% (P < 0.001), and by 100 μM of DBZBIM by 50% (P < 0.005). SMAA mRNA was suppressed by 1 mM of DBIM by 49% (P < 0.05), by 2 mM of DPIM by 78% (P < 0.0001), by 50 μM of AMIM by 23% (P < 0.05), and by 50 μM of TMPHIM by 49% (P < 0.05). Fibronectin mRNA was suppressed by DPM by 48% (P < 0.05), by TMPHIM by 29% (P < 0.005), and by DBZIM by 26% (P < 0.005). Collal mRNA was suppressed by DBM by 58% (P < 0.0005), by DPM by 61% (P < 0.0001), by TMPHM by 62% (P < 0.0001), and by DPPHM by 47% (P < 0.05). EGCG did not show significant inhibition on HSC activation and fibrosis. This might due to the chemical instability of EGCG under the prolonged incubation of 48 h. It was reported that the half life of EGCG was only 30 min in the culture medium for human esophageal cancer cells (Hou et al. 2005) and ~ 72 min in rat blood at a dosage of 30 mg/kg (Lin et al. 2004). Among all the MSs, DPM and TMPHM showed significant suppression on all four genes, and DPIM was the most potent agent under the test conditions.
[00276] Effect IMSs on liver fibrosis in mice
[00277] DBZIM and DPM were tested in two liver fibrosis models in mice induced by liver toxin thioacetamide (TAA) or by a surgical procedure called bile duct ligation (BDL), respectively. As shown in Figure 15, the two MSs, when administered in drinking water, showed anti-fibrotic properties in the two mouse models of liver fibrosis. Particularly DBZM attenuated fibrosis in the TAA model (at 500mg/L, 12 weeks) (Figure 15a) and the BDL model (at 10mg/L, 4weeks) (Figure 15b), whereas the DPM displayed its effect in the BDL model at lg/L (4 wks) (Figure 15c). This data suggests that MSs have potential as anti-liver fibrosis therapeutic compounds.
[00278] DPIM attenuated liver fibrosis
[00279] DPM was chosen for further in vivo study, To ensure proficiency in BDL procedure, a number of exercises and tests were performed to eventually achieve over 90% success rate on the BDL surgery and over 4 weeks of survival time for the operated mice. The extent of fibrosis was assessed by the Sirius-Red collagen staining at 4 weeks post-BDL. An extensive collagen accumulation around the sinusoidal areas was observed in the BDL group (Fig. 16C), whereas only minimal collagen staining was seen in the sham operated group (Fig. 16A). Treatment of sham mice with lg/1 of DPM for 4 weeks did not induce any significant build-up of collagen (Fig. 16B). Four weeks after compound treatment, the collagen contents were dramatically reduced by 750mg/l (Fig.16E) and lg/1 (Fig.16F) of DPM. The effect by the 500mg/l of DPlM was not conclusive, since only one mouse was treated with this dose at the time of data collection (Fig. 16D). Quantitatively the Sirius-Red positive areas were reduced from 8.69 to 4.33 by the dose of 750mg/l, and even more (and dose-dependently) down to 3.42 by the dose of lg/1 (Fig.17).
[00280] DPIM reduced fibrotic cellular proliferation
[00281 ] Liver weight increases in BDL-mice mainly due to 1) the build-up of bile, and 2) fibrotic cellular proliferation. The livers of different treated groups were weighed, 4 weeks after BDL, liver weight was increased by 83% compared with sham operated mice (1.50 ± 0.19g for sham operated, vs. 2.74 + 0.5 Ig for BDL 4 weeks, P<0.05). However, DPIM compound treatments showed a significant liver weight reduction (P < 0.05, DPIM compound treatment vs. BDL 4 weeks): 1.97 ± 0.44g for the 750mg/l treatment group, 1.65 ± 0.2Og for the lg/1 treatment group (Fig. 18). This dose-dependant effect was consistent with Sirius red staining areas. Compared with sham operated mice, DPIM treatments in sham operated mice for 4 weeks did not result in significant liver weight change (1.56 ± 0.08g for DPIM treated vs.1.50 ± 0.19g for sham operated, P = 0.46)(E-g. 18).
[00282] H&E staining of the liver sections (Fig. 19) showed that DPIM treatment (Fig. 19C) reduced the number of proliferative cells (identified by the nuclear staining of Hamehexlin) around the sinusoidal area, when compared to the DBL group (Fig. 19B) and control sham group (Fig 19A). At the messenger level, DPM also reduced the collagen IaI mRNA (Fig. 20).
[00283] Effect of DBZEM on Hepatocellular Carcinoma and Other Tumour Cells
[00284] DBZIM inhibited cell proliferation and disrupted cell cycle in HLE cells
[00285] The effect of DBZM on the HCC cell proliferation and cell cycle was characterized on a high-content screening (HCS) platform. Under each concentration, at lease 1000 cells were analyzed. 5-bromo-2'-deoxyuridine (BrdU), which incorporates into replicating DNA, is used to identify cells which have progressed through S phase of cell cycle. DAPI staining was used for nuclear identification and DNA content determination. DBZM inhibited the HLE cell proliferation as indicated by less total cell number and less BrdU positive cells in a dose-dependant manner (Fig. 21A), and arrested HLE cells in the G0/G1 phase (Fig. 21B).
[00286] DBZJM induced cell death through apoptosis in HCC cells
[00287] - Cell-based caspase 3/7 activity and lactate dehydrogenase (LDH) release assay allow the differentiation of apoptosis and necrosis of cells under the influence of DBZEM. As shown in Fig. 22A-D, caspase 3/7 activity (indicative of apoptosis) in both HLE and HepG2 cells was enhanced by DBZIM, while LDH release (indicative of cytotoxicity) was least affected under the same treatment regimen. It suggested that DBZM used at the current treatment condition caused the HCC cell death mainly by apoptosis, rather than necrosis.
[00288] Apoptosis was further confirmed by annexin V staining. In the initial phase of apoptosis, cells translocated the membrane phosphatidylserine (PS) from the inner face of the plasma membrane to the cell surface. The surface PS is readily detected by fluorescent conjugate of annexin V, which has a high affinity for PS and often used as an apoptosis marker. As shown in the scatter plots in Fig. 22E-F, DBZIM dose-dependently induced apoptosis in both HepG2 and HLE cells. It was noted that as a positive control, the TRAIL (TNF-Related Apoptosis Inducing Ligand) only induced apoptosis in HepG2, but not in HLE cells.
[00289] DBZIM triggered mitochondria-mediated apoptosis pathway
[00290] In order to understand the apoptosis pathway involved by DBZIM, caspase 3, 8, 9 activities were assayed using colorimetric assays. As shown in Fig. 23 A, caspase 8 activity was not affected by DBZTM, whilst caspase 9 and 3 activities were dose-dependently enhanced by DBZTM, pointing a mitochondria-mediated apoptosis pathway. However, DBZIM was found to cause a minimal release of cytochrome C from the mitochondria to the cytosol (Fig. 23B).
[00291 ] The mitochondria-mediated intrinsic apoptosis pathway is mainly regulated- by the
Bcl-2 and the IAP (Inhibitor of Apoptosis Protein) families. As shown with Western blotting in Fig.24, the anti-apoptotic protein BCI-XL and the pro-apoptotic protein Bak were slightly down- regulated and up-regulated by DBZIM respectively, whereas the other two members of the Bcl-2 . family, Bcl-2α and B ax, were not affected by the treatment. Notably in the IAP family, survivin was significantly suppressed by DBZIM, *whereas xIAP and cIAP were not affected (Fig. 24). Ia addition, HCS analysis revealed that survivin was translocated from the nucleus to the cytoplasm under the influence of DBZIM (Fig. 25).
[00292] DBZIM induced cytoplasm-to-nucleus translocation of AIF
[00293] Apoptosis-inducing factor (AIF) is normally localized in the mitochondria, and translocated to the nucleus upon induction of apoptosis. It induces mitochondria to release caspase 9. In nucleus, AIF also induces chromatin condensation and DNA fragmentation, a process known as caspase independent apoptosis pathway. DBZIM was found to induce the translocation of AIF to the nucleus by Western blotting (Fig. 26A). Immunocytostaining showed that AIF was translocated from the mitochondria/cytoplasm to the nucleus (Fig. 26B). It is worth noting that under DBZIM treatment the nuclear AEF (arrows) tends to be associated with the smaller condensed nuclei which are presumably going through the apoptosis. Quantitative analysis with HCS suggested that the DBZIM-induced translocation is dose-dependent (Fig. 26C).
[00294] DBZIM induced apoptosis through inducing ROS and AP-I
[00295] Reactive oxygen species (ROS) not only directly modifies cellular macromolecules, it also actively participates in various cellular signaling. An elevated level of ROS has been reported to trigger me programmed cell death by apoptosis. When treated with DBZIM at the rmllimolar range, HLE cells generated significant amount of ROS and coincidently underwent mass cell death (Fig. 27).
[00296] A sustained and elevated level of transcription factor activation protein 1 (AP-I) complex is known to participate in apoptosis. Western blotting (Fig. 28) of nuclear extracts from HLE cells showed that DBZIM significantly induced several key members of the AP-I complex, including C-Jun, Jun B, Jun D, and Fra-1, but not Fra-2.
[00297] DBZIM induced apoptosis and reduced pAKT in several other tumor cell lines
[00298] To investigate whether the effect of DBZIM is liver cell specific, tumor cells of lung, breast and gastric cancer cells were treated with DBZIM, and it was found that the same agent caused significant cell death in all the tumor cell lines tested. The IC50 value (determined at day 3) was 50 μM for H1299 (lung cancer), 50 μM for MCF-7 (breast cancer), and 40 μM for AGS gastric cancer at day 3 (Table 3). Interestingly, DBZIM showed much less cytotoxicity on the normal (noncancerous) lung cells (IMR90). Further, treatment of AGS and H1299 with DBZIM resulted in morphological (apoptotic look) changes and significant reduction in the number of cells (Fig 29A and 29B). It was also found that DBZIM significantly reduced the growth of two glioma cell lines, U87MG (human) and C6 (rat) (Fig. 30A and 30C), with IC50 (determined at day 2) for U87 MG and C6 being 2.1 mM and 1.0 mM, respectively.
[00299] To determine whether cells undergo apoptosis, Western blotting was first performed for a caspase-3/9 and PARP activation with proteins from MCF-7 and AGS treated with DBZIM. Results showed that DBZIM time-dependently increased caspase-3/9 and PARP activity, suggesting that the DBZM-treated cells undergo apoptosis in breast cancer cell line MCF-7 (Fig. 31A) and in wild-type p53 gastric cancer cell AGS (Fig. 31B). Similarly we observed a DBZIM-induced elevation of caspase 3/7 in C6 (by 3 folds at 2.0 mM) and in U87 MG (by 17 folds at 2 mM) (Fig. 3OB and 30D).
[00300] The Akt (also known as protein kinase B) kinase signaling pathway is activated in a variety of carcinomas of lung, breast, colon, and pancreatic and other tumor types through over- expressing the Akt protein itself and/or increasing phosphorylation of AM. This study provided evidence that DBZM is particularly efficacious in inducing apoptosis in AGS, H1299 and MCF-7 cells through a mechanism involving the reduction in the levels of me, phospho-AKT. Next it was investigated whether DBZM modulated the Akt pathway in H1299, MCF-7, AGS and NKN28, DBZIM treatment resulted in an appreciable down-regulation of the active form of Akt (phospho- Ser473-Akt) from 24 h to 72 h, without any changes in β-actin in all four cancer cell lines tested (Fig. 32). Taken together, these data suggest that DBZM is a potent suppressor of AKT-mediated signaling and is independent of p53 status.
[00301 ] DBZIM reduced tumor growth in a xenograft HCC mouse model
[00302] Several HCC cell lines (HepG2, Hep3B, Huh7, PLC, and HLE) were tested for their ability to efficiently induce HCC in nude mice, and it was found that Huh7 line was among the most efficient. Therefore a murine HCC model was established using the Huh7 cells for testing the antitumor efficacy of DBZM in vivo. Three weeks into the treatment, difference in tumor size was apparent between mice in the control and the treatment group (Fig. 33A). Measurement showed that the tumor volume in the DBZM-treated mice was significantly reduced by 40% (P<0.01) when compared to the control mice (Fig. 33B). It was noted that the body weight (including a reduction in the tumor weight) of the DBZM-treated mice decreased by 17% (P< 0.01) after three weeks of treatment (Fig. 33C).
[00303] Anticancer activities of additional IMSs
[00304] Determination ofIC50for 46 IMS in HLE cells
[00305] A total of 46 IMSs were cultured in the HLE cell line for 48 h to determine rough IC50 values for individual compounds. (Table 2) After the initial screening, seven of the MSs, compound C, compound 9, IBN-15, IBN-19, IBN-24, IBN-25 and IBN-32, were used for subsequent experiments.
[00306] Endogenous expression ofp53
[00307] The expression level of ρ53 in Hep 3B (ρ53null), HepG2 (p53 wild type), and HLE and PLC (p53 Mutant) hepatocarcinoma cells was determined by Western Blot. (Fig. 34). The expression of ρ53 was high in cells bearing mutant p53, as compared to cells with the wild type P53.
[00308] IMSs inhibited proliferation of HLE cells
[00309] The effect of IMSs (EBN-15, IBN-19, D3N-24, IBN-25 and JBN-32) on the proliferative properties of p53 mutant cell line HLE was examined. The cytotoxicity activity and proliferation inhibition of EMSs in tumor cells were measured by MTT. The treatment of HLE liver cancer cells with IMSs resulted in a dose dependent inhibition of cell proliferation. The control experiments witih DMSO alone (0.05% v/v) had no effect on cell proliferation. The ICS0 value for IBN-15, IBN-19, IBN-24, IBN-25, and IBN-32 in HLE cells was estimated to be 60 μm, 90 μm, 90 μm, 30 μm, and 20 μm at 72 h. While the IC50 value for EBN-15, EBN-19, IBN-24, IBN-25 and IBN-32 in HLE cells at 120 h was found to be 20 μm, 28 μm, 30 μm, 35 μm, and 20 μm. (Table 4) Microscopic observation on the cell cultures visually confirmed the inhibition on cell proliferation by IMSs. (Fig. 35)
[00310] IMSs induced apoptosis
[00311 ] As the data indicated that IMSs significantly inhibited cell growth in HLE cells,
Annexin V-PI dual-staining assay was performed in the same cell type under the same conditions to investigate whether the growth inhibition resulted from cell apoptosis. Cells undergoing apoptosis would stain positive for Annexin V-FITC and negative for PI (quadrant 4, Q4). Cells which stained positive for both Annexin V-FITC and PI (Q2) are either at the end stage of apoptosis or undergoing necrosis, and those which stained negative for both Annexin V-FITC and PI (Q3) were alive or undergoing undetectable apoptosis. In contrast, cell debris stained only for PI (Ql). As shown in Fig 36 A and B, IMSs IBN-15, IBN-19, EBN-24, IBN-25, and IBN-32 induced 33%, 29%, 29%, 21%, and 18% (Q2) cell apoptosis, respectively, whereas fewer apoptotic cells (7.2%) were observed for DMSO treated cells, confirming that the IMS-induced cell growth inhibition has a significant apoptotic component
[00312] IMSs changed cell cycle distribution
[00313] To analyze whether the MSs-induced inhibition of cell growth in HLE cells was accompanied by alterations in cell cycle distribution, the percentage of cells in the different phases of, cell cycle and apoptotic index were analysed by flow cytometry. When cells were treated with EBN-15, E3N-19, EBN-24, EBN-25, and IBN-32 at a concentration corresponding to their respective TC-50 value (i.e., 60 μm, 90 μm, 90 μm, 30 μm, and 20 μm) for 72 h, a significant accumulation of cells in the sub-GO phase and a corresponding reduction in the number of cells in the G2/M phase was found. The representative results are shown in Fig. 37A, 37B and 37C). Two percent of the cells in the control culture (with DMSO) were in sub-GO phase, whereas the numbers dramatically climbed up to 20% in the IMS-treated cultures. Similarly in control culture, 50 % cells were in the Gl phase, 24% were in the S phase, and 30% were in the G2/M phase at 72 h, whereas in the IMS- treated cultures (with IBN-15, IBN-19, IBN-24, IBN-25, and IBN-32), 42%, 29%, 28.6%, 34.5 %, and 32.3 % of the cells were in the Gl phase; and 19.7%, 19.8%, 11.34%, 14.3%, and 14.6% in the S phase; and 21.%, 23.7%, 16.3%, 19.4%, and 18.9% in the G2/M phase, respectively.
[00314] IMSs induced apoptosis via caspase activation
[00315] To further determine whether the apoptotic activity of IMSs is due to caspase activation, Western blot analysis was performed with total protein lysates from HLE cells. It was observed that IBN 19, 24 and 25 induced significant activation and cleavage of caspase 3 and caspase 9 and PARP (Fig. 38), suggesting that the compounds induce cells to undergo apoptosis.
[00316] IMSs increased the expression ofp53 and phosphorylated p53 (Serl5, 20, 46 and Ser
392)
[00317] In order to determine whether the activation of the p53 pathway was involved in
IMS-induced apoptosis, analyses of p53 and its down stream effectors were performed in HLE cells, which carry the mutant p53. Immunocytochemical analyses revealed that IMSs (IBN-19 and IBN-24) induced p53 expression and accumulation in the nucleus (Fig. 39).
[00318] Because phosphorylation at the Ser 15 site of p53 by ATM is often observed when cells receive DNA damage signals. It was speculated that ATM may also respond to IMSs in the HLE cells. Tt has been reported that chemical agents that damage DNA act through posttranslational modifications of p53 and activate its downstream targets in various human cancer cells (Banin et al. 1998; Canman et al. 1998). The present results also demonstrated that these IMSs induced p53 modifications, such as phosphorylation at Ser-15 (Fig. 40) due to up-regulation of ataxia telangiectasia-mutated (ATM) kinase gene. Furthermore, IMSs induced phosphorylation at position at Ser-20, -46 and -392. These posttranslational modifications of p53 appear to be responsible for the cell cycle arrest (Fig. 40).
[00319] IMSs induced the execution of apoptosis through activation of the mitochondrial pathway [00320] To investigate the mitochondrial events involved in IMSs-induced apoptosis, the changes in the levels of pro-apoptotic protein Bax, and anti-apoptotic protein Bcl-2 were analyzed. Immunoblot analysis showed that treatment of HLE cells with various MSs increased Bax protein level and simultaneously decreased Bcl-2 level (Fig. 41).
[00321 J Real-time RT-PCR profiling of gene expression in IMS-treated cells
[00322] To obtain more information on how the IMSs influence other gene expression in the HLE cells, 84 genes related to p53 signal transduction were profiled by using the RT2 Profiler array (Super Array Bioscience) (Fig 42A, B, C, D, E). The array includes keys genes relevant to p53, apoptosis, cell cycle, cell growth, proliferation, differentiation, and DNA repair. It was found that ATM gene expression at 48 h after IBN- 19, IBN24 and IBN-25 treatment was significantly increased by 36-, 4.6- and 9-folds respectively, when compared to that of the control (Fig 42E). These data show that IMSs induce transcriptional up-regulation of ATM gene, which in term elicits a specific p53 phosphorylation at Ser-15 in the HLE cells.
[00323] The results demonstrated that IBN-24 and IBN-25 significantly down regulated anti- apoptotic genes BCl-2 (Fig 42A). The results from the quantitative RT-PCR analysis also indicated that SESN2 gene was markedly induced by IBN-19, as compared to untreated (Fig. 42B). Further, the results also demonstrated that cell proliferation genes McIl, Egrl, Foxo3, Jun, Mdm4, NfI, PrmlD, Sesnl, E2fll, Prkca and, Brcal were significantly down regulated in the IBN-24 and IBN- 25 treated cells as compared to control cells. (Fig 42 D) The important cell cycle regulatory genes cdk4, cdc25A, cdc2, e2fl, and Hk2 were significantly downregulated. (Fig 42C)
[00324] Compound C (DBZMIM) and compound 9 (MABZEd) inhibit proliferation of gastric and breast cancer cells
[00325] In this study, the anti-cancer effects of compound C and compound 9 on p53 mutant gastric cancer cell line MKN 28 and p53 mutant breast cancer cell line MDAMB231, as well as on wild type ρ53 breast epithelial cells (MCF-IOA) was preliminarily explored. First, the cytotoxic effect of these drugs on these cells was investigated using the MTT assay. Cells were seeded into 96 well plates and treated with increasing concentrations of these synthetic organic molecules for 72 h, and then the cytotoxic effect was measured. In response to these synthetic organic molecules, the normal MCF-IOA cells showed resistance to both organic molecules with no IC50 (concentration of the drug that leads to 50% lethality) (Table 7). Both the cancer cell lines MKN 28 and MDAMB 231 were the most sensitive towards compound 9, with IC-50 being 10 and 20 μM (Tables 5 and 6). On the other hand, compound C killed both MKN 28 and MDAMB231 with the same IC50 of 35 μM (Tables 5 and 6). These IC50 values were similar to those obtained with a commonly used cancer drag 5-FIurouracil (5-FU). Morphology of the cells treated with compound C and compound 9 respectively was shown in Fig.43 A and 43B.
[00326] Compound 9 inhibited proliferation of HCC Huh.7 cells in mice
[00327] Several HCC cell lines (HepG2, Hep3B, Huh7, PLC, and HLE) were tested for their ability to efficiently establish HCC in nude mice, and found that Huh7 line was among the most efficient. Therefore a murine HCC model was established using the Huh.7 line and one of the MSs (compound 9) was tested to assess its anti-tumor efficacy. Three weeks into the treatment, significant difference in tumor size was apparent between mice in the control and the treatment group (Fig 44A). Measurement showed that the tumor volume in the C9-treated mice was significantly reduced by 43% and 62% (P<0.01) at 0.6g/l and 1.5g/L respectively, when compared to the control mice (Fig. 44B). No change in body weight or gross pathology was noted for the C9- treated mice, when compared to the control mice (Fig. 44C), implying lack of significant general toxicity associated with the C9 under the current dosing scheme.
[00328] Discussion
[00329] Anti-oxidative, anti-inflammatory and anti-fibrotic properties of IMSs
[00330] Toxicity of IMSs
[00331 ] The IC50 of the most frequently tested compound in this study, DBZM, was 1.7 mM, and the IC50 values for DPIM and DBIM with aliphatic substitutions were 2-4 mM. The in vivo toxicity for DPIM and DBIM was roughly one-tenth of that for DBZOVL In contrast, the IC50 values for natural antioxidants EGCG and genistein in HSCs were much lower (25-75 μM) (Kang et al. 2001, Chen et. al 2002, Chen et. al 2003, Higashi et al. 2005, Zhang et al. 2006). Intriguingly, preliminary in vitro and in vivo data seemed to suggest, in contrast to common belief, that the synthetic IMSs might have a better safety profile than the natural anti-oxidants. The mild toxicity of this class of compounds certainly helped to put forth the prospect of using IMSs as the basic units to synthesize additional candidates with novel properties and functionalities tailored to various specific therapeutic needs.
[00332] IMSs Demonstrated Anti-Oxidative Property Through Neutralization of ROS and Induction of Phase II Anti-Oxidative Enzyme
[00333] Cells or tissues maintain a redox hemeostasis when the rate of ROS production and scavenging capacity are essentially in balance. Changes in oxidant/anti-oxidant balance will trigger xosponsive redox signaling. It has been reported that oxidative stimuli can induce expression of anti- oxidative defense enzymes such as SODs, CAT and GPx, etc., to restore the original redox homeostasis or to reach a new equilibrium (Droge 2002). Redox regulation is also partly mediated through the cellular GSH and the GSH/GSSG redox state. GSH plays a key role in cellular anti- oxidative processes by serving as an electron donor to GPx in the reduction of hydrogen peroxide to water, and as a nucleopMIic co-substrate to GST in the detoxification of xenobiotics. Anti-'oxidants exert their effect mainly through three different pathways: (1) neutralization of cellular ROS generated during metabolism and immune response, (2) induction of endogenous anti-oxidative enzymatic activity, and (3) chelation of iron or copper ions that catalyze the generation of hydroxyl radical. GPx, CAT and SOD, which constitute the first line of cellular anti-oxidative defense, are directly involved in the neutralization of ROS. GPx enzyme reduces H2O2 to H2O, while GSH functions as a cofactor and is consequently oxidized to GSSG. CAT is a peroxisomal enzyme and converts H2O2 to H2O. SOD catalyzes the dismutation of superoxide. There are three forms of SOD: extracellular and intracellular copper/zinc (Cu/Zn) SODs, and a mitochondrial manganese (Mn) SOD. GST is a phase π anti-oxidant enzyme. It has been reported that activation of HSCs is associated with the loss of GST activity in culture (Whalen et. al 1990).
[00334] La this study, IMSs effectively attenuated ROS (in particular H2O2 and lipid peroxides, which react to 2',7'-dichloro-fluorescein diacetate (DCF-DA) probe (Halliwell et al. 2004) in a dosage-dependent manner. While the level of GSH remained relatively constant, the amount of GSSG was dramatically decreased, resulting in a significant increase in the GSH/GSSG ratio. Moreover, the enzymatic activities of GPx and CAT, which are responsible for reducing hydrogen peroxide to water, were slightly attenuated. Taking all the data together, it is proposed that IMSs might have mainly exerted their anti-oxidant effect through the direct neutralization of ROS, such as hydrogen, peroxide. Therefore, the burden on cellular anti-oxidative defense was alleviated by the exogenous synthetic anti-oxidative IMSs, resulting in a decrease in the expression of first-tier anti-oxidative enzymes. In the meantime, the effect of IMSs on GSH synthesis and anti- oxidative enzymes showed a general pattern of attenuation at low dosages, less potent attenuation and even enhancement at high dosages. This suggested that DBZM worked through different mechanisms at low and high dosages. At a low dosage, the anti-oxidative pathway predominated by direct reaction to remove H2O2 so that GPx and CAT activities were reduced. In contrast, there might be a synergistic effect between direct neutralization of H2O2 and induction of anti-oxidant enzymes at a high dosage. It was reported that a reduction in the battery of anti-oxidative enzymes GPx, CAT and SOD as observed in the experimental cholestasis can be reversed by the endogenous anti-oxidant melatonin (Padillo et al. 2004). Interferon-alpha has been shown to increase GPx activity in activated HSCs (Lu et al. 2002). On the other hand, it has been shown that oxidative stimuli could induce anti-oxidative enzymatic activities (Droge 2002). One recent paper also showed that a high dosage of NAC (30 rtiM) shifted cellular redox state towards oxidative stress, as indicated by enhanced reduced glutathione, oxidized glutathione, GPx activity and ROS production in endothelial cells. In this study, high-dosage IMSs showed a trend of enhanced GPx and CAT activities. This result, along with other findings that high-dosage IMSs imposed greater effect in attenuating ROS production (Figs. 2a and 2b) and GSSG (Figs. 3c and 3d), and inducing GSH/GSSG ratio (Figs. 3e and 3f), suggested that IMSs at high dosage enhanced cellular defense activity against oxidative stress by synergizing neutralization of ROS and induction of endogenous anti-oxidant enzymes. In addition, IMSs had little effect on the total SOD (Gu/Zn-SOD and Mn- SOD) activity, suggesting that the compounds did not react with superoxide and have little influence on the cellular response to remove superoxide.
[00335] Meanwhile, phase II enzymes such as total GST were enhanced in activity under the treatment of IMSs. It was known that GSTs protect the cells against oxidative toxicants by conjugating the xenobioti.es to GSH, thereby neutralizing their electrophilic sites and rendering the product water-soluble. The enhancement of GST activity indicated that the cells have better capability of getting rid of oxidants such as lipid peroxidation products, thereby minimizing the cellular oxidative injuries.
[00336] The anti-oxidative property of IMSs was also demonstrated in primary HSC cells. DMSO induced cellular oxidative stress, and imposed lethal effect on primary HSC cells. The presence of IMSs effectively protected the primary HSC cells from dying in the culture medium containing 0.2% (v/v) of DMSO. This finding implies a potential application of this group of compounds in cell-based drug screening assays and tissue engineering.
[00337] IMSs showed Anti-Inflammatory Effect Through Suppression of NF- KB and IL-6
[00338] Transcription factor NF-κB is found in the cytosol bound to inhibitory protein known as IKB. When the cells are under various stress conditions, such as infection, inflammation and tissue repair, NF-KB is activated and translocated into the nucleus where it binds to DNA and regulates the transcription of gene-encoding proteins involved in immune or inflammatory responses (Vasiliou et al. 2000, AIi et al. 2004). The activation of NF-κB is associated with phosphorylation and subsequent degradation of IKB. The NF-κB -responsive site has been identified and characterized in the promoters and enhancers of many genes, including IL-6 and ICAM-I (AIi et al. 2004). NF-KB is also known to be sensitive, to oxidative stress. Most agents activating NF-KB are either modulated by ROS or oxidant themselves. It has been reported that the treatment of antioxidant resveratrol (Chavez et al. 2007) or vitamin E (Liu et al. 1995) attenuated NF-κB elevation induced in carbon tetrachloride experimental fϊbrotic rodents.
[00339] To test whether IMSs are able to suppress NF-κB activation and in turn regulate immune and inflammatory responses, the protein expression of active NF-κB was assayed by Western blotting and NF-κB-responsive IL-6 gene transcription. Our data showed that MSs could effectively interrupt the activation of NF-KB, and as a result, down-regulate the gene expression of IL-6 (Figs. lOa-c and 11). These data indicated that IMSs have anti-inflammatory effect by down- regulating IL-6 transcription through NF-KB signaling.
[00340] IMSs Showed Anti-Fϊbrotic Properties Through Suppression of HSC Activation
[00341 ] HSCs are well-recognized cellular regulators in the development of hepatic fibrosis. HSC activation, associated with enhanced secretion of pro-fibrogenic, pro-inflammatory cytokines (such as TGF-βl and IL-6) is characterized by the over-expression of SMAA, and results in the over-deposition of ECM proteins including collal and fibronectin. The anti-fibrotic effects of IMSs were evident by the suppression of HSC activation markers (GFAP and SMAA) and fibrotic endpoints (collal and fibronectin) in a dosage- and time-dependent manner. A transiently induced SMAA expression was observed under the treatment of high-dosage TDBZIM (Fig. 7b). The underlying mechanism was not clear, but might be related to the radical-like nature of NHC with bulky N-substitαents (Bourissou et al. 2000). SMAA might have a transient over-response to excess radicals. Nevertheless, IMSs of moderate concentrations were generally able to attenuate SMAA expression (Figs. 6a, 6b and 14b). IMSs also consistently suppressed the mRNA and protein expression of collal and fibronectin (Figs, δa-b, 7c-d, 13 and 14c-d), which as a net effect prevented or slowed down the progression of fibrosis.
[00342] TGF-β 1 is the most potent pro-fibrogenic- cytokine, which is over-expressed in activated HSCs. TGF-βl signaling is initiated by the binding of the active form of TGF-βl to type π receptor, and subsequently leading to the phosphorylation of type I receptor (TGFβ RI). Activated TGFβ RI recruits- Smads protein, and propagates the downstream signal to regulate the gene expression of matrix proteins (Fu et al. 2006). To understand IMSs' effect on treating liver fibrosis, TGF-βl and TGFβ RI in T6 cells under IMS treatment were measured. The data showed that IMSs could effectively interfere with TGF-βl signaling by suppressing the gene and protein expressions of total TGF-βl and TGPβ RI (Figs. 9a-c and Fig. 13). It was recently reported that EGCG could decrease the active form of TGF-βl in primary HSC cells (Fu et al. 2006). In the •present study it was also shown that EGCG could suppress the production of total TGF-βl protein in T6 cells (Fig. 13). Some IMSs, particularly DPIM, showed greater suppression of TGF-βl and TGFβ RI than EGCG under the test conditions. Resveratrol, another anti-oxidant from grape skin, has been reported to effectively attenuate TGF-βl protein elevation induced in an experimental fibrotic rat model (Chavez et al. 2007).
[00343] Activation of HSC is also associated with elevated levels of NF-κB and NF-κB- responsive genes, including IL-6 (Mann et al. 2006). The role of NF-κB is to maintain HSCs in the activated state and promote a chronic wound healing response. In fact, NF-KB has been implicated in various aspects of liver disorders, including hepatic inflammation, fibrosis, and the development of hepatocellular carcinoma (Elsharkawy et al. 2007). In this study, IMSs greatly suppressed active NF-KB p65 protein (Fig. 11), while not affecting the cytosolic NF-κB P65 level (data not shown). AP-I is a homodimer or heterodimer composed of at least one Jun family protein (c-Jun, JunB and JunD) and another member from the Fos family (c-Fos, Fral and Fra2). It has been reported that JunD is functionally the most important AP-I factor in activated HSCs required for the induction of IL-6 andthe tissue inhibitor of metalloproteinase I (TIMP-I) gene transcription (Smart el al. 2001). It is known that HSC T6 cells express all members of Jun and Fos proteins (Zhang et al., 2006). Under the influence of IMSs, c-Fos, JunD and Fra-1 levels were attenuated (Fig. 11). In line with the down-regulation of JunD, IL-6 gene transcription was suppressed (Figs. lOa-c). In short, IMSs might prevent hepatic fibrosis through the suppression of HSC activation, and by decreasing the deposition of ECM proteins (collal and fibronectin) and the production of pro-fibrogenic cytokines (TGF-β 1 , TGFβ RI and IL-6) mediated through NF-κB and AP- 1 signaling.
[00344] NHC Precursors Could Be Drug-Like Compounds
[00345] ROS has been implicated in many physiological conditions, including neurodegenerative diseases, cardiovascular diseases, stroke, heart attack, cancer, aging and fibrosis, etc. Therefore, anti-oxidation has been pursued as a therapeutic strategy for a number of diseases. Dietary anti-oxidants have been widely used to ameliorate excessive oxidative stress both in animal models and humans. For example, resveratrol has been shown to extend the lifespan of various species, and to be effective at improving the health and survival of mice on a high-calorie diet (Baur et al. 2006). Tea polyphenols (EGCG being the most abundant catechin) have been shown to inhibit carcinogen-induced DNA damage in animal models of skin, lung, colon, liver and pancreatic. cancers (Frei et al. 2003). However, stringent scientific proof for the efficacy of natural anti- axidants has not been established (Droge et al. 2001) due to various factors. Some of the notable limitations for using natural anti-oxidants as therapeutics include low potency and fast turnover during metabolism. By comparison, less effort has been devoted towards studying synthetic antioxidants due to safety concerns. Nevertheless, some progress has been made in this direction. For example, natural anti-oxidant has been modified to enhance its potency (Keum et al. 2007). Synthetic mimics of SOD and catalase have been shown to be effective in rodent models of ischemia and Parkinson's disease (Peng et al. 2005). Even more encouragingly, a class of nitron- free radical trap agents, alpha-phenyl-N-tert-butyl-nitron (PBN) and disodium 2,4-disulfophenyl-N- tert-butylrdtrone (NXY-059), has been shown to be potent neuroprotective agent (Maples et al. 2004), and has demonstrated anti-cancer activity in hepatocellular carcinoma through its antiinflammatory properties (Floyd 2006). These examples suggest that it is possible to develop therapeutic drug candidates based on synthetic anti-oxidants. It is believed that this report represents the first effort towards exploring the potential therapeutic effect of IMSs. IMSs are precursors of NHCs, in which the electronic structure and stability, and thus, therapeutic safety and efficacy can be tuned by varying the N-substituents. NHCs with bulky substituents favor a triplet ground state and exhibit radical-like reactivity, while those with smaller substituents adopt a singlet ground state and show both nucleophilic and electrophilic behavior (Bourissou et al. 2000).
[00346] In this study, IMSs demonstrated potent anti-oxidative, anti-inflammatory and anti- fibrotic properties in cultured HSCs. At the same time, IMSs showed relatively low and tunable toxicity with IC50 in the range of 35 μM to 3.1 mM. It is hypothesized that the anti-fϊbrogenic and anti-inflammatory effects of the IMS compounds are derived through the anti-oxidative property targeted at inhibiting HSC activation. The radical scavenging property of the MSs is presumably originated from the product after a series of chemical reactions including:(l) the spontaneous conversion of IMSs to NHCs preferably under a basic condition, (2) the interaction of the NHCs at the carbon 2-position with free radicals such as ROS resulting in the formation of intermediate active radicals and the neutralization of the free radicals.
[00347] The findings of this study support this hypothesis. Although the underlying molecular mechanism is not yet well understood, the effects of IMSs are shown to be mediated through NF-KB and AP-I transcription factors, and TGF-βl and TL-6 signaling pathways. Large- scale screening of lead compounds for the treatment of liver fibrosis can be performed using the surrogate GFAP biomarker driving a fluorescent reporter. The radical scavenging and the antiinflammatory properties are central to IMSs' therapeutic effect in treating liver fibrosis in this study. Further exploration may reveal therapeutic potential for this group of compounds in treating other degenerative and aging-related diseases.
[00348] Effect of DBZIM on Hepatocellular Carcinoma and Other Tumour Cells
[00349] Hepatocellular carcinoma (HCC) is known to develop over liver insults, which leads to hepatocyte inflammation and regeneration, matrix protein remodeling, fibrosis/cirrhosis and eventually HCC. HCC is inherently chemotherapy-resistant and is know to express the drug- resistant gene MDR-I. (Huang et al. 1999, Kato et al. 2001) Advanced HCC also shows poor prognosis even after a successful surgical resection (Ariizumi et al.2004). The data showed that 1 IMS compound DBZIM can induce apoptosis an in both differentiated (HepG2) and undifferentiated (HLE) HCC cell line without significant cytotoxicity (Fig. 22). DBZM can also inhibit the proliferation of HLE (Fig. 21 A) via cell cycle arrest at G0/G1 phase (Fig. 21B).
[00350] Apoptosis can be triggered through either the extrinsic pathway or the intrinsic pathway. The extrinsic pathway is initiated through the simulation of transmembrane death receptors such as Fas receptors and activation of caspase 8 which leads to caspase 3/7 cleavage. The intrinsic pathway is initiated through the release of apoptosis activators from mitochondria and activation of caspase 9 is involved to cleave caspase 3/7 which leads to apoptosis. Ih this study, enzyme activity assays showed that DBZIM induced the activities of caspase 9 and 3 but not caspase 8 (Fig. 23). Further experiment by Western blotting showed that DBZTM also regulated the expression of Bcl-XL and Bak to induce the apoptosis (Fig. 24). This evidence suggests that DBZIM mainly caused the mitochondria-mediated apoptosis through the regulation of Bcl-2 favoring the occurrence of apoptosis. Meanwhile, it was found that AIF but not cytochrome C (Fig. 23d and Fig. 26) was released from mitochondria to activate caspases and/or cause chromatin condensation / DNA fragmentation. AIF has been generally implicated in the caspase-independent mode of cell apoptosis and been proposed as a new drug target. (Mignotte et al. 1998, Lorenzo et al. 2007).However it has also been shown to act in a caspase-dependant manner. (Amoult et al. 2003) DBZIM was also shown to down-regulate survivin (Fig. 24) and induce cytoplasm location of survivin (Fig. 25). Survivin is one member of IAPs protein family, expressed during human foetal development and in several neoplasms, but not in normal tissue except in thymus and placenta. It has been shown that nuclear translocation of surviving in HCC promotes the cancer cell growth. (Moon et al. 2003) Nuclear and cytoplasmic expression of survivin has also been related to prognosis of pancreatic cancer (Tonini et al. 2005). DBZIM perturbated the regulatory mechanism by translocation of survivin and suggesting a possible new angle for searching for HCC chemotherapy. [00351 ] The alteration of mitochondria function is associated with the early phases of the cell death. Meanwhile, observed inhibition of the mitochondrial respiratory chain causes the over production of ROS, which mediates the death signaling pathway. (Mignotte et al. 1998) The mode of cell death, either by apoptosis or necrosis is largely dependant on the level of intracellular ROS . Also it is difficult to differentiate whether ROS is the result or the cause of the killing process or both. In this study, it was interesting that MSs of different concentration had different physiological effect, in terms of ROS production. In HCC cells, when DBZIM was in the micromolar range, it attenuated the ROS level slightly, which agreed with the findings in HSC-T6 cells. When DBZIM was at much higher concentration in the milimolar range, it promoted the accumulation of ROS (Fig. 27). Meanwhile, accompanying the ROS production, some members of the transcription factor AP-I complex was found to be up-regulated by DBZM (Fig. 28). It was reported that the integration of oxidative stress and the up-regulation of JNK/c-Jun/AP-1 cascade is necessary to promote the cell death (Singh et al. 2007). Taking these results together, it is suggested that the cell killing caused by DBZIM at high dose may be mostly due to apoptosis and ROS induction.
[00352] The observations that DBZM not only inhibited cell proliferation in HCC lines, but also several other tumor cell lines commonly used in various cancer studies, through multiple mechanisms (cell cycle arrest, apoptosis, ROS, AP-I, inhibition of pAKT, etc.) provided further support for the speculation that DBZIM indeed exerts a number of beneficial anti-tumor properties in vitro. Its potential in vivo efficacy will need to be tested in various specific tumor models for confirmation.
[00353] In the initial in vivo trial, DBZM was administered to a HCC xenograft mouse model (Huh7cells in nude mice) via drinking water, after the tumor growth was evident and at the log phase about 8 weeks after cell inoculation. Several observations were made. First, 3 weeks of treatment with DBZIM resulted in 40% reduction in tumor volume, which is a preliminary and significant demonstration for using DBZM as an effective anti-tumor agent. Second, the effective dose observed for in vivo study (2g/I) is much lower that those for in vitro cell culture study (generally in 1-2 mM). This disparity could suggest. that DBZM may have a very different PK/PD profile in vivo than in vitro. Third, the effectiveness achieved by dosing the agent in drinking water (with weekly replacement) indicates that the DBZM is rather stable in water and can be absorbed through gastrointestinal (GI) tract effectively to enter the general circulation. Finally, we also noticed a significant loss in body weight (by 17%) in the 3-week treatment with DBZM, which may suggest some type of general toxicity in the mice, though no gross pathological abnormality could be identified in all major organs. Perhaps an alternative dosing scheme using this agent can be further explored to achieve maximal therapeutic effect, yet with minimal side effect.
[00354] In conclusion, by using DBZM as a model compound for research in IMSs, it has been demonstrated that this agent has anti-tumor activity in vitro and in vivo, which warrants further efforts in chemically synthesizing and biologically characterizing additional IMS compounds with different substituents on the imidazolium ring, in order to identify candidates with more potent efficacy with reduced side effect.
[00355] Anticancer activities of additional IMSs
[00356] The ability of "DBZDVt-like" candidate compounds to induce cancer cell death was investigated and molecular mechanisms (apoptosis and cell cycle arrest) underlying this ability were dissected. It was discovered that the basic anti-tumor properties (apoptosis and cell cycle arrest) embedded in the model DBZIM were preserved in the some of additional IMSs with different subgroups, mainly including EBN-15, -19, -24, -25, -32, compound C (DBZMM) and compound 9 (MABZM).
[00357] It was found that IMSs effectively inhibit tumor cell growth concomitant with induction of cell cycle arrest and apoptosis. IMSs activate ATM system and subsequently the phosphorylation and stabilization of p53. A correlation was drawn between the level of p53, phospho-p53 (Ser-15) and ATM in HLE liver cancer cells. It is well known that ATM and ATR are activated by DNA damage. Activation of the ATM pathways has long been associated with the cell cycle checkpoint induced by several DNA damage/genotoxic stress. Once activated, ATM phosphorylates various downstream molecules such as p53, MDM2, Chkl, Chk2, resulting in cell cycle arrest or cell death. Tumor suppressor gene p53 is a key element in the induction of cell cycle arrest and apoptosis following DNA damage or cellular stress in human cells and cell cycle arrest. (Saito et al. 2002, Hofseth et al, 2003) In this study, it has been shown that treatment of HLE cells with IMSs resulted in the accumulation of p53 and phospho-p53 (Serl5). It has also been found that the induction of ATM gene expression causes DNA damage andp53 activity, suggesting that p53 induced by DNA damage play an important role in IMSs-mediated cell cycle arrest. Indeed, this result is consistent with the report of Canman et al., (1998) who showed that phosphorylation of p53 at serine 15 was in response to DNA damage. The in vitro studies also showed that phosphorylation of this specific site is critical for the p53 response and subsequent IMS-induced anticancer activity. It was also found that MSs decrease the expression of a number of genes important for cell cycling, including Cdc25A, Cdc2, and SESN2.
[00358] Up-regulated p53 protein can activate the pro-apoptotic protein BAX and inhibits Bcl-2 to cause mitochondrial dysfunction and cytochrome c release, stimulating the mitochondrial apoptotic pathway (Melino et al., 2004). In this study, it was demonstrated that IMSs inhibited BcI- 2 and activated. Bax in the HLE cancer cells. It was found that IMSs effectively inhibit tumor cell growth, concomitant with induction of cell cycle arrest and apoptosis. Furthermore, the fact that IMSs do not exhibit any significant toxicity in normal lung and breast cells suggests that IMSs possesses selectivity between normal and cancer cells. The data on caspase-3 activation strongly suggest that the mechanism of IMS induced apoptosis in these liver cancer-derived cell lines might be mediated by caspase-dependent signaling leading to caspase-3 activation. In the present study, it was shown that IMSs have cytotoxic activity toward cancer cells harboring mutant p53 and can activate mutant p53. In the present study, the inhibitory effects of IMSs to malignant tumor cell lines with mutant p53 status was investigated. The mitochondrial apoptotic pathway has been described as important in signalling apoptotic cell death for mammalian cells (Pilch et al 2003; Kluck et al., 1997). Following the treatment of HLE cells with IMSs, it was observed that IMSs treatment resulted in a significant increase of Bax expression, and a decrease of Bcl-2 suggesting that changes in the ratio of pro-apoptotic and anti-apoptotic Bcl-2 proteins might contribute to the apoptosis-promotion activity of IMSs. The activation of caspase-3 and caspase-9 was also observed after HLE cells were treated with IMSs. These results confirm that IMS-induced apoptosis is associated with regulation of BAX and Bcl-2 proteins.
[00359] Here it is demonstrated that, in addition to DBZIM, compound 9 or C9 displays a potent anti-tumor activity in the HCC xenograft model, and yet shows no apparent general toxicity. This preliminary evidence provides proof of the concept that chemical modification based on a core DBZIM structure can significantly improve the anti-tumor efficacy and reduce side effects. For example, C9 at concentration (1.5g/l) can have much higher potency than DBZTM at a higher concentration (2g/l), when administered via same route for the same period of time.
[00360] Thus the present study demonstrated that: (1) human liver cancer HLE cells are highly sensitive to growth inhibition by IMSs. (2) IMSs reduced survival of HLE cells via cell cycle arrest and apoptosis induction in a p53-dependent manner, and by decreasing the expression of Cdc2, Cdc25A. (3) IMSs inhibited cell growth in the HLE cells via activation of ATM, which stabilizes p53 by phosphorylation of p53 at Serl5 and decreasing the interaction of p53 and MDM2. (4) IMSs triggered mitochondrial apoptotic pathway by regulation of Bcl-2 and BAX proteins expression. These findings suggest that IMSs are a promising class of low toxic chemopreventive agents against human liver cancer cells, and other tumor cell types.
[00361 ] All publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. The citation of any publication is for its disclosure prior to the filing date and should not be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention.
[00362] Concentrations given in this specification, when given in terms of percentages, include weight/weight (w/w), weight/volume (w/v) and volume/volume (v/v) percentages.
[00363] As used in this specification and the appended claims, the singular forms "a", "an" and "the" include plural reference unless the context clearly dictates otherwise. As used in this specification and the appended claims, the terms "comprise", "comprising", "comprises" and other forms of these terms are intended in the non-limiting inclusive sense, that is, to include particular recited elements or components without excluding any other element or component. Unless defined otherwise all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention belongs.
References
AIi S, Mann D A. Signal transduction via the NF-κB pathway: A targeted treatment modality for infection, inflammation and repair. Cell Biochem Funct 2004; 22:67-79.
Agarwal MX., Taylor W.R., Chernov M. V., Chernova O.B. and Stark, G.R. The p53 network, J. Biol. Chem. 273 (1998), pp. 1-4.
Amoult D, Gaume B, KarbowsM M, Sharpe J C, Cecconi F, Youle R J. Mitochondrial release of AIF and EndoG requires caspase activation downstream of Bax/Bak-mediated permeabilization. EMBO J. 2003; 22(17):4385-4399.
Anderson C.W., and Appella E. Signaling to the p53 tumor suppressor through pathways activated by genotoxic and non-genotoxic stresses in: RA Bradshaw, E.A. Dennis (Eds.), Handbook of cell signaling, vol.3 Academic Press, New York, 2003.
Apella E., and Anderson CW. Post- translational modifications and activation of p53 by genotoxic stresses, Eur. J. Biochem. 268(2001) 2764-2772.
Arϋzumi S, TakasaM K, Yamamoto M, Ohtsubo T, Katsuragawa H, Katagiri S. Histopathologic differentiation of the main nodule determines outcome after hepatic resection for synchronous multi-centric hepatocellular carcinomas. Hepatogastroenterology. 2004; 51:500-504.
Avila MA, Berasain C, Sangro B, Prieto J. New therapies for hepatocellular carcinoma. Oncogene 2006; 25:3866-3884.
Bachem M G, Meyer D, Melchior R, et al. Activation of rat liver perisinusoidal lipocytes by transforming growth factors derived from myofibroblastlike cells. A potential mechanism of self- perpetuation in liver fibrogenesis. J Clin Invest 1992; 89:19-27. Baker M V, Barnard P J, Berners-Price S J, Brayshaw S K, Hickey J L, Skelton B W, White A H. Cationic, linear Au(I) N-heterocyclic carbene complexes: Synthesis, structure and anti- mitochondrial activity. Dalton Trans 2006; 3708-3715.
Banin S., Moyal L., Sheih S., Taya Y., Anderson C.W., Chessa L., Smorodinsky NJ. , Prives C, Reiss Y., Shiloh Y, and Ziv Y. , Enhanced phosphorylation of p53 by ATN in response to DNA damage, Science 281(1998) 1674-1677.
Barnard P J, Baker M V, Berners-Price S J, Day D A. Mitochondrial permeability transition induced by dinuclear gold(I)-carbene complexes: Potential new antimitochondrial antitumour agents. J Inorg Biochem 2004; 98:1642-1647.
Bataller R, Brenner DA (2005) Liver fibrosis. J CUn Invest 115(2): 209-218.
Baur J A, Pearson K J, Price N L, Jamieson H A, Lerin C, Kalra A, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature 2006; 444: 337-342.
Bergamo A, Sava G. Ruthenium complexes can target determinants of tumour malignancy. Dalton Trans. 2007; 1267-1272.
Bourissou D, Guerret O, Gabbai F P, Bertrand G. Stable carbenes. Chem. Rev. 2000; 100:39-91.
Boydston A. J. , Williams K. A., Bielawski C. W., J. Am. Chem. Soc, 2005, 127, 12496.
Britton R S, Bacon B R. Role of free radicals in liver diseases and hepatic fibrosis. Hepatogastroenterol 1994; 41:343-348.
Bykov, VJ; Issaeva, N; Shilov, A; Hultcrantz, M; Pugacheva, E; Chumakov, P; Bergman, J; Wiman, KG; Selivanova, G. Restoration of the tumor suppressor function to mutant p53 by a low- molecular-weight compound. Nat Med. 2002; 8:282-8.
Canman C.E., . Lim D.S, Cimprich K.A., Taya Y., Tamai K., Sakaguchi K. A , Appella E., Kastan M.B., Siliciano J.D, Activation of the ATM kinase by ionizing radiation and phosphorylation of p53 Science 281(1998) 1677-1679
Cao X G, Li X X, Bao Y Z, Zing N Z, Chen Y. Responses of human lens epithelial cells to quercetin and DMSO. Invest Ophthalmol Vis Sci.2007;48(8):3714-3718.
Caron de Fromentel, C; Gruel, N; Venot, C; Debussche, L; Conseiller, E; Dureuil, C; Teillaud, JL; Tocque, B; Bracco, L. Restoration of transcriptional activity of ρ53 mutants in human tumor cells by intracellular expression of anti-p53 single chain Fv fragments. Oncogene. 1999; 18:551-7.
Casanovas A, Olmos G, Ribera J, Boronat M A, Esquerda J E, Garcia-Sevilla J A. Induction of reactive astrocytosis and prevention of motoneuron cell death by the I2-imidazoline receptor ligand LSL 60101. Br J Pharmacol 2000; 130: 1767-1776.
Casini A, Ceni E, Salzano R, Biondi P, Parola M, GaIh" A, Foschi M, et al. Neutrophil- derived superoxide anion induces lip peroxidation and stimulates collagen synthesis in human hepatic stellate cells: Role of nitric oxide. Hepatology 1997;25:361-367. Chan D.W., S.C. Son, W. Block, R. Ye, K.K. Khanna, M.S. Wold, P. Douglas, A.A. Goodazri, J. Pelley, Y. Taya, M.F. Lavin, S. P. Lees-Miller, Purification and characterization of human placenta: a manganese-dependent, wortmannin-sensitive serine/threonine protein kinase, J. Biol Chem. 275 (2000) 7803-7810.
Chavez E, Reyes-Gordillo K, Segovia J, Shibayama M, Tsutsumi V, Vergara P, Moreno M G, Muriel P. Resveratrol prevents fibrosis, NF-κB activation and TGF-β increases induced by chronic CCl4 treatment in rats. J. Appl. Toxicol. 2007; in press.
Chen A, Zhang L, Xu J, Tang J. The antioxidant (-)-epigallocatechin-3-gallate inhibits activated hepatic stellate cell growth and suppresses acetaldehyde-induced gene expression. Biochem J 2002;368(Pt 3):695-704.
Chianese R. A. and Cratree R. H. Organometallics, 2005, 24, 4432.
Das A, Banik N L, Ray S K. Garlic compounds generate reactive oxygen species leading to activation of stress kinases and cysteine proteases for apoptosis in human glioblastoma T98G and U87MG cells. Cancer. 2007; 10(5)1083-1095.
De Bleser P J, Xu G, Rombouts K, Rogiers V, Geerts A. Glutathione levels discriminate between oxidative stress and transforming growth factor-β signaling in activated rat hepatic stellate cells. J Biol Chem. 1999:274(8)3381-3387.
Deuffϊc S, Poynard T, Buffat L, Valleron AJ. Trends in primary liver cancer. Lancet 1998; 351:214-215.
Dominianni S J, Yen T Y. Oral Hypoglycemic Agents. Discovery and Structure-Activity Relationships of Phenacylimidazolium Halides. J. Med. Chem. 1989; 32:2301-2306.
Droge W. Free radicals in the physiological control of cell function. Physiol Rev 2002;82:47-95.
El-Serag HB, Davila JA, Petersern NJ, McGlynn KA. The continuing increase in the incidence of hepatocellular carcinoma in the United States: an update. Ann Intern Med 2003; 139:817-823.
El-Serag HB, Mason AC. Rising incidence of hepatocellular carcinoma in the United States. N Engl J Med 1999; 340:745-750.
Elsharkawy A M, Mann D A. Nuclear factor-kappaB and the hepatic inflammation-fibrosis- cancer axis. Hepatology 2007;46(2):590-597.
Fei P, and El-Diery,W.S. P53 and radiation responses, Oncogene 22(2003)5774-5783. Floyd R A. Nitrones as therapeutics in age-related diseases. Aging cell 2006;5(l):51-57.
Frei B, Higdon J V. Antioxidant activity of tea polyphenols in vivo: Evidence from animal studies. J Nutr 2003;133:3275S-3284S.
Friedler, A; Hansson, LO; Veprintsev, DB; Freund, SM; Rippin, TM; Nikolova, PV; Proctor, MR; Rudiger, S; Fersht, AR. A peptide that binds and stabilizes p53 core domain: Chaperone strategy for rescue of oncogenic mutants. Proc Natl Acad Sci U S A. 2002; 99:937-942. Friedman SL (2008) Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver. Physiol Rev 88(1): 125-172.
Friedman SL (2008b) Mechanisms of hepatic fibrogenesis. Gastroenterology 134(6): 1655- 1669.
Friedman S L. Liver fibrosis - from bench to bedside. J Hepatol 2003; 38(Suppl l):S38-53.
Fu Y, Zhou Y, Zheng S, Chen A. The antifibrogernc effect of (-)-epigallocatechin gallate results from the induction of de novo synthesis of glutathione in passaged rat hepatic stellate cells. Lab Invest 2006;86:697-709.
Galli A, Svegliati-Baroni G, Ceni E, Milani S, Ridolfi F, Salzano R, Tarocchi M, Grappone C, Pellegrini G, Benedetti A, Surrenti G, Casini A, Oxidative stress stimulates proliferation and invasiveness of hepatic stellate cells via a MMP2-mediated mechanism. Hepatology 2005; 41(5): 1074-1084.
Giaccia AJ. and Kastan M.B., The complexity of p53 modulation: emerging patterns from divergent signals, Genes Dev. 12 (1998), pp.2973-2983.
Harrison S A, Torgerson S, Hayashi P, Ward J, Schenker S. Vitamin E and vitamin C treatment improves fibrosis in patients with non-alcoholic steatohepatitis. Am J Gastroenterol 2003;98:2485-2490.
Halliwell B, Whiteman M. Measuring reactive species and oxidative damage in vivo and in cell culture: How should you do it and what do the results mean? Br J Pharmacol 2004; 142:231- 255.
Harlow K J, Hill A F, Welton T, Convenient and general synthesis of symmetric N5N'- disubstituted imidazolium halides, Synthesis, 1996;6:697-698.
Hofseth LJ. , S. Saito, S.P. Hussain, M.G. Espey, K.M. Miranda, Y. AraM, C. Jhappan, Y. Higashimotto, RHe. S.P. Linke, M.M. Quezado, I. Zurer, V. Rotter, D.A. Wink, E. Appella, CC. Harris, Nitric oxide- induces cellular stress and p53 activation in chronic inflammation Proc. Natl. Acad. Sci. USA 100(2003) 143-148.
Hollstein, M; Sidransky, D; Vogelstein, B; Harris, CC. p53 mutations in human cancers. Science. 1991; 253:49-53.
Hou Z, Sang S, You H, Lee M J, Hong J, Chin K V, Yang C S. Mechanism of action of (-)- eρigallocatechin-3-gallate: Auto-oxidation-dependent inactivation of epidermal growth factor receptor and direct effects on growth inhibition in human esophageal cancer KYSE 150 cells. Cancer Res 2005;65(17):8049-8056.
Huang A, Liu G. The study of innate drug resistance of human hepatocellular carcinoma Bel7402 cell line. Cancer left. 1999; 135:97-105.
Hupp, TR; Sparks, A; Lane, DP. Small peptides activate the latent sequence-specific DNA binding function of p53. CeU. 1995; 83:237-45. doi: 10.1016/0092-8674(95)90165-5. Hussain, SP; Hofseth, LJ; Harris, CC. Radical causes of cancer. Nat Rev Cancer. 2003; 3:276-285.
Iredale JP (2007) Models of liver fibrosis: exploring the dynamic nature of inflammation and repair in a solid organ. J Clin Invest 117(3): 539-548.
Iredale JP (2003) Cirrhosis: new research provides a basis for rational and targeted treatments. Bmj 327(7407): 143-147.
Jakubikova J, Sedlak J. Garlic-derived organosufides induce cytotocity, apoptosis, cell cycle arrest and oxidative stress in human colon carcinoma cell line. Neoplasma. 2006; 53(3): 191-199.
Kato A, MiyazaM M, Ambiru S, Yoshitomi H, Ito H, Nakagawa K, Shimizu H, Yokosuka O, Nakajima N. Multidrug resistance gene (MDR-I) expression as a useful prognostic factor in patients with human hepatocellular carcinoma after surgical resection. J Surg Oncol. 2001; 78(2): 110-115.
Katzenellenbogen M, Nizrahi L, Pappo R, Klopstock N, 01am D, Barash H, Domany E, Galun E, Glodenberg D. Molecular mechanisms of the chemopreventive effect on hepatocellular carcinoma development in Mdr2 knockout mice. MoI Cancer Ther. 2007; 6(4): 1283-1291.
Kawada N, Seki S, Inoue M, Kuroki T. Effect of antioxidants, resveratrol, wuercetin, and N- acetylcysteine, on the functions of cultured rat hepatic stellate cells and Kupffer cells. Hepatology 1998;27: 1265-1274.
Keum Y S, Chang P P J, Kwon J H, Yuan X L, Li W, Hu L, Kong A N T. 3- Morpholinoproply isothiocyanate (3MP-ITC) is a novel synthetic isothiocyanate that strongly induces the antioxidant response element (ARE)-dependent Nrf2-rnediated detoxifying/antioxidant enzymes in vitro and in vivo. Carcinogenesis 2007; in press.
Kluck RM, Bossy-Wetzel E, Green DR, Newmeyer DD The release of cytochrome c from mitochondria: A primary site for Bcl-2 regulation of apoptosis. Science 275 (1997):1132-1136.
Ko, LJ, Prives, C. p53: puzzle and paradigm. Genes & Dev. 1996; 10:1054-72.
Lin L C, Hung L C, Tsai T H. Determination of (-)-epigallocatechin gallate in rat blood by microdialysis coupled with liquid chromatography. J Chromatogr 2004;1032(l-2):125-128.
Liu S L, Degli Esposti S, Yao T, Diehl A M, Zern M A. Vitamin E therapy of acute CCl4- induced hepatic injury in mice is associated with inhibition of nuclear factor-kappaB binding. Hepatology 1995;22:1474-1481.
Llovet JM, Burroughs A, Bruix J. Hepatocellular carcinoma. Lancet. 2003; 362:1907-17.
Lorenzo H K, Susin S A. Therapeutic potential of AIF-mediated caspase-independent programmed cell dealth. Drug Resist Updat. 2007; 10(6):235-255
Lu G, Shimizu I, Cui X, Itonaga M, Tamaki K, Fukuno H, Inoue H, Honda H, Ito S. Interferon-alpha enhances biological defense activities against oxidative stress in cultured rat hepatocytes and hepatic stellate cells. J Med Invest 2002;49(3-4):172-181. Maples K R, Green A R, Floyd R A. Nitrone-related therapeutics: Potential of NXY-059 for the treatment of acute ischaemic stroke. CNS Drugs 2004;18(15):1071-1084.
Maubach G, Lim M C C, Zhang C Y, Zhuo L. GFAP promoter directs lacZ expression specifically in a rat hepatic stellate cell line. World J of Gastroentero 2006; 12:723-730.
Meurer S K, Lahme B, Tihaa L, WeisMrchen R, Gressner A M. N-acetyl-L-cysteine suppresses TGF-β signaling at distinct molecular steps: The biochemical and biological efficacy of a multifunctional, antifibrotic drug. Biochem Pharmaco 2005;70: 1026-1034.
Mignotte B, Vayssiere J-L. Mitochondria and apoptosis. Eur. J. Biochem. 1998; 252:1-15.
Moon W Sung, TarnawsM A. Nuclear Translocation of survivin in hepatocellular carcinoma: a key to cancer cell growth. Hum Pathol. 2003; 34(11):1119-1126.
Nakanuta M, Higashi N, Kohjima M, Fukushima M, Ohta S,- Kotoh K, Kobayashi N, Enjoji M. Epigallocatechin-3-gallate, a polyphenol component of green tea, suppresses both collagen production and collagenase activity in hepatic stellate cells. Int J MoI Med 2005;16:677-681.
Naugler W E, Sakurai T, Kim S, Maeda S, Kin K H, Elsharkawy A M, Karin M. Differences in MyD88-dependent IL-6 production. Science 2007;317:121-124.
Nishikawa T, Nakajima T, Moriguchi M, Jo M, Sekoguchi S, Ishϋ M, Takashima H, Katagishi T, Kimura H, Minami M, Itoh Y, Kagawa K, Okanoue T. A green tea polyphenol, epigalocatechin-3-gallate, induces apoptosis of human hepatocellular carcinoma, possible through inhibition of Bcl-2 family protein. J. Hepatol. 2006; 44:1074-1082.
Olmos G, DeGregorio-Rocasolano N, Regalado M P, Gasull T, Boronat M A, Trullas R, Villarroel A, Lerma J, Garcia-Sevilla J A. Protection by imidazol(ine) drags and agmatine of glutamate-induced neurotoxicity in cultured cerebellar granule cells through blockade of NMDA receptor. Br J Pharmacol 1999;127:1317-1326.
Orr J G, Leel V, Cameron G A, Marek G J, Haughton E L, Elrick L J, Trim J E, Hawksworth G M, Halestrap A P, Wright M C. Mechanism of action of the antiiϊbrogenic compound gliotoxin in rat liver cells. Hepatology 2004;40(l):232-242.
Oster, BA; Coffey, HA; Morin, MJ; Rastinejad, F. Pharmacological rescue of mutant p53 conformation and function. Science. 1999; 286:2507-10.
Padillo F J, Cruz A, Navarrete C, Bujalance I, Briceno J, Gallardo J I, Marchal T, Caballero R, Tύnez I, Muntane J, Montilla P, Pera-Madrazo C. Melatonin prevents oxidative stress and hepatocyte cell death induced by experimental cholestasis. Free Radic Res 2004;38(7):697-704.
Parkin DM, Bray F, Ferlay J, Pisani P. Estimating the world cancer burden: Globocan 2000. Int J Cancer 2001; 94:153-156.
Parola M, Pinzani M, Casini A, Leonarduzzi G, Marra F, Caligiuri A, Ceni E, et al. Induction of procollagen type I gene expression and synthesis in human hepatic stellate cells by 4- hydroxy-2,3-nonenal and other 4-hydroxy-2,3-alkenals is related to their ' molecular structure. Biochem Biophys Res.Commun 1996;222:261-264. Peng J, Stevenson EF, Doctrow SR, Andersen JK. Superoxide dismutase/catalase mimetics are neuroprotective against selective paraquat-mediated dopaminergic neuron death in the substantial nigra: Implications for Parkinson disease. J Biol Chem. 2005;280:29194-29198.
Pilch D.R., O.A. Sedelnikova, C. Redon, A. Celeste, A. Nussenzweig, W.M. Bonner, characteristics of gamma-H2AX foci at DNA double stranded breaks sites, Biochem. Cell. Biol. 81 (2003) 123-129.
Rakoff-Nahoum S, Why Canacer and Inflammation, Yale Journal of Biology and Medicine (2006); 79; 123-130.
Saito S., H. Yamaguchi, Y, Hi'gashimoto, C. Chao, Y. Xu, AJ. Forance Jr. E. Apella, CW. Anderson, phosphorylation site interdependence of human p53 post- translational modifications in response to stress, J. Biol. Chem. 278(2003) 37536-37544)
Saito S., A.A. Goodazri, Y.Higashimoto, Y. Noda, S.p. Lees-miller, E. Appella, CW. Anderson, ATM mediates phosphorylation at multiple p53 sites, including ser(46), in response to ionizing radiation, J. Biol. Chem. 277 (2002) 12491-12494.
Selivanova, G; Iotsova, V; Okan, I; Fritsche, M; Strom, M; Groner, B; Grafstrom, RC; Wiman, KG. Restoration of the growth suppression function of mutant p53 by a synthetic peptide derived from the p53 C-terminal domain. Nat Med. 1997; 3:632-8. doi: 10.1038/nm0697-632.
Shieh S. Y., Eceda M., Taya Y. and Prives C, DNA damage-induced phosphorylation of p53 alleviates inhibition by MDM2, Cell 91 (1997), pp. 325-334.
Siliciano J.D. , CE. Canman, Y. Taya, K. A Sakaguchi, E. Appella, M.B. Kastan, DNA damage induces phosporylation of aminoterminus of p53, Genes Dev. 11 (1997) 3471-3481.
. Smart DE, Vincent KJ, Arthur MJP, et al. Control of the tissue inhibitor of metalloproteinases-1 and interleukin-6 genes in activated hepatic stellate cells. J Biol Chem. 2001;276:244l4-24421.
Smith G.C., R.B. Cary, N.D. LaMn, B.C. Hann, S. H. Teo, DJ. Chen, S. P. Jackson, Purification, of DNA binding properties of ataxia- telangiectasia gene product ATM, Proc. Natl. Acad, Sci. USA96 (1999) 11134-11139.
Taylor-Robinson SD, Foster GR, Arora S, Hargreaves S, Thomas HC Increase in primary 'liver cancer in the UEC, 1979-1994. Lancet 1997; 350: 1142-1143.'
Thomas M B. Hepatocellular Carcinoma: The Need for Progress. J. Clin. Oncol. 2005; 23(13): 2892-2899
Thorgeirsson S S, Grisham J W. Molecular phthogenesis of human heptocelluϊar carcinoma. Nature Genetics. 2002; 31: 339-346
Tonini G, Vinvenzi B, Santhii D, Scarpa S, Vasaturo f, Malacrino C, Coppola R, magistrelli P, Borzomati D, Baldi A, Antinori A, Caricato M, Nuzzo G, Picciocchi A. Nuclear and cytoplasmic expression of suvivin in 67 sugically resected pancreatic cancer patients. Br J Cancer. 2005; 92:2225-2232. Tsukamoto H, Rippe R, Niemela O, Lin M. Roles of oxidative stress in activation of Kupffer and Ito cells. J Gastroenterol Hepatol 1995;10:s50-s53.
Tsukamoto H. Cytokine regulation of hepatic stellate cell in liver fibrosis. Alcohol Clin Exp Res 1999;23:911-916.
VaIi L, Hahn O, Kupcsulik P, Drahos A, Sarvary E, Szentmihalyi K, Pallai Z, Kurucz T, Sipos P, Blazovics A. Oxidative stress with altered element content and decreased ATP level of erythrocytes in hepatocellular carcinoma and colorectal liver metastases. Eur J Gastroenterol Hepatol. 2008; 20(5); 393-398.
Valko M, Izakovic M, Mazur M, Rhodes C, Tesler J Role of oxygen radical in DNA damage and cancer incidence. MoI. Cell. Biochem. 2004; 266; 37-56.
Vasiliou V, Lee J, Pappa A, Petersen DR. Involvement of p65 in the regulation of NF-kB in rat hepatic stellate cells during cirrhosis. Biochem Biophys Res Commun 2000;273:546-550.
Vogelstein, B; Lane, D; Levine, AJ. Surfing the p53 network [news] [In Process Citation]. Nature. 2000; 408:307-10. doi: 10.1038/35042675.
Wagle D R, Vasan S, Gall M. Method of treating fibrotic diseases or other indications with imidazolium agents. Patent Application US 2007/0043016 Al. 2007.
Wahl G.M. and A.M. Carr, The evolution of diverse biological responses, to DNA damage: insights from yeast and p53, Nat. Cell Biol. 3(2001) E277-286.
Wei Michael C, Wei-Xing Zong, Emily H.-Y. Cheng, Tullia Lindsten, ViIy Panoutsakopoulou, Andrea J. Ross, Kevin A. Roth, Grant R. MacGregor, Craig B. Thompson, Stanley J. Korsmeyer. Proapoptotic BAX and BAK: A requisite gateway to mitochondrial dysfunction and death. Science 292 (2001):727-730.
Weiskirchen R, Gressner A M. Isolation and culture of hepatic stellate cells. Methods MoI Med 2005;117:99-113.
Whalen R, Rockey DC, Friedman SL, Boyer TD. Activation of rat hepatic stellate cells leads to loss of glutathione S-transferases and their enzymatic activity against products of oxidative stress. Hepatology 1999;30(4):927-933.
Zhang Y, Ngeow K. C. and Yiαg J. Y. Organic Letters, 9 [18] (2007) 3495-3498.
Zhang CY, Zhuo L. Epigallocatechin gallate and genistein attenuate glial fibrillary acidic protein elevation induced by fibrogenic cytokines in hepatic stellate cells. Int J MoI Med 2006;18:1141-1151.
Zhao L, Zhang C, Zhuo L, Zhang Y, Ying, Y Y J. Imidazolium Salts: A Mild Reducing and Anti-oxidative Reagent JACS. 2008; 130(38):12586-7.

Claims

WHAT IS CLAIMED IS:
1. A method for delivering an anti-fibrotic or anti-cancer agent to a cell, the method comprising contacting the cell with an effective amount of a compound of general formula I or an oligomer or polymer thereof: •
Figure imgf000080_0001
wherein:
the dashed line is absent or is present as a bond to form a second bond between the carbon to which R1 and R3 are attached and the carbon to which R2 and R4 are attached;
R1 and R2: (i) are each independently H, straight or branched C1-C6 alkyl, straight or branched C2-
Ce alkenyl, straight or branched C2-C6 alkynyl, C6-C1O aryl;
(ϋ) together with their ring atoms form a 6- to 10-membered fused saturated, unsaturated or aromatic ring system; (ϋi) R1 and R5 together with their ring atoms, or R2 and R6 together with their ring atoms, form a 5- to 10-membered fused saturated, unsaturated or aromatic ring system and the other of R1 and R2 is as defined above in (i); or
(iv) R1 and R5 together with their ring atoms and R2 and R6 together with their ring atoms each form a 5- to 10-membered fused saturated, unsaturated or aromatic ring system;
R and R are both H, or, when R and R together with, their ring atoms form a 6- to 10-membered ■ fused aromatic ring system or when the dashed line is present as a bond, R3 and R4 are absent;
R5 or R6: (i) are as defined above for R1 and R2; or (ϋ) are each independently straight or branched C1-Ce alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C18 cycloalkyl including fused cycloalkyl ring systems, C6-C10 aryl, C6-C10 aryl-Ci-C6 alkyl, C6-Ci0 aryl-C2-C6 alkenyl, or C6-C10 aryl-C2-C6 alkynyl, C1-C6 alkyl-C6-C10 aryl, C2-C6 alkenyl-C6-C10 aryl, or C2-C6 alkynyl-C6-C10 aryl;
R is H, C1-C6 alkyl, phenyl, substituted C1-C6 alkyl or halo;
in which any of R1 to R7, where applicable, optionally has one or more carbon atoms replaced with a heteroatom selected from N, O, S and P and is optionally substituted with one or more of straight or branched C1-Cg alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3- C18 cycloalkyl including fused cycloalkyl ring systems, C6-C10 aryl, fluoro, tri-fluoro-methyl, cyanato, isocyanato, carboxyl, C1-C6 acyloxy, C1-C6 acyl, carbonyl, amino, acetyl, acetoxy, oxo, nitro, hydroxyl, C1-C6 alkylcarboxy, Cj-C6 alkoxy, C2-C6 alkenoxy, C2-C6 alkynoxy; and
in which one of the ring carbon atom to which R1 and R5 are attached and the ring carbon to R2 and R6 are attached is optionally replaced with a nitrogen atom;
or a pharmaceutically acceptable salt thereof.
2. The method of claim 1 wherein the compound is an imidazolium or an imidazolium salt.
3. The method of claim 1 wherein the compound is an imidazohnium or an imidazoh'nium salt.
4. The method of claim 1 wherein R5 is the same as R6.
5. The method of claim 1 wherein R5 and R6 are hydrocarbons.
6. The method of claim 1 in wherein the pharmaceutically acceptable salt is a chloride, bromide, tetrafluoroborate or hexafluorophosphate salt.
7. The method of claim 1 wherein the compound is l-ethyl-3-methylimidazolium, 1,3- bisbenzylinύdazoliurα, 1,3-dϋsoproρyIimidazolium, 1,3-di-tert-butylimidazolinium, 1,3-Ms(I- adamantyl)imidazolium, l,3-bis(2,4,6-trimethylphenyl)-imidazoUnium, l,3-bis(2,6-diisopropyl- phenyl)-imidazolinium, 1,3-diaUylirrridazolium, l-benzyl-3-methyBmidazolium, l-butyl-3- methyHmidazolium, 1 -( 1 -adamanryl)-3-(2,4,6-1iimemylphenyl)-4,5-dmydroirnidazolium, 2- benzyUmidazo[l,5-σ]quinolinium, l,3-bis(l-adamantyl)-benzimidazolium, 1,3- dicyclohexylbenzimidazolium, 1,3-dϋsopropylimidazoKnium tetrafluoroborate, 1,3- diisopropylimidazolium, 2-(2,6-dϋsopropylphenyl)-5-methyHmidazo[l,5-α]pyridinium, l-(2,6- diisopropylphenyl)-3-(2,4,6-trimemylphenyl)^ αjpyridinium, 2-mesityl-2,5,6J-tefr^ 1,3-Ms(I- adamantyl)imidazolinium, l-butyl-3-(2-pyridiny]jne1tyl)-lH-imidazolium, 6,7-dihydro-2- pentafluoroplmeyl-5H-pyrrolo[24-c3-l,2,4-trizoli-um, l-memyl-3-(2-hydroxylemyl)-iinidazoUuni, l-methyl-3-(4-isocynatobenzyl)-imidazolium, l-methyl-3-(4-carboxylbenzyl)-imidazolium 1- methyl-3-(4-acetate-ben2yl)-imidazolium, 1 -methyl-3-(252-dime1hoxylethyl)-imidazolium5 1 -(2,4,6- trimethylphenyl)-3 -(4-acetate-benzyl)-imidazolium, 1 ,3 -Dibenzyl-5-phenylimidazolium, 1 -benzyl- 3-(4-carboxylbenzyl)-2-methyliinidazolium, l-benzyl-3-(3,4,5-trimemoxylbenzyl)-2- methylimidazoliυm, l-benzyl-3-(4-acetate-beiizyl)-2-metb.yl-imidazoliuni, l-benzyl-3-(4- methylcarboxylateben2yl)-2-meth.yl-itnidazoliuni, l-benzyl-3-(2,2-dimethoxyleth.yl)-2-methyl- imidazoUum, 2,6-di-(3-benzyl-iinidazoHuin)-pyridine, 2,2'-di-(3-ben2yl-itnidazolium)-l5r- binaphthalene, l-ben2yl-3-(4-metibιylbe]izyl)-irmdazoKum, l-benzyl-3-(2-trifluorometh.ylbenzyl)-2- methylimidazolium, 1 -benzyl-3 -(4-me1iιylcarboxylatebenzyl)-5-plienyl-imidazolium, 1 -benzyl-3 - (4-acetateben2yl)-5-phenyl-imidazolium5 1 -benzyl-3-(4-methylbenzyl)-5-phenylimidazolium, (1 ,2- 4,5-di'irm'dazolium)-N,N' ,N",N' "-tetrabenzyl-benzene, 1 -benzyl-3 -(2-propyn- 1 -yl)-imidazolium, 1 - benzyl-3-(3-hydroxyl-propyl)-imidazolium, l,3-di(2-phenylethyl)-imidazolium, l-ben2yl-3-(4- acetateben2yl)-imidazolium, l-benzyl-S-φyridin^-y^-imidazolium, l53,5-tris-(4-methyl- imiazolium)-linked cyclophane, l,3-diben2yl-2-(l,3-dibenzyl-lH-imidazol-2(3H)-ylidene)-2,3- dihydro-lH-imidazole, 1 -benzyl-3 -methyl-imidazoliiim, l-(4-cyanatobenzyl)-3-methyl- imidazolium, l-(4-carboxybenzyl)-3-metb.yl-imidazolium, l-meth.yl-3-(4-acetate-ben2yl)- imidazolium, l-methyl-3-(252-dimetlioxyethyl)-inαidazolium, or l-(2,4,6-trimeth.ylplienyl)-3-(4- acetate-benzyl)-imidazolium, or a pharmaceutically acceptable salt thereof.
8. The method of claim 1 wherein the compound is a dimer of a compound having a structure of general formula I.
9. The method of claim 8 wherein the compound is l53-dibenzyl-2-(l,3-dibenzyl-lH- imidazol-2(3H)-yMene)-253-dmydro-m-imidazole, (l52-455-diimidazoUum)-N5N',N'', N'''- tetrabenzyl-benzene, 256-di-(3-benzyl-imidazolium)-pyridine or 2,2'-di-(3-benzyl-imidazolium)- ljl'-binaphthalene, or a pharmaceutically acceptable salt thereof.
10. The method of claim 1 in which the compound is a trimer of a compound having a structure of general formula I.
11. The method of claim 10 wherein the compound is l,3,5-tris(4-memyl-imidazolium)-Knked cyclophane or a pharmaceutically acceptable salt thereof.
12. The method of claim 1 wherein the cell is in vitro.
13. The method of claim 1 wherein the cell is in vivo.
14. The method of claim 13 wherein the method comprises administering the agent to a subject for the treatment of fibrotic disease.
15. The method of claim 14 wherein the fibrotic disease is hepatic fibrosis.
16. The method of claim 15 wherein the compound is 1,3-dϋsopropylimidazoliιιm or a pharmaceutically acceptable salt thereof.
17. The method of claim 13 wherein the method comprises administering the agent to a subject for the treatment of cancer.
18. The method of claim 17 wherein the cancer is hepatocellular carcinoma, lung cancer, breast cancer, stomach cancer or glioma.
19. The method of claim 18 wherein the compound 1,3-Bisbenzylimidazolium, 1, 3,-Dibenzyl- 2-memylimidazoHum,l-(2,4,6-ttimemylphenyl)-3-(4-acetate-benzyl)-imidazolium, l,3-Dibenzyl-5- phenylimidazoliurα , l-benzyl-3-(4-acetate-benzyl)-2-methyl-imidazolium, l-benzyl-3-(4- methylbenzyl)-imidazoiium, l-benzyl-3-(2-trifiuoromethylbenzyl)-2-methylimidazolium or 1,3- di(2-phenylethyl)-imidazolium, or a pharmaceutically acceptable salt thereof.
20. Use of a compound having a structure of general formula I, or an oligomer or polymer thereof, for delivering an anti-fibrotic or anti-cancer agent to a cell in vivo:
Figure imgf000083_0001
wherein:
the dashed line is absent or is present as- a bond to form a second bond between the carbon to which R1 and R3 are attached and the carbon to which R2 and R4 are attached;
R1 and R2: (i) are each independently H, straight or branched C1-C6 alkyl, straight or branched C2-
C6 alkenyl, straight or branched C2-C6 alkynyl, C6-Ci0 aryl; (ii) together with their ring atoms form a 6- to 10-membered fused saturated, unsaturated or aromatic ring system; (iii) R1 and R5 together with their ring atoms, or R2 and R6 together with their ring atoms, form a 5- to 10-membered fused saturated, unsaturated or aromatic ring system and the other of R1 and R2 is as defined above in (i); or (iv) R1 and R5 together with their ring atoms and R2 and R6 together with their ring atoms each form a 5- to 10-membered fused saturated, unsaturated or aromatic ring system;
R3 and R4 are both H, or, when R1 and R2 together with their ring atoms form a 6- to 10-membered fused aromatic ring system or when the dashed line is present as a bond, R3 and R4 are absent;
R5 or R6:
(i) are as defined above for R1 and R2; or
(ii) are each independently straight or branched Ci-C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C18 cycloalkyl including fused cycloalkyl ring systems, C6-Ci0 aryl, C6-Ci0 aryl-Ci-C6 alkyl, C6-CiO 8TyI-C2-C6 alkenyl, or C6-Ci0 aryl-C2-C6 alkynyl, Ci-C6 alkyl-C6-Cio aryl, C2-C6 alkenyl-C6-Ci0 aryl, or C2-C6 alkynyl-C6-Ci0 aryl;
R7 is H, Ci-C6 alkyl, phenyl, substituted Ci-C6 alkyl or halo;
in which any of R1 to R7, where applicable, optionally has one or more carbon atoms replaced with a heteroatom selected from N, O, S and P and is optionally substituted with one or more of straight or branched Ci-Cg alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-Cg alkynyl, C3- Ci8 cycloalkyl including fused cycloalkyl ring systems, C6-Ci0 aryl, fluoro, tri-fluoro-methyl, cyanato, isocyanato, carboxyl, Ci-C6 acyloxy, Ci-C6 acyl, carbonyl, amino, acetyl, acetoxy, oxo, nitro, hydroxyl, Ci-C6 alkylcarboxy, Ci-C6 alkoxy, C2-C6 alkenoxy, C2-C6 alkynoxy; and
in which one of the ring carbon atom to which R1 and R5 are attached and the ring carbon to R2 and R6 are attached is optionally replaced with a nitrogen atom;
or a pharmaceutically acceptable salt thereof.
21. Use of a compound having a structure of general formula I, or an oligomer or polymer thereof, in the manufacture of a medicament for delivering an anti-fibrotic or anti-cancer agent to a cell in vivo:
Figure imgf000085_0001
wherein:
the dashed line is absent or is present as a bond to form a second bond between the carbon to which R and R3 are attached and the carbon to which R2 and R4 are attached;
R1 and R2:
(i) are each independently H, straight or branched Ci-C6 alkyl, straight or branched C2-
C6 alkenyl, straight or branched C2-C& aϊkynyl, C6-Ci0 aryl; (ϋ) together with their ring atoms form a 6- to 10-membered fused saturated, unsaturated or aromatic ring system; (iii) R1 and R5 together with their ring atoms, or R2 and R6 together with their ring atoms, form a 5- to 10-membered fused saturated, unsaturated or aromatic ring system and the other of R1 and R2 is as defined above in (i); or (iv) R1 and R5 together with their ring atoms and R2 and R6 together with their ring atoms each form a 5- to 10-membered fused saturated, unsaturated or aromatic ring system;
R3 and R4 are both H, or, when R1 and R2 together with their ring atoms form a 6- to 10-membered fused aromatic ring system or when the dashed line is present as a bond, R3 and R4 are absent;
R5 Or R6:
(i) are as defined above for R1 and R2; or
(ii) are each independently straight or branched Ci-C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3-C1S cycloalkyl including fused cycloalkyl ring systems, C6-CiO aryl, C6-C10 aryl-Ci-C6 alkyl, C6-Ci0 aryl-C2-C6 alkenyl, or C6-Ci0 aryl-C2-C6 alkynyl, Ci-C6 alkyl-C6-Ci0 aryl, C2-C6 alkenyl-C6-Ci0 aryl, or C2-C6 alkynyl-C6-Ci0 aryl; R is H, C1-C6 alkyl, phenyl, substituted C1-C6 alkyl or halo;
in which any of R1 to R7, where applicable, optionally has one or more carbon atoms replaced with a heteroatom selected from N, O, S and P and is optionally substituted with one or more of straight or branched C1-C6 alkyl, straight or branched C2-C6 alkenyl, straight or branched C2-C6 alkynyl, C3- Ci8 cycloalkyl including fused cycloalkyl ring systems, C6-C10 aryl, fluoro, tri-fluoro-methyl, cyanato, isocyanato, carboxyl, Ci-C6 acyloxy, Ci-C6 acyl, carbonyl, amino, acetyl, acetoxy, oxo, nitro, hydroxyl, Ci-C6 alkylcarboxy, Ci-C6 alkoxy, C2-C6 alkenoxy, C2-C6 alkynoxy; and
in which one of the ring carbon atom to which R1 and R5 are attached and the ring carbon to R2 and Rδ are attached is optionally replaced with a nitrogen atom;
or a pharmaceutically acceptable salt thereof.
22. The use of claim 20 or claim 21 wherein the compound is an imidazolium or an imidazolium salt.
23. The use of claim 20 or claim 21 wherein the compound is an imidazolinium or an imidazolinium salt.
24. The use of any one of claims 20 to 23 wherein R5 is the same as R6.
25. The use of any one of claims 20 to 23 wherein R5 and R6 are hydrocarbons.
26. The use of any one of claims 20 to 23 wherein the pharmaceutically acceptable salt is a chloride, bromide, tetrafluoroborate or hexafluorophosphate salt.
27. The use of claim 20 or claim 21 wherein the compound is l-ethyl-3-methylimidazolium, 1,3-bisbenzyljbmdazolium, 1,3-diisopropylimidazolium, l,3-di-tert-buty]imidazoMnium, l,3-bis(l- adamantyl)imidazolium, l,3-bis(2,4,6-trimethylphenyl)-irnidazoliniurn, l,3-bis(2,6-diisopropyl- phenyl)-imidazolinium, 1,3-diallylimidazolium, l-benzyl-3-methylimidazolium, l-butyl-3- methylirnidazolium, l-(l-adamantyl)-3-(2,4,6-trimethylphenyl)-4)5-dihydroimidazolium, 2- benzylimidazo[l,5-α]quinblinium, l,3-bis(l-adamantyl)-benzimidazolium, 1,3- dicyclohexylbenzimidazolium, 1,3-dϋsopropylimidazolinium tetrafluoroborate, 1,3- diisopropylimidazolium, 2-(2,6-diisopropylphenyl)-5-methyiimidazo[l,5-α]pyridinium, l-(2,6- ffisopropylphenyl)-3-(2,4>6-Mmemylphenyl)-iniidazolinium, 2-mesityl-5- methylimidazo[l ,5- α]pyridinium, 2-mesityl-2,5,6,7-tetrahydropyrrolo[2,l-c][l,2,4]triazθl-4-ium, l,3-bis(l- adamantyl)imidazolinium,' l-butyl-3-(2-pyridinylmethyl)- IH-imidazolium, 6,7-dihydro-2- pentafluoroρhneyl-5H-pyrrolo[2,l-c]-l,2,4-trizolium, l-methyl-3-(2-hydroxylethyl)-imidazolium, l-methyl-3-(4-isocynatobenzyl)-imidazolium, l-methyl-3-(4-carboxylbenzyl)-imidazolium, 1- methyl-3-(4-acetate-benzyl)-imidazolium, 1 -methyl-3-(2,2-diDiethoxylethyl)-imidazolium, l-(2,4,6- trimethylplienyl)-3-(4-acetate-benzyl)-inxidazoliuin, l,3-Dibenzyl-5-phenylimidazolium, 1-benzyl- 3-(4-carboxylbenzyl)-2-methylimidazolium, l-benzyl-3-(3,4,5-trimethoxylbenzyl)-2- methylimidazolium, l-benzyl-3-(4-acetate-benzyl)-2-methyl-imidazoliuni, l-benzyl-3-(4- methylcarboxylatebenzyl)-2-methyl-imidazolium, l-benzyl-3-(2,2-dimethoxylethyl)-2-methyl- imidazolium, 2,6-di-(3-benzyl-imidazolium)-pyridine, 2,2'-di-(3-benzyl-imidazolium)-l,r- binaphthalene, l-benzyl-3-(4-methylbenzyl)-imidazolium, l-benzyl-3-(2-triiluoromethylbenzyl)-2- memylrmidazolium, l-benzyl-3-(4-metb.ylcarboxylatebenzyl)-5-phenyl-irαidazolium, l-benzyl-3- (4-acetatebenzyl)-5-phenyl-inήdazolium, l-benzyl-3-(4-methylbenzyl)-5-phenylimidazolium, (1 ,2- 4,5-diimidazolium)-N,N' ,N",N'"-tetrabenzyl-benzene, l-benzyl-3-(2-propyn-l-yl)-imidazolium, 1- benzyl-3-(3-hydroxyl-propyl)-imidazolium, l,3-di(2-phenylethyl)-imidazolium, l-benzyl-3-(4- acetatebenzyl)-imidazolium, 1 -benzyl-3 -(pyridin-2-yl)-imidazolium, 1 ,3 ,5-tris-(4-methyl- imiazolium)-linked cyclophane, l,3-dibenzyl-2-(l}3-dibenzyl-lH-imidazol-2(3H)-ylidene)-2,3- dihydro-lH-imidazole, l-benzyl-3-methyl-imidazolium, l-(4-cyanatoben2yl)-3-metiιyl- imidazolium, l-(4-carboxybenzyl)-3-metb.yl-imidazolium5 l-metb.yl-3-(4-acetate-benzyl)- imidazolium, l-melliyl-3-(2,2-dimethoxyethyl)-imidazoliuin, or l-(2,4,6-trimeth.ylphenyl)-3-(4- acetate-benzyl)-imidazolium, or a pharmaceutically acceptable salt thereof.
28. The use of claim 20 or claim 21 wherein the compound is a dimer of a compound having a structure of general formula I.
29. The use of claim 28 wherein the compound is l,3-dibenzyl-2-(l,3-dibenzyl-lH-imidazol- 2(3H)-yUdene)-2,3-dihydro-lH-imidazole, (l,2-4,5-diimidazoKum)-N,N' ,N" , N" ' -tetrabenzyl- benzene, 2,6-di-(3-benzyl-imidazolium)-pyridine or 2,2'-di-(3-benzyl-imidazolium)-l,r- binaphthalene, or a pharmaceutically acceptable salt thereof.
30. The use of claim 20 or claim 21 in which the compound is a trimer of a compound having a structure of general formula I.
31. The use of claim 30 wherein the compound is l,3,5-tris(4-methyl-irnidazolium)-linked cyclophane or a pharmaceutically acceptable salt thereof.
32. The use any one of claims 20 to 31 wherein the anti-fibrotic agent is delivered for the treatment of fibrotic disease.
33. The use of claim 32 wherein the fibrotic disease is hepatic fibrosis.
34. The use of claim 33 wherein the compound is 1,3-diisopropylimidazolium or a pharmaceutically acceptable salt thereof.
35. The use of any one of claims 20 to 31 wherein the anti-cancer agent is delivered for the treatment of cancer.
36. The use of claim 35 wherein the cancer is hepatocellular carcinoma, lung cancer, breast cancer, stomach cancer or glioma.
,
37. The use of claim 36 wherein the compound is 1,3-Bisbenzylήmdazolium, 1, 3,-Dibenzyl-2- methylimidazolium, l-(2,4,6-1ximethylphenyl)-3-(4-acetate-benzyl)-imidazolium, 1 ,3-Dibenzyl-5- phenylimidazolium , l-benzyl-3-(4-acetate-benzyl)-2-methyl-imidazolium, l-benzyl-3-(4- methylben2yl)-imidazolium, l-benzyl-3-(2-trifluoromethylbenzyl)-2-methylimidazolium or 1,3- di(2-phenylethyl)-imidazolium, or a pharmaceutically acceptable salt thereof.
38. A compound that is an imidazolium or imidazolinium that is l-ethyl-3-methylimidazolium, l-methyl-3-(2-hydroxylethyl)-imidazolium, l-methyl-3-(4-isocynatobenzyl)-imidazo]ium, 1- methyl-3-(4-carboxylbenzyl)-imidazolium, l-methyl-3-(4-acetate-benzyl)-imidazolium, 1-methyl- 3-(2,2-dimethoxylethyl)-imidazolium, l-(2,4,6-ttimethylphenyl)-3-(4-acetate-benzyl)-imidazolium, l,3-Dibenzyl-5-phenylimidazolium, l-benzyl-3-(4-carboxylbenzyl)-2-methylimidazolium, 1- benzyl-3-(3,4,5-trimethoxylbenzyl)-2-methylimidazolium, l-benzyl-3-(4-acetate-benzyl)-2-methyl- imidazolium, l-benzyl-3-(4-methylcarboxylatebenzyl)-2-methyl-imidazolium, l-benzyl-3-(2,2- dimethoxylethyl)-2-methyl-imidazolium, 2,6-di-(3-benzyl-imidazolium)-pyridine, 2,2' -di-(3- benzyl-imidazolium)-l,r-binaphthalene, l-benzyl-3-(4-methylbenzyl)-imidazolium, l-benzyl-3-(2- -trifluoromethylbenzyl)-2-methylimidazolium, l-benzyl-3-(4-methylcarboxylatebenzyl)-5-phenyl- imidazolium, l-benzyl-3-(4-acetatebenzyl)-5-phenyl-imidazolium, l-benzyl-3-(4-methylbenzyl)-5- phenylimidazolium, (l,2-4,5-diimidazolium)-N,N',]SP',N'"-tetrabenzyl-benzene, l-benzyl-3-(2- propyn-l-yl)-imidazolium, l-benzyl-3-(3-hydroxyl-propyl)-imidazolium, l,3-di(2-phenylethyl)- imidazolium, l-benzyl-3-(4-acetatebenzyl)-imidazolium, l-benzyl-3-(pyridin-2-yl)-imidazolium, 1 ,3 ,5-1xis-(4-metih.yl-irniazolium)-linked cyclophane, 1 ,3 -dibenzyl-2-(l ,3 -dibenzyl- lH-imidazol- 2(3H)-ylidene)-2,3-dihydro-lH-imidazole, l-benzyl-3-methyl-imidazolium, l-(4-cyanatobenzyl)-3- methyl-imidazolium, l-(4-carboxybenzyl)-3-methyl-imidazolium, l-methyl-3-(4-acetate-benzyl)- imidazolium, l-methyl-3-(252-dimethoxyethyl)-imidazolium, or l-(2,4,6-trimethylphenyl)-3-(4- acetate-benzyl)-imidazolium, or a pharmaceutically acceptable salt thereof.
PCT/SG2009/000037 2008-01-30 2009-01-30 Method for treating fibrosis and cancer with imidazolium and imidazolinium compounds WO2009096905A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP09706910A EP2249832A4 (en) 2008-01-30 2009-01-30 Method for treating fibrosis and cancer with imidazolium and imidazolinium compounds
US12/865,668 US9072729B2 (en) 2008-01-30 2009-01-30 Method for treating fibrosis and cancer with imidazolium and imidazolinium compounds
JP2010544928A JP5559701B2 (en) 2008-01-30 2009-01-30 Methods of treating fibrosis and cancer with imidazolium and imidazolinium compounds
CN2009801120807A CN101983057A (en) 2008-01-30 2009-01-30 Method for treating fibrosis and cancer with imidazolium and imidazolinium compounds
US14/789,928 US20160331727A9 (en) 2008-01-30 2015-07-01 Method for treating fibrosis and cancer with imidazolium and imidazolinium compounds

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US676908P 2008-01-30 2008-01-30
US61/006,769 2008-01-30

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/865,668 A-371-Of-International US9072729B2 (en) 2008-01-30 2009-01-30 Method for treating fibrosis and cancer with imidazolium and imidazolinium compounds
US14/789,928 Continuation US20160331727A9 (en) 2008-01-30 2015-07-01 Method for treating fibrosis and cancer with imidazolium and imidazolinium compounds

Publications (1)

Publication Number Publication Date
WO2009096905A1 true WO2009096905A1 (en) 2009-08-06

Family

ID=40913057

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/SG2009/000037 WO2009096905A1 (en) 2008-01-30 2009-01-30 Method for treating fibrosis and cancer with imidazolium and imidazolinium compounds

Country Status (5)

Country Link
US (2) US9072729B2 (en)
EP (1) EP2249832A4 (en)
JP (1) JP5559701B2 (en)
CN (1) CN101983057A (en)
WO (1) WO2009096905A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2271338A1 (en) * 2008-03-31 2011-01-12 Agency for Science, Technology And Research Method for treating neurological disorders with imidazolium and imidazolinium compounds
EP2701511A4 (en) * 2011-04-29 2015-06-10 Univ Akron Azolium and purinium salt anticancer and antimicrobial agents
US9855280B2 (en) 2015-11-06 2018-01-02 National Dong Hwa University Nanoparticles containing azolium and N-heterocyclic carbene compounds and use thereof
CN108777957A (en) * 2016-02-12 2018-11-09 新加坡科技研究局 Antibacterial patterned surface and its manufacturing method
WO2023281388A1 (en) * 2021-07-05 2023-01-12 Uniwersytet Medyczny W Białymstoku Imidazolium salt, which is a derivative of lithocholic acid, a method of its preparation and its use
US11584722B2 (en) 2017-04-07 2023-02-21 Xiamen Vivohealths Technology Co., Ltd. Substituted imidazole salt compounds, preparation method thereof, pharmaceutical composition thereof and application thereof

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102952079B (en) * 2012-11-27 2014-10-15 河北工业大学 Novel imidazolium salt ionic liquid and related luminescent material thereof
EP3386952A4 (en) * 2015-12-11 2019-08-28 The Board of Regents of The University of Texas System Substituted benzimidazolium, pyrido-imidazolium, or pyrazino-imidazolium compounds as chemotherapeutics
JP7224968B2 (en) * 2018-03-05 2023-02-20 東ソー株式会社 Method for producing aromatic compound
CN109232265B (en) * 2018-10-22 2021-09-28 苏州大学 Method for preparing benzyl amine compound
CN111909092A (en) * 2019-05-10 2020-11-10 厦门华绰生物医药科技有限公司 Fluorine-containing substituted imidazole salt compound, preparation method thereof, medicinal composition and application thereof
CN114835648A (en) * 2022-05-30 2022-08-02 平顶山学院 Bromobenzyl benzimidazole quaternary ammonium salt derivative and antibacterial application thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001077486A1 (en) * 2000-04-07 2001-10-18 Sofitech N.V. Scale removal
US20030176426A1 (en) * 2000-07-13 2003-09-18 Alteon, Inc. Method for treating fibrotic diseases or other indications with imidazolium agents
WO2005019183A1 (en) * 2003-07-21 2005-03-03 Basf Aktiengesellschaft Method for the production of purified 1,3-substituted imidazolium salts
US20050282803A1 (en) * 2004-06-17 2005-12-22 Haley Gregory J CIS-imidazolines

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3849557A (en) 1971-05-13 1974-11-19 Allen & Hanburys Ltd Pharmaceutical compositions containing 1,1-azo bis-(1h-imidazo(1,2-a)pyridinium)dibromide compounds and the use thereof
US4483835A (en) * 1983-11-14 1984-11-20 Chevron Research Company Process for preparing molecular sieves using imidazole template
US7041871B1 (en) 1995-10-10 2006-05-09 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US6194447B1 (en) 1998-07-02 2001-02-27 Neurosearch A/S Bis (benzimidazole) derivatives serving as potassium blocking agents
WO2000025793A1 (en) 1998-10-30 2000-05-11 Merck Patent Gmbh Compositions for the treatment and prevention of neurological and pathopsychological diseases
US6544994B2 (en) 2000-06-07 2003-04-08 Eprov Ag Pharmaceutical preparation for treating or preventing cardiovascular or neurological disorders by modulating of the activity of nitric oxide synthase
MXPA03000386A (en) 2000-07-13 2004-02-26 Alteon Inc Cyanomethyl substituted thiazoliums and imidazoliums and treatments of disorders associated with protein aging.
CA2448306A1 (en) 2001-05-30 2002-12-05 Alteon Inc. Method for treating fibrotic diseases or other indications
US7758890B2 (en) 2001-06-23 2010-07-20 Lyotropic Therapeutics, Inc. Treatment using dantrolene
EP1554253A4 (en) 2002-06-03 2006-09-20 Smithkline Beecham Corp Imidazolium cxcr3 inhibitors
CA2505608A1 (en) 2002-12-06 2004-06-24 Alcon, Inc. Superoxide dismutase mimics for the treatment of ocular disorders and diseases
CA2527236A1 (en) 2003-05-27 2004-12-23 Attenuon Llc Thiotungstate analogues and uses thereof
US20050058702A1 (en) 2003-09-17 2005-03-17 Ben-Sasson Shmuel A. Compositions capable of facilitating penetration across a biological barrier
WO2006119589A2 (en) 2005-05-11 2006-11-16 Ramaekers, Vincent Prevention and therapy of cerebral folate deficiency
EP2271338A4 (en) 2008-03-31 2011-05-04 Agency Science Tech & Res Method for treating neurological disorders with imidazolium and imidazolinium compounds

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001077486A1 (en) * 2000-04-07 2001-10-18 Sofitech N.V. Scale removal
US20030176426A1 (en) * 2000-07-13 2003-09-18 Alteon, Inc. Method for treating fibrotic diseases or other indications with imidazolium agents
US20070043016A1 (en) * 2000-07-13 2007-02-22 Wagle Dilip R Method of treating fibrotic diseases or other indications with imidazolium agents
WO2005019183A1 (en) * 2003-07-21 2005-03-03 Basf Aktiengesellschaft Method for the production of purified 1,3-substituted imidazolium salts
US20050282803A1 (en) * 2004-06-17 2005-12-22 Haley Gregory J CIS-imidazolines

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
"lo-li-tec", IONIC LIQUIDS TECHNOLOGIES, 10 October 2007 (2007-10-10), pages 2, Retrieved from the Internet <URL:http//www.iolitec.de/ionic_e.htm.> *
BEYAZ, A ET AL.: "Synthesis and CMC studies of 1-methyl-3-(pentafluorophenyl) imidazolium quaternary salts", COLLOIDS AND SURFACES B: BIOINTERFACES, vol. 36, 2004, pages 71 - 74, XP008139668 *
IONIC LIQUIDS DATABASE - (ILTHERMO)., 29 September 2006 (2006-09-29), Retrieved from the Internet <URL:http//ilthermo.boulder.nist.gov/TLThermo/ionall.uix.do?event=goto&source=ionasallV,htm> *
See also references of EP2249832A4 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2271338A1 (en) * 2008-03-31 2011-01-12 Agency for Science, Technology And Research Method for treating neurological disorders with imidazolium and imidazolinium compounds
EP2271338A4 (en) * 2008-03-31 2011-05-04 Agency Science Tech & Res Method for treating neurological disorders with imidazolium and imidazolinium compounds
EP2803356A1 (en) * 2008-03-31 2014-11-19 Agency for Science, Technology and Research Method for treating neurological disorders with imidazolium and imidazolinium compounds
EP2701511A4 (en) * 2011-04-29 2015-06-10 Univ Akron Azolium and purinium salt anticancer and antimicrobial agents
US9278951B2 (en) 2011-04-29 2016-03-08 The University Of Akron Azolium and purinium salt anticancer and antimicrobial agents
US9855280B2 (en) 2015-11-06 2018-01-02 National Dong Hwa University Nanoparticles containing azolium and N-heterocyclic carbene compounds and use thereof
CN108777957A (en) * 2016-02-12 2018-11-09 新加坡科技研究局 Antibacterial patterned surface and its manufacturing method
US11584722B2 (en) 2017-04-07 2023-02-21 Xiamen Vivohealths Technology Co., Ltd. Substituted imidazole salt compounds, preparation method thereof, pharmaceutical composition thereof and application thereof
WO2023281388A1 (en) * 2021-07-05 2023-01-12 Uniwersytet Medyczny W Białymstoku Imidazolium salt, which is a derivative of lithocholic acid, a method of its preparation and its use

Also Published As

Publication number Publication date
US20110178040A1 (en) 2011-07-21
US20160000757A1 (en) 2016-01-07
EP2249832A4 (en) 2011-12-21
US20160331727A9 (en) 2016-11-17
JP5559701B2 (en) 2014-07-23
EP2249832A1 (en) 2010-11-17
CN101983057A (en) 2011-03-02
US9072729B2 (en) 2015-07-07
JP2011510979A (en) 2011-04-07

Similar Documents

Publication Publication Date Title
US9072729B2 (en) Method for treating fibrosis and cancer with imidazolium and imidazolinium compounds
Ahsan et al. Phosphocreatine protects endothelial cells from oxidized low-density lipoprotein-induced apoptosis by modulating the PI3K/Akt/eNOS pathway
Hu et al. E Platinum, a newly synthesized platinum compound, induces autophagy via inhibiting phosphorylation of mTOR in gastric carcinoma BGC-823 cells
AU2012322660B2 (en) Pyrazol-3-ones that activate pro-apoptotic BAX
ZA200203653B (en) Bicyclic and tricyclic heteroaromatic compounds.
Bavisotto et al. The dissociation of the Hsp60/pro-Caspase-3 complex by bis (pyridyl) oxadiazole copper complex (CubipyOXA) leads to cell death in NCI-H292 cancer cells
JP6316859B2 (en) Substances and methods for inhibiting replication protein A and uses thereof
WO2016109470A1 (en) Small molecule stimulators of steroid receptor coactivator proteins and their use in the treatment of cancer
JP2009543874A (en) Thiazolidinone derivatives
EP3071234A1 (en) Translation inhibitors in high-dose chemo- and/or high-dose radiotherapy
Osmaniye et al. Novel imidazole derivatives as potential aromatase and monoamine oxidase-B inhibitors against breast cancer
Li et al. Synergistic promotion of breast cancer cells death by targeting molecular chaperone GRP78 and heat shock protein 70
KR20120079254A (en) Pharmaceutical composition for leukemia therapy
EP2911667B1 (en) Triazine derivatives for the treatment of conditions associated with nicotinamide adenine dinucleotide phosphate oxidase
Chen et al. Design, synthesis and biological evaluation of novel 9-N-substituted-13-alkylberberine derivatives from Chinese medicine as anti-hepatocellular carcinoma agents
CA2969051C (en) Compounds for use in the treatment of conditions associated with nadph oxidase
EP3364971B1 (en) Heterocyclic pdk1 inhibitors for use to treat cancer
Huang et al. Synthesis and biological evaluation of a novel c-Myc inhibitor against colorectal cancer via blocking c-Myc/Max heterodimerization and disturbing its DNA binding
Rosolen et al. Intracellular ROS generation mediates maleimide-induced cytotoxicity in leukemia cells
ES2653674B2 (en) Amines derived from 5- Nitroindazole with antiprotozoal properties
RU2717310C1 (en) Aldosterone synthase inhibitors based on 2-amino-4h-pyran-3-carbonitrile derivatives
EP4232044A1 (en) Phosphaplatin compounds as therapeutic agents selectively targeting highly glycolytic tumor cells and methods thereof
Elgaafary Pharmacological studies on phytochemicals for the treatment of prostate cancer
US20150051220A1 (en) Compounds for use in therapy
WO2014011120A1 (en) Endoperoxides, synthesis and uses thereof for the treatment of neoplastic diseases such as cancer

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200980112080.7

Country of ref document: CN

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

Ref document number: 09706910

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2010544928

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2009706910

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12865668

Country of ref document: US