WO2015028662A1 - Compositions and methods for characterization and amelioration of rheumatoid arthritis - Google Patents

Compositions and methods for characterization and amelioration of rheumatoid arthritis Download PDF

Info

Publication number
WO2015028662A1
WO2015028662A1 PCT/EP2014/068501 EP2014068501W WO2015028662A1 WO 2015028662 A1 WO2015028662 A1 WO 2015028662A1 EP 2014068501 W EP2014068501 W EP 2014068501W WO 2015028662 A1 WO2015028662 A1 WO 2015028662A1
Authority
WO
WIPO (PCT)
Prior art keywords
choka
fls
rheumatoid arthritis
subject
inhibitor
Prior art date
Application number
PCT/EP2014/068501
Other languages
French (fr)
Inventor
Gary S. Firestein
Mónica GUMA
Michael Karin
Elsa SÁNCHEZ LÓPEZ
Juan Carlos LACAL
Stefano TIZIANI
Original Assignee
Consejo Superior De Investigaciones Cientificas (Csic)
University Of California
Board Of Regents, The University Of Texas System
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 Consejo Superior De Investigaciones Cientificas (Csic), University Of California, Board Of Regents, The University Of Texas System filed Critical Consejo Superior De Investigaciones Cientificas (Csic)
Publication of WO2015028662A1 publication Critical patent/WO2015028662A1/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
    • A61K31/435Heterocyclic 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
    • A61K31/4425Pyridinium derivatives, e.g. pralidoxime, pyridostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis

Definitions

  • Embodiments herein relate generally to compositions and methods for treatment of and characterization of rheumatoid arthritis. More particularly, some embodiments relate to choline kinase a (ChoKa), inhibitors thereof, and uses of such inhibitors.
  • ChoKa choline kinase a
  • a method of ameliorating rheumatoid arthritis in a subject in need of such amelioration comprises administering a therapeutically effective dose of a choline kinase a (ChoKa) inhibitor to the subject.
  • the ChoKa inhibitor comprises a small molecule.
  • the ChoKa inhibitor comprises 1 ,4-(4-4'- Bis-((4-(dimethylamine)pyridinium-1 -yl) methyl)diphenyl) butane dibromide (MN58b).
  • the ChoKa inhibitor consists of MN58b.
  • the dose of MN58b comprises about 1 mg per kg to about 10mg per kg, for example about 1 mg per kg, 2, 3, 4, 5, 6, 7, 8, 9, 10 mg per kg, including ranges between any two of the listed values. In some embodiments, the dose of MN58b comprises about 3mg per kg. In some embodiments, administration comprises injecting the ChoKa inhibitor. In some embodiments, administration of the ChoKa inhibitor inhibits proliferation of at least one resident synovial fibroblast (FLS) of the subject. In some embodiments, administration of the ChoKa inhibitor inhibits migration of at least one FLS of the subject. In some embodiments, administration of the ChoKa inhibitor reduces at least one of IL-1 b expression or MMP3 expression by the subject.
  • FLS resident synovial fibroblast
  • the method further comprises detecting an amount of proliferation or migration of at least one FLS of the subject. In some embodiments, the method further comprises detecting a presence or absence of at least one of Akt activation or MAPK activation in at least one FLS of the subject. In some embodiments, the method further comprises detecting an amount of expression of at least one of IL-1 b expression or MMP3 expression by the subject.ln some embodiments, a pharmaceutical composition comprising a dosage of a choline kinase a (ChoKa) inhibitor suitable for treatment of rheumatoid arthritis is provided.
  • ChoKa choline kinase a
  • the dosage comprises about 1 mg per kg to about 10mg per kg of body mass of a subject in need of treatment of rheumatoid arthritis.
  • the dosage further comprises an antibody for treatment of rheumatoid arthritis.
  • the antibody comprises an anti-CD20 antibody, for example rituximab.
  • a method of determining a response to at least one clinical treatment of rheumatoid arthritis administered to a subject comprises measuring the amount of at least one of ChoKa mRNA, ChoKa protein, or a choline metabolite in the subject.
  • an amount of ChoKa mRNA or ChoKa protein is measured in an FLS of the subject.
  • at least one clinical treatment of rheumatoid arthritis comprises treatment with an anti-CD20 antibody, for example, rituximab.
  • at least one clinical treatment of rheumatoid arthritis comprises treatment with a ChoKa inhibitor, for example MN58b.
  • ChoKa inhibitors can be used in conjunction with embodiments herein, for example small molecules ⁇ e.g. MN58b and 1 ,1 '-(biphenyl- 4,4'-diylmethylene)bis [4-(4-chloro-N-methylanilino-)quinolinium]dibromide (RSM- 932A); see, e.g. U.S. Pat. No. 8,481256 and U.S. Pub. Nos. 2007/0185170 and 2010/0068302, each of which is incorporated by reference in its entirety herein), antibodies ⁇ e.g monoclonal antibodies such as AD3, AD8 and AD1 1 , described in PCT Pub. No.
  • RNAi see U.S. Pub. No. 2010/0068302
  • antisense nucleic acids ribozymes, aptamers, and the like.
  • a method of identifying a metabolic profile indicative of osteoarthritis or rheumatoid arthritis comprises comparing the level of at least one of GPC, ChoKa mRNA, ChoKa protein, or a choline metabolite in individuals with osteoarthritis or rheumatoid arthritis to individuals who do not have osteoarthritis or rheumatoid arthritis.
  • the method of diagnosing osteoarthritis or rheumatoid arthritis comprises determining whether a subject has level of at least one of GPC, ChoKa mRNA, ChoKa protein, or a choline metabolite indicative of osteoarthritis or rheumatoid arthritis. In some embodiments, the method further comprises administering a treatment for osteoarthritis or rheumatoid arthritis if the subject has level of at least one of GPC, ChoKa mRNA, ChoKa protein, or a choline metabolite indicative of osteoarthritis or rheumatoid arthritis.
  • Figure 1 is a photograph illustrating a RA synovial that was immunostained with IgG (A) and cholinekinase a antibody (B).
  • FIG. 2 is a photograph of a western blot. RA FLS were stimulated by several cytokines and ChoKa expression was confirmed by WB.
  • C control; T: TNF; P: PDGF; E: EGF; L: LPS.
  • Figures 3A and 3B are graphs depicting NMR spectra.
  • ⁇ NMR spectra obtained from FLS RA Figure 3A
  • Figure 3A grown in culture (blue) no stimulus (red) after 24hrs of PDGF (10ng/ml) stimulation.
  • Figure 3B IMR-90 fibroblast cell line spectra.
  • Figure 4 is a photograph of a western blot. Lysates of RA FLS were prepared when indicated after PDGF stimulation and were analyzed for the expression of the indicated proteins.
  • Figure 5 is a graph depicting cell growth. FLS were cultured in presence of PDGF with different MN58b concentrations. Growth was measured on day 4 using MTT.
  • Figure 6 is a series of photographs depicting effects of ChoKa inhibitor on synoviocyte migration.
  • FIGURES 7A-7D ARE GRAPHS DEPICTING CLINICAL SCORES (FIGURE 7A), HISTOLOGICAL SCORES (FIGURE 7B), RELATIVE IL-1 B MRNA EXPRESSION (FIGURE 7C), AND RELATIVE MMP3 MRNA EXPRESSION (FIGURE 7D) IN PASSIVE K/BXN SERUM TRANSFER ARTHRITIS AFTER VEHICLE AND CHOKa INHIBITOR (3 MG/KG) TREATMENT.
  • Choline kinase a (ChoKa) is an enzyme essential for phosphatidylcholine (PtdCho ) biosynthesis, and is involved in cell proliferation, growth and invasion. It has been recently recognized as both a prognostic marker and a therapeutic target in various types of human cancers.
  • RA Rheumatoid Arthritis
  • synovial hyperplasia contributes to inflammation and joint destruction.
  • Synovial fibroblast (FLS) in the intimal lining, especially in the pannus are the major effectors of cartilage damage through production of extracellular matrix degrading enzymes such as MMPs and cathepsins.
  • RA FLS possess unique aggressive phenotype, such as cartilage invasion.
  • ChoKa expression is regulated by inflammatory cytokines and regulates key FLS functions that might contribute to cartilage destruction in RA.
  • ChoKa inhibition in a model of inflammatory arthritis decrease paw swelling and histological score.
  • ChoKa inhibition is a novel therapy for RA and will be a non-invasive biomarker of inflammation/joint damage and response to therapy.
  • RA rheumatoid arthritis
  • FLS fibroblast-like synoviocytes
  • FLS contribute to synovial inflammation by producing inflammatory mediators, extracellular matrix degrading enzymes such as MMPs and cathepsins and recruiting and activating immune cells.
  • RA FLS possess unique aggressive phenotype, such as cartilage invasion.
  • MMPs and cathepsins extracellular matrix degrading enzymes
  • RA FLS possess unique aggressive phenotype, such as cartilage invasion.
  • Small molecule inhibitors that inhibit key signaling molecules in RA have the potential to improve efficacy and to overcome limitations of antibody based approaches.
  • new rationally designed disease modifying agents that directly target FLS are needed to complement current therapies.
  • ChoKa is essential for PtdCho biosynthesis, which is required for the increased cancer cell proliferation, tumor progression and invasiveness, making it a potential prognostic marker of some cancers.
  • Selective inhibition of ChoKa also attenuates MAPK and P13K/Akt signaling, which can be associated with a decrease in the growth of cells.
  • RA cultured FLS possess a unique tumor like behavior, and aggressive phenotype, such as cartilage invasiveness.
  • MAPK and P13K/Akt are signal pathways that regulate synoviocyte function in RA such as metalloproteinase (MMP) expression and synoviocyte growth and survival, which may explain its aggressive phenotype.
  • MMP metalloproteinase
  • RA rheumatoid arthritis
  • FLS synovial fibroblasts
  • FLS in the intimal lining, especially in the pannus are the major effectors of cartilage damage through production of extracellular matrix degrading enzymes such as MMPs and cathepsins[2].
  • RA FLS possess unique aggressive phenotype, such as cartilage invasion[3].
  • the molecular mechanisms that regulate FLS behavior in RA are poorly understood and represent a major obstacle for developing therapeutic interventions that modulate these functions.
  • Small molecule inhibitors that inhibit key signaling molecules in RA have the potential to improve efficacy and to overcome limitations of antibody based approaches[4-6].
  • new rationally designed disease modifying agents that directly target FLS can be useful for replacing or complementing current therapies.
  • Metabolomics allows for a global assessment of a cellular state within the context of the immediate environment, taking into account genetic regulation, altered kinetic activity of enzymes, and changes in metabolic reactions[7]. Compared with genomics or proteomics, metabolomics reflects changes in phenotype and therefore function[7]. Studying patients using a metabolomic strategy may reveal underlying biochemical phenomena associated with the disease, thus providing insights that help the development of a better understanding of mechanisms underlying disease, and to develop new strategies for treatment. Metabolic profiling has also been used to identify biomarkers for several diseases. Systemic diseases like RA are likely associated with changes in a complicated array of chemical reactions and metabolites that stem from a diverse set of metabolic pathways. However, few works have addressed metabolic changes in RA[8-12].
  • tumor metabolome is beginning to be characterized[13-15].
  • tumors in general, display elevated phospholipid levels characterized by increases in the levels of phophocholine (PCho) and total choline-containing metabolites together with decreases in the glycerophosphocholine (GPC)/PCho ratio (a phenomenon known as the "GPC-to-PCho switch"; Fig.1 )[16-18].
  • GPC glycerophosphocholine
  • Fig.1 glycerophosphocholine
  • Fig.1 glycerophosphocholine
  • Elevated PCho levels can be partially attributed to an increased activity of choline kinase (ChoK) catalyzing the first step in the Kennedy pathway, an enzyme recently recognized as both a prognostic marker and a therapeutic target in various types of human cancers[18, 23, 24].
  • ChoK can also take on a rate-limiting, regulatory role in phosphatidylcholine (PtdCho) biosynthesis under some circumstances.
  • PtdCho phosphatidylcholine
  • ChoK is primarily located in the cytoplasm of cells from various tissues. At least three isoforms of ChoK exist in mammalian cells, and these are encoded by two genes: ChoKa and ChoKb.
  • ChoKa-1 and ChoKa-2 are derived from ChoKa by alternative splicing. Homodimeric or heterodimeric forms of ChoK are enzymatically active.
  • the upregulation of ChoK activity in cancer probably results from an increase in ChoKa expression, which would lead to a higher proportion of ChoKa homodimers in cancer cells and in turn a higher ChoK activity level.
  • ChoKa is essential for PtdCho biosynthesis, which is required for the increased cancer cell proliferation, tumor progression and invasiveness, making it a potential prognostic marker of some cancers.
  • Selective inhibition of choline kinase also attenuates MAPK and PI3K/Akt signaling, which was associated with a decrease in the growth of cells[25].
  • RA cultured FLS possess a unique tumor like behavior, and aggressive phenotype, such as cartilage invasiveness[2, 3].
  • MAPK and PI3K/Akt are signal pathways that regulate synoviocyte function in RA such as metalloproteinase (MMP) expression and synoviocyte growth and survival, which may explains its aggressive phenotype[26, 27].
  • MMP metalloproteinase
  • MN58b[28] (1 ,4-(4-4'-Bis-((4- (dimethylamine)pyridinium-l -yl) methyl ⁇ diphenyl) butane dibromide) exhibits selective inhibition of choline kinase, inhibits proliferation of cancer cells in vitro with an IC50 of 1 -1 OnM, and displays therapeutic activity against human tumor xenografts in vivo.
  • ChoKa regulates key FLS functions that might contribute to cartilage destruction in RA.
  • Initial studies also show a novel role for choline kinase in vivo.
  • ChoKaD is a key regulator ofD FLS functions that contribute to aggressive behavior and joint destruction in RA and that selective ChoKa inhibition in RA will be disease modifying by directly modulating synoviocyte mediated cartilage destruction and that choline-containing compounds detected by MRS might be a non-invasive biomarker of inflammation/joint damage and response to therapy.
  • MN58b (1.4-(4-4'-Bis-((4- (dimethylamine)pyridinium-l -yl) methyl ⁇ diphenyl) butane dibromide) exhibits selective inhibition of ChoKa, inhibits proliferation of cancer cells in vitro with an IC 50 of 1 -10 ⁇ , and displays therapeutic activity against human tumor xenografts in vivo.
  • Our data shows that ChoKa regulates key FLS functions that might contribute to cartilage destruction in RA.
  • Initial studies also show that ChoKa inhibition in a model of inflammatory arthritis decrease paw swelling and histological score.
  • a ChoKa inhibitor for example MN58b, also comprises a pharmaceutically acceptable excipient. Therefore, some embodiments are also directed to a pharmaceutical composition as disclosed above, wherein the pharmaceutical composition additionally comprises a pharmaceutically acceptable excipient.ln general all excipients known by a person skilled in the art are suitable within embodiments herein.
  • excipients examples include calcium carbonate, kaolin, sodium hydrogen carbonate, lactose, D-mannitol, starches, crystalline cellulose, talc, granulated sugar, porous substances, etc.
  • a ChoKa inhibitor for example MN58b, may be used as bulk itself but usually be formulated into pharmaceutical preparations together with a suitable amount of "carrier for pharmaceutical preparation" according to ordinary methods.
  • compositions and methods described herein may also contain additionally diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials well known in the art.
  • carriers for pharmaceutical preparation comprises, for example, excipients as defined herein, binders, e.g., dextrin, gums, a-starch, gelatin, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, pullulan, etc., thickening agents, e.g., natural gums, cellulose derivatives, acrylic acid derivatives, etc., disintegrators, e.g., carboxy- methyl cellulose, croscarmellose sodium, crospovidone, low-substitution hydroxypropyl cellulose, partial a-starch, etc., solvents, e.g., water for injections, alcohol, propylene glycol, macrogol, sesame oil, corn oil, etc., dispersants, e.g., Tween 80, HCO60, polyethylene glycol, carboxymethyl cellulose, sodium alginate, etc., solubilizers, e.g., poly- ethylene glycol, propylene glycol, propylene
  • Preferred methods of administration of the pharmaceutical compositions described above include oral and parenteral, e.g., i.v. infusion, i.v. bolus and i.m. injection formulated so that a unit dosage comprises a therapeutically effective amount of each active component or some submultiples thereof.
  • the compounds may be employed in powder or crystalline form, in liquid solution, or in suspension.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • compositions for injection may be prepared in unit dosage form in ampules, or in multidose containers.
  • the composition will generally be sterile and pyrogen- free, when intended for delivery by injection into the subject.
  • the injectable compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain various formulating agents.
  • the active ingredient may be in powder (lyophilized or non-lyophilized) form for reconstitution at the time of delivery with a suitable vehicle, such as sterile water.
  • Carriers suitable for an injectable pharmaceutical composition are typically comprised sterile water, saline or another injectable liquid, e.g., peanut oil for intramuscular injections. Also, various buffering agents, preservatives and the like can be included.
  • the pharmaceutical composition according to some embodiments may also be administered parenterally in a sterile medium. Depending on the vehicle and concentration used, the drug can either be suspended or dissolved in the vehicle.
  • adjuvants such as local anaesthetic, preservative and buffering agents can be dissolved in the vehicle.
  • the proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants.
  • microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. It is also preferred to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatine. Intra-venous infusion is another possible route of administration for the compounds used according to some embodiments herein.
  • Orally administrable pharmaceutical compositions may be in the form of tablets, capsules, powders, granules, lozenges, liquid or gel preparations, such as oral, topical, or sterile parenteral solutions or suspensions.
  • the oral compositions may utilize carriers such as conventional formulating agents, and may include sustained release properties as well as rapid delivery forms.
  • Such compositions and preparations should contain at least 0.1 % of active compounds.
  • the percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 60% of the weight of a given unit dosage form. The amount of active compound in such therapeutically useful compositions is such that an effective dosage level will be obtained.
  • Tablets and capsules for oral administration may be in unit dose presentation form, and may also contain conventional excipients such as binding agents, for example syrup, acacia, gelatine, sorbitol, tragacanth, or polyvinylpyrrolidone; fillers for example lac- tose, sugar, maize-starch, calcium phosphate, sorbitol or glycine; tabletting lubricant, for example magnesium stearate, talc, polyethylene glycol or silica; disintegrates for example potato starch, or acceptable wetting agents such as sodium lauryl sulphate.
  • the tablets may be coated according to methods well known to a person skilled in the art.
  • Oral liquid preparations may be in the form of, for example, aqueous or oily sus- pensions, solutions, emulsions, syrups or elixirs, or may be presented as a dry product for reconstitution with water or other suitable vehicle before use.
  • Such liquid preparations may contain conventional additives such as suspending agents, for example sorbitol, syrup, methyl cellulose, glucose syrup, gelatine hydrogenated edible fats; emulsifying agents, for example lecithin, sorbitan monooleate, or acacia; non-aqueous vehicles which may include edible oils, for example almond oil, fractionated coconut oil, oily esters such as glycerine, propylene glycol, or ethyl alcohol; preservatives, for example methyl or propyl p-hydroxybenzoate or sorbic acid, and if desired conventional flavouring or colouring agents.
  • suspending agents for example sorbitol, syrup, methyl cellulose, glucose syrup, gelatine hydrogenated edible fats
  • emulsifying agents for example lecithin, sorbitan monooleate, or acacia
  • non-aqueous vehicles which may include edible oils, for example almond oil, fractionated coconut oil, oily esters such as glycerine, prop
  • compositions according to some embodiments herein may also be prepared in suitable forms for absorption through the mucous membranes of the nose and throat or bronchial tissues and may conveniently take the form of powder or liquid sprays or inhalants, lozenges, throat paints, etc.
  • the preparations may be presented as individual capsules, in liquid or semi-solid form, or may be used as drops, etc.
  • Suitable subjects for the administration of the formulation of some embodiments herein include mammals, primates, humans, and other animals.
  • the animal subject is a mammal, generally a domesticated farm mammal, e.g. horse, pig, cow, sheep, goat etc., or a companion animal, e.g. cat, dog etc..
  • In vitro antibacterial activity is predictive of in vivo activity when the compositions are administered to a mammal infected with a susceptible bacterial organism.
  • mice were injected with K/BxN sera on day 0.
  • MN58b (3mg/kg) was injected daily i.p. beginning on day 0 or day 4 after serum administration.
  • Clinical arthritis scores were serially assessed. Joints were evaluated for inflammation and joint damage using histology and a semiquantitative scoring system.
  • ChoKa mRNA and protein were highly expressed in RA synovial tissue and in cultured FLS. Its expression in FLS was increased 2-3-fold after tumor necrosis factor (TNF) and PDGF stimulation, respectively with peak expression within 48 hours. Metabolomic studies of choline-containing compounds in cultured FLS extracts showed increased levels of phosphocholine in RA FLS, confirming activation of this pathway. ChoKa regulates key FLS functions that might contribute to cartilage destruction in RA. For example, ChoKa inhibition with MN58b (5 ⁇ ) reduced proliferation by 79 ⁇ 3.2% and migration by 54 ⁇ 15% (p ⁇ 0.05). ChoKa inhibition also markedly increased H 2 O 2 -induced apoptosis in FLS.
  • ChoKa protein is localized to the intimal lining cells, we evaluated its expression and regulation in cultured RA FLS derived from this region. Protein expression was confirmed with Western blot analysis. To determine whether ChoKa is regulated by inflammatory mediators implicated in RA, we stimulated RA FLS with TNF, IL-1 , PDGF, EGF or LPS for 48 hrs, and protein was assayed by WB. ChoKa increased following TNF, PDGF and EGF stimulation (Fig. 2) while LPS had no effect.
  • FIG. 3A One-dimensional 1 H NMR spectra of aqueous extracts revealed and activated choline profile in RA FLS (Fig. 3A).
  • the relative areas of signal components due to individual PC metabolites (GPC, PCho and Cho) are more similar to tumor than non-tumor cells (Fig. 1 ), PCho becoming the predominant metabolite, which is further increased after PDGF stimulation (GPC:PC ratio ⁇ 0.1 ).
  • Fig. 3 shows 1 H NMR spectra of normal IMR-90 fibroblast cell line, which shows a GPC/PC ratio around 1.
  • EXAMPLE 6 CHOLINE KINASE REGULATES RA FLS GROWTH
  • PDGF is a known chemotactic agent for mesenchymal cells
  • ChoKa is required for FLS migration.
  • FLS were grown to a confluent monolayer in a 6-well plates and wounded area was generated with 1 ml micropipette tip.
  • Cells were cultured in low serum Med (1 % FBS) alone or with PDGF-BB 10 ng/ml +/- MN58b (1 ⁇ ) or vehicle (Fig. 6).
  • Cell migration in response to PDGF was dramatically decreased in presence of CHoKa inhibitor MN58b at 1 ⁇ .
  • EXAMPLE 8 EFFECT OF CHOKa INHIBITOR IN KXB/N MODEL OF
  • MN58b a choline kinase inhibitor
  • WT mice were injected with 150ul serum from adult K/BxN mice on day 0.
  • MN58b significantly decreased paw swelling in this model (p ⁇ 0.01 for vehicle compared with daily 3 mg/kg) (Fig. 7A). It also decreased histological score (Fig. 7B) and IL-1 b and MMP3 mRNA expression (Fig 7 C and D).
  • mice were immunized with an intradermal injection on day 0 with methylated BSA (mBSA) in complete Freund's adjuvant. 2 weeks later mice were bled and antibodies against mBSA were analyzed by ELISA. MN58b treatment (daily 3mg/kg) did not change the titer of antibodies against mBSA suggesting that the inhibitor was acting more in the innate than in the adaptive response compartment.
  • rheumatoid synoviocytes display certain unique features that are reminiscent of transformed cells, it is not clear whether these features are inherent in RA FLS (transformed aggressors) or are somehow imprinted due to exposure to cytokines in the rheumatoid milieu in vivo (passive responders). Studies using normal and OA FLS after cytokine stimulation will determine if cytokines mimic the change in choline metabolism in RA FLS.
  • synovium and synoviocytes Preparation of synovium and synoviocytes. Synovium and FLS will be obtained from patients undergoing total joint replacement or synovectomy who meet the 1987 revised American College of Rheumatology criteria for seropositive RA or patients with OA as previously described[29]. RA patients will discontinue methotrexate for at least 1 month prior to surgery to minimize the influence of methotrexate on folate metabolism and methyl donors[30]. For FLS lines, tissue is enzymatically dispersed and cells allowed to adhere overnight. Nonadherent cells are washed off, and the adherent FLS are grown in DMEM containing 10% FCS.
  • FLS are used from passage 3 through 8 during which time they are a homogeneous population of cells ( ⁇ 1 % CD1 1 b positive, ⁇ 1 % phagocytic, and ⁇ 1 % FcR II and FcR III receptor positive)[31].
  • 1 H-MRS platform has become an established tool for the comprehensive analysis of the metabolome in biological samples for both polar and lipophilic metabolites.
  • RA FLS have a tumor-like metabolite pattern
  • cells from normal, RA and OA FLS (5 cell lines each), as well as tumor fibroblasts (e.g., PVNS) and dermal fibroblasts will be isolated and prepared for the 1 H-MRS analysis as described[21 , 32].
  • Acquisition of the 1 H-MRS metabolic profiles of both the polar and apolar fractions will be performed using a Bruker Avance 700 MHz NMR spectrometer equipped with high throughput robotics.
  • RA FLS and synovium Is choline kinase expression greater in RA FLS and synovium? Expression will be determined by qPCR and WB. We will also determine the expression of ChoKa in OA, RA and normal FLS (5 cell lines each) before and after stimulation with cytokines (IL-1 at 2ng/ml, TNF or PDGF BB at 10ng/ml) by Western blot (WB) and qPCR. RA, OA and normal synovium will be immunostained (5 tissues each) to localize ChoKa. Digital image analysis will determine 1 ) whether intimal lining expression is greater than sublining expression; and 2) whether these proteins are more abundant in RA compared with OA or normal tissues.
  • EXAMPLE 12 DETERMINING THE EFFECT OF CHOLINE KINASE INHIBITION ON GENE EXPRESSION, SIGNALING AND FUNCTION
  • ChoKa will probably decrease P-protein and MMP expression based on our preliminary studies. Cytokines, especially TNF and IL-1 , enhance the destructive properties of RA FLS and their effects will probably be potentiated by growth factors like PDGF.
  • Apoptosis Human and murine FLS will be treated with anti-Fas antibody or 100 uM hydrogen peroxide to induce apoptosis. Cells will be evaluated from 4 to 24 hr later using trypan blue dye exclusion. A histone release assay performed by ELISA will be performed to confirm the results.
  • Proliferation Cell proliferation will be evaluated using cultured FLS (human and murine) and stimulating with medium or PDGF (10ng/ml) in the presence of ChoKa inhibitor. Tritiated thymidine will be used to quantify DNA synthesis after 2-7 days.
  • ChoKa regulate migration and MMP expression through activation of Akt/MAPK pathway respectively Because ChoKa regulates MAPK and Akt signaling after PDGF stimulation in RA FLS, and these signaling have been involved in MMP and migration respectively, we will determine the effect of Akt and MAPK siRNA on those functions together with ChoKa inhibition. We hypothesize that if the effect of ChoKa is through these two pathways, ChoKa inhibitor will not add any further effect after blockade of MAPK and Akt by siRNAs or chemical inhibitors.
  • MAPK either JNK, p38 or ERK siRNA
  • MAPK will be chosen according to the effect of ChoKa inhibition on those phospho-protein after PDGF stimulation
  • Akt will be knocked-down by siRNA in RA FLS and at day 5
  • MMP expression and migration assays after PDGF stimulation will be realized and assessed as detailed above after chemical ChoKa inhibition.
  • EXAMPLE 13 DETERMING EFFECTS OF CHOLINEKINASE INHIBITION IN
  • Clinical scoring will be done daily and experiments will be terminated on day 10.
  • the endpoints for our analysis 1 ) Clinical scores and ankle diameter; 2) Ankle histology, for synovial hyperplasia, cartilage erosion, cartilage proteoglycan, inflammation and bone destruction 3) IHC for infiltrating cell types 4) Inflammatory gene expression (mRNA and protein analysis) and 5) Alteration in synovial signaling in the arthritic joints.
  • ⁇ Synovial proliferation and cartilage erosion Hind paws of mice are harvested for paraffin sections and are stained with H&E, safranin O-fast green for proteoglycan content. We will determine in situ FLS proliferation by performing IHC for PCNA and Ki67, nuclear antigens used as an index of cell proliferation in tissue. Results will be quantified by image analysis.
  • ⁇ Synovial mediator expression Ankle extracts will be assayed by tissue extract ELISA and qPCR of pro-inflammatory cytokines (TNF and IL-6), prototypical Th1 , Th2, and Th17 cytokines (IFNy, IL-4 and IL-17A), and proteases (MMP3, MMP13) as possible mediators regulated by the Akt/MAPK pathway.
  • MN58b inhibitor is well tolerated by mice at the doses tested, and preliminary data already shows no changes in histology of liver and kidney.
  • Biomarkers associated with clinical response might not be the same biomarkers that predict risk of further joint damage.
  • MRS is a commonly used analytical method to analyze the metabolome of body fluids such as urine and blood serum[36].
  • Recent studies demonstrate the applicability of NMR-based metabolomics using serum samples for the diagnosis and prognosis[37].
  • NMR-based metabolomics using serum samples for the diagnosis and prognosis[37].
  • RA a non-invasive biomarker of transformation, staging and response to therapy disease.
  • RA metabolic changes in RA and little information of this metabolic pathway has been addressed in inflammation.
  • We will take advantage of samples that we already have in our department to explore the relationship between choline metabolites and clinical response.
  • a range includes each individual member.
  • a group having 1 -3 articles refers to groups having 1 , 2, or 3 articles.
  • a group having 1 -5 articles refers to groups having 1 , 2, 3, 4, or 5 articles, and so forth.

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

According to some embodiments herein, methods and compositions for amerlioration of rheumatoid arthritis are provided. According to some embodiments herein, methods and compositions for characterization of rheumatoid arthritis are provided. The methods and compositions of some embodiments include inhibitors of ChoKα.

Description

COMPOSITIONS AND METHODS FOR CHARACTERIZATION AND AMELIORATION OF RHEUMATOID ARTHRITIS
Field
Embodiments herein relate generally to compositions and methods for treatment of and characterization of rheumatoid arthritis. More particularly, some embodiments relate to choline kinase a (ChoKa), inhibitors thereof, and uses of such inhibitors.
SUMMARY
In some embodiments, a method of ameliorating rheumatoid arthritis in a subject in need of such amelioration is provided, in which the method comprises administering a therapeutically effective dose of a choline kinase a (ChoKa) inhibitor to the subject. In some embodiments, the ChoKa inhibitor comprises a small molecule. In some embodiments, the ChoKa inhibitor comprises 1 ,4-(4-4'- Bis-((4-(dimethylamine)pyridinium-1 -yl) methyl)diphenyl) butane dibromide (MN58b). In some embodiments, the ChoKa inhibitor consists of MN58b. In some embodiments, the dose of MN58b comprises about 1 mg per kg to about 10mg per kg, for example about 1 mg per kg, 2, 3, 4, 5, 6, 7, 8, 9, 10 mg per kg, including ranges between any two of the listed values. In some embodiments, the dose of MN58b comprises about 3mg per kg. In some embodiments, administration comprises injecting the ChoKa inhibitor. In some embodiments, administration of the ChoKa inhibitor inhibits proliferation of at least one resident synovial fibroblast (FLS) of the subject. In some embodiments, administration of the ChoKa inhibitor inhibits migration of at least one FLS of the subject. In some embodiments, administration of the ChoKa inhibitor reduces at least one of IL-1 b expression or MMP3 expression by the subject. In some embodiments, the method further comprises detecting an amount of proliferation or migration of at least one FLS of the subject. In some embodiments, the method further comprises detecting a presence or absence of at least one of Akt activation or MAPK activation in at least one FLS of the subject. In some embodiments, the method further comprises detecting an amount of expression of at least one of IL-1 b expression or MMP3 expression by the subject.ln some embodiments, a pharmaceutical composition comprising a dosage of a choline kinase a (ChoKa) inhibitor suitable for treatment of rheumatoid arthritis is provided. In some embodiments, the dosage comprises about 1 mg per kg to about 10mg per kg of body mass of a subject in need of treatment of rheumatoid arthritis. In some embodiments,the dosage further comprises an antibody for treatment of rheumatoid arthritis. In some embodiments, the antibody comprises an anti-CD20 antibody, for example rituximab.
In some embodiments, a method of determining a response to at least one clinical treatment of rheumatoid arthritis administered to a subject is provided, in which the method comprises measuring the amount of at least one of ChoKa mRNA, ChoKa protein, or a choline metabolite in the subject. In some embodiments, an amount of ChoKa mRNA or ChoKa protein is measured in an FLS of the subject. In some embodiments, at least one clinical treatment of rheumatoid arthritis comprises treatment with an anti-CD20 antibody, for example, rituximab. In some embodiments, at least one clinical treatment of rheumatoid arthritis comprises treatment with a ChoKa inhibitor, for example MN58b. The skilled artisan will recognize that a variety of ChoKa inhibitors can be used in conjunction with embodiments herein, for example small molecules {e.g. MN58b and 1 ,1 '-(biphenyl- 4,4'-diylmethylene)bis [4-(4-chloro-N-methylanilino-)quinolinium]dibromide (RSM- 932A); see, e.g. U.S. Pat. No. 8,481256 and U.S. Pub. Nos. 2007/0185170 and 2010/0068302, each of which is incorporated by reference in its entirety herein), antibodies {e.g monoclonal antibodies such as AD3, AD8 and AD1 1 , described in PCT Pub. No. WO2007/138143, hereby incorporated by reference in its entirety), RNAi (see U.S. Pub. No. 2010/0068302), antisense nucleic acids, ribozymes, aptamers, and the like. In some embodiments, a method of identifying a metabolic profile indicative of osteoarthritis or rheumatoid arthritis is provided, in which the method comprises comparing the level of at least one of GPC, ChoKa mRNA, ChoKa protein, or a choline metabolite in individuals with osteoarthritis or rheumatoid arthritis to individuals who do not have osteoarthritis or rheumatoid arthritis. In some embodiments, the method of diagnosing osteoarthritis or rheumatoid arthritis is provided, in which the method comprises determining whether a subject has level of at least one of GPC, ChoKa mRNA, ChoKa protein, or a choline metabolite indicative of osteoarthritis or rheumatoid arthritis. In some embodiments, the method further comprises administering a treatment for osteoarthritis or rheumatoid arthritis if the subject has level of at least one of GPC, ChoKa mRNA, ChoKa protein, or a choline metabolite indicative of osteoarthritis or rheumatoid arthritis.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a photograph illustrating a RA synovial that was immunostained with IgG (A) and cholinekinase a antibody (B).
Figure 2 is a photograph of a western blot. RA FLS were stimulated by several cytokines and ChoKa expression was confirmed by WB. C: control; T: TNF; P: PDGF; E: EGF; L: LPS.
Figures 3A and 3B are graphs depicting NMR spectra. Ή NMR spectra obtained from FLS RA (Figure 3A) grown in culture (blue) no stimulus (red) after 24hrs of PDGF (10ng/ml) stimulation. (Figure 3B) IMR-90 fibroblast cell line spectra.
Figure 4 is a photograph of a western blot. Lysates of RA FLS were prepared when indicated after PDGF stimulation and were analyzed for the expression of the indicated proteins. Figure 5 is a graph depicting cell growth. FLS were cultured in presence of PDGF with different MN58b concentrations. Growth was measured on day 4 using MTT. Figure 6 is a series of photographs depicting effects of ChoKa inhibitor on synoviocyte migration.
FIGURES 7A-7D ARE GRAPHS DEPICTING CLINICAL SCORES (FIGURE 7A), HISTOLOGICAL SCORES (FIGURE 7B), RELATIVE IL-1 B MRNA EXPRESSION (FIGURE 7C), AND RELATIVE MMP3 MRNA EXPRESSION (FIGURE 7D) IN PASSIVE K/BXN SERUM TRANSFER ARTHRITIS AFTER VEHICLE AND CHOKa INHIBITOR (3 MG/KG) TREATMENT. DETAILED DESCRIPTION
Choline kinase a (ChoKa) is an enzyme essential for phosphatidylcholine (PtdCho ) biosynthesis, and is involved in cell proliferation, growth and invasion. It has been recently recognized as both a prognostic marker and a therapeutic target in various types of human cancers. In Rheumatoid Arthritis (RA), synovial hyperplasia contributes to inflammation and joint destruction. Synovial fibroblast (FLS) in the intimal lining, especially in the pannus are the major effectors of cartilage damage through production of extracellular matrix degrading enzymes such as MMPs and cathepsins. RA FLS possess unique aggressive phenotype, such as cartilage invasion. It is appreciated herein that ChoKa expression is regulated by inflammatory cytokines and regulates key FLS functions that might contribute to cartilage destruction in RA. As shown herein, ChoKa inhibition in a model of inflammatory arthritis decrease paw swelling and histological score. ChoKa inhibition is a novel therapy for RA and will be a non-invasive biomarker of inflammation/joint damage and response to therapy.
The enzyme has also been implicated in cancer disease progression, metastasis, and invasiveness. The unique tumor-like behavior of rheumatoid arthritis (RA) fibroblast-like synoviocytes (FLS) led us evaluate whether this pathway could play a role in inflammation and joint damage due to synovitis. Therefore, we examined the expression and function of ChoKa in RA FLS and performed a targeted metabolomics assessment of this pathway. These studies suggest that ChoKa contributes to the rheumatoid aggressive phenotype. Pathogenesis of RA disease can involve a complex interaction between the innate and the adaptive arm of the immune system in concert with the resident synovial fibroblasts (FLS). FLS contribute to synovial inflammation by producing inflammatory mediators, extracellular matrix degrading enzymes such as MMPs and cathepsins and recruiting and activating immune cells. RA FLS possess unique aggressive phenotype, such as cartilage invasion. However, the molecular mechanisms that regulate FLS behavior in RA are poorly understood and represent a major obstacle for developing therapeutic interventions that modulate these functions. Small molecule inhibitors that inhibit key signaling molecules in RA have the potential to improve efficacy and to overcome limitations of antibody based approaches. Thus, new rationally designed disease modifying agents that directly target FLS are needed to complement current therapies.
To the best of Applicant's knowledge at the time of filing, there are no disease modifying agents that directly target FLS. There are no specific biomarker of inflammation/joint damage and response to therapy in rheumatoid arthritis.
ChoKa is essential for PtdCho biosynthesis, which is required for the increased cancer cell proliferation, tumor progression and invasiveness, making it a potential prognostic marker of some cancers. Selective inhibition of ChoKa also attenuates MAPK and P13K/Akt signaling, which can be associated with a decrease in the growth of cells. RA cultured FLS possess a unique tumor like behavior, and aggressive phenotype, such as cartilage invasiveness. Moreover, MAPK and P13K/Akt are signal pathways that regulate synoviocyte function in RA such as metalloproteinase (MMP) expression and synoviocyte growth and survival, which may explain its aggressive phenotype. As such, it was determined if this unique aggressive phenotype could be secondary to a higher ChoKa activity level, similar to what it is observed in tumor cells, and to determine suitability as therapeutic target and biomarker of inflammation/joint damage.The hallmark of rheumatoid arthritis (RA) is synovial inflammation, hyperplasia and joint destruction[1]. Pathogenesis of the disease involves a complex interaction between the innate and the adaptive arm of the immune system in concert with the resident synovial fibroblasts (FLS). FLS contribute to synovial inflammation by producing inflammatory mediators and recruiting and activating immune cells[2]. In addition, FLS in the intimal lining, especially in the pannus are the major effectors of cartilage damage through production of extracellular matrix degrading enzymes such as MMPs and cathepsins[2]. RA FLS possess unique aggressive phenotype, such as cartilage invasion[3]. However, the molecular mechanisms that regulate FLS behavior in RA are poorly understood and represent a major obstacle for developing therapeutic interventions that modulate these functions. Small molecule inhibitors that inhibit key signaling molecules in RA have the potential to improve efficacy and to overcome limitations of antibody based approaches[4-6]. Thus, new rationally designed disease modifying agents that directly target FLS can be useful for replacing or complementing current therapies.
Metabolomics allows for a global assessment of a cellular state within the context of the immediate environment, taking into account genetic regulation, altered kinetic activity of enzymes, and changes in metabolic reactions[7]. Compared with genomics or proteomics, metabolomics reflects changes in phenotype and therefore function[7]. Studying patients using a metabolomic strategy may reveal underlying biochemical phenomena associated with the disease, thus providing insights that help the development of a better understanding of mechanisms underlying disease, and to develop new strategies for treatment. Metabolic profiling has also been used to identify biomarkers for several diseases. Systemic diseases like RA are likely associated with changes in a complicated array of chemical reactions and metabolites that stem from a diverse set of metabolic pathways. However, few works have addressed metabolic changes in RA[8-12]. In other fields, like oncology, the tumor metabolome is beginning to be characterized[13-15]. Using standard metabolomic methods, tumors, in general, display elevated phospholipid levels characterized by increases in the levels of phophocholine (PCho) and total choline-containing metabolites together with decreases in the glycerophosphocholine (GPC)/PCho ratio (a phenomenon known as the "GPC-to-PCho switch"; Fig.1 )[16-18]. Because choline-containing compounds are detected by MRS, increased levels of these compounds provide a non-invasive biomarker of transformation, staging and response to therapy[19-23]. Elevated PCho levels can be partially attributed to an increased activity of choline kinase (ChoK) catalyzing the first step in the Kennedy pathway, an enzyme recently recognized as both a prognostic marker and a therapeutic target in various types of human cancers[18, 23, 24]. ChoK can also take on a rate-limiting, regulatory role in phosphatidylcholine (PtdCho) biosynthesis under some circumstances. ChoK is primarily located in the cytoplasm of cells from various tissues. At least three isoforms of ChoK exist in mammalian cells, and these are encoded by two genes: ChoKa and ChoKb. The two functional isoforms, ChoKa-1 and ChoKa-2, are derived from ChoKa by alternative splicing. Homodimeric or heterodimeric forms of ChoK are enzymatically active. The upregulation of ChoK activity in cancer probably results from an increase in ChoKa expression, which would lead to a higher proportion of ChoKa homodimers in cancer cells and in turn a higher ChoK activity level. ChoKa is essential for PtdCho biosynthesis, which is required for the increased cancer cell proliferation, tumor progression and invasiveness, making it a potential prognostic marker of some cancers. Selective inhibition of choline kinase also attenuates MAPK and PI3K/Akt signaling, which was associated with a decrease in the growth of cells[25].
As mentioned above, RA cultured FLS possess a unique tumor like behavior, and aggressive phenotype, such as cartilage invasiveness[2, 3]. Moreover, MAPK and PI3K/Akt are signal pathways that regulate synoviocyte function in RA such as metalloproteinase (MMP) expression and synoviocyte growth and survival, which may explains its aggressive phenotype[26, 27]. All together led us to do a targeted metabolomics assessment of choline metabolism in RA FLS to determine if its unique aggressive phenotype could be secondary to a higher ChoKa activity lever, similar to what it is observed in tumor cells, and to determine suitability as therapeutic target and biomarker of inflammation/joint damage. Our preliminary data demonstrates that ChoKa is highly expressed in RA synovial tissue, especially in the intimal lining and in cultured FLS, and its expression in FLS is regulated by inflammatory cytokines. MRS studies of choline-containing compounds in RA FLS showed an increase of PCho similar to what is observed in tumor cells. We used then a specific competitive choline kinase inhibitor, MN58b, to determine ChoK functions in vivo and in vitro. MN58b[28] (1 ,4-(4-4'-Bis-((4- (dimethylamine)pyridinium-l -yl) methyl}diphenyl) butane dibromide) exhibits selective inhibition of choline kinase, inhibits proliferation of cancer cells in vitro with an IC50 of 1 -1 OnM, and displays therapeutic activity against human tumor xenografts in vivo. We show here that ChoKa regulates key FLS functions that might contribute to cartilage destruction in RA. Initial studies also show a novel role for choline kinase in vivo. We hypothesized that ChoKaDis a key regulator ofD FLS functions that contribute to aggressive behavior and joint destruction in RA and that selective ChoKa inhibition in RA will be disease modifying by directly modulating synoviocyte mediated cartilage destruction and that choline-containing compounds detected by MRS might be a non-invasive biomarker of inflammation/joint damage and response to therapy.
Despite the successes in the last two decades, treatment of RA remains an unmet medical need. There is an ongoing effort to develop more effective, safer and less costly therapies to achieve free remission. Furthermore, to the best of Applicant's knowledge, the currently available disease modifying drugs do not directly target synoviocytes, cells that play a major role in RA pathogenesis. New rationally designed disease modifying agents that directly target key signaling proteins in FLS are needed to replace or complement current therapies. Metabolomics is an emerging field of biomedical research can offer a better understanding of mechanisms underlying disease, and help to develop new strategies for treatment. Variations in metabolite concentrations can also serve as diagnostic or prognostic biomarkers. Unique metabolomic profiles have been identified in the serum of patients with several diseases. Little is known about metabolomics changes in RA that could help in treatment decision-making. Of note, University of California, San Diego has been recently awarded by the NIH Common Fund and will play a central role in a new program from the National Institutes of Health (NIH) to accelerate metabolomics. Using a targeted metabolomics approach, we have identified a potential target in synoviocytes, ChoKa, that has been related to cancer cell proliferation, tumor progression and invasiveness. A highly selective choline kinase inhibitor could potentially block synovial hyperplasia and cartilage invasion by synoviocytes. We will explore the unexpected choline kinase function and signaling in synoviocytes biology and in RA pathogenesis. We will use selective small molecule inhibitor to test our hypothesis in vitro cell culture and in preclinical models of arthritis. We will also explore choline metabolites as biomarkers. If successful, these studies could pave the way for new treatments for patients with RA.
Our data demonstrates that ChoKa is highly expressed in RA synovial tissue, especially in the intimal lining and in cultured FLS, and its expression in FLS is regulated by inflammatory cytokines. MRS studies of choline-containing compounds in RA FLS showed an increase of PCho similar to what is observed in tumor cells. We used then a specific competitive choline kinase inhibitor, MN58b, to determine ChoKa functions in vivo and in vitro. MN58b (1.4-(4-4'-Bis-((4- (dimethylamine)pyridinium-l -yl) methyl}diphenyl) butane dibromide) exhibits selective inhibition of ChoKa, inhibits proliferation of cancer cells in vitro with an IC50 of 1 -10 μΜ, and displays therapeutic activity against human tumor xenografts in vivo. Our data shows that ChoKa regulates key FLS functions that might contribute to cartilage destruction in RA. Initial studies also show that ChoKa inhibition in a model of inflammatory arthritis decrease paw swelling and histological score.
The data is using the specific competitive ChoKa inhibitor, MN58b. In some embodiments, any of a series of other compounds could also be included since findings herein imply cause of existing ChoKa inhibitorAccording to some embodiments herein, a ChoKa inhibitor, for example MN58b, also comprises a pharmaceutically acceptable excipient. Therefore, some embodiments are also directed to a pharmaceutical composition as disclosed above, wherein the pharmaceutical composition additionally comprises a pharmaceutically acceptable excipient.ln general all excipients known by a person skilled in the art are suitable within embodiments herein. Examples of such excipients are calcium carbonate, kaolin, sodium hydrogen carbonate, lactose, D-mannitol, starches, crystalline cellulose, talc, granulated sugar, porous substances, etc.A ChoKa inhibitor, for example MN58b, may be used as bulk itself but usually be formulated into pharmaceutical preparations together with a suitable amount of "carrier for pharmaceutical preparation" according to ordinary methods.
Thus, compositions and methods described herein may also contain additionally diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials well known in the art.
Further on, "carriers for pharmaceutical preparation" comprises, for example, excipients as defined herein, binders, e.g., dextrin, gums, a-starch, gelatin, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, pullulan, etc., thickening agents, e.g., natural gums, cellulose derivatives, acrylic acid derivatives, etc., disintegrators, e.g., carboxy- methyl cellulose, croscarmellose sodium, crospovidone, low-substitution hydroxypropyl cellulose, partial a-starch, etc., solvents, e.g., water for injections, alcohol, propylene glycol, macrogol, sesame oil, corn oil, etc., dispersants, e.g., Tween 80, HCO60, polyethylene glycol, carboxymethyl cellulose, sodium alginate, etc., solubilizers, e.g., poly- ethylene glycol, propylene glycol, D-mannitol, benzyl benzoate, ethanol, trisami- nomethane, triethanolamine, sodium carbonate, sodium citrate, etc., suspending agents, e.g., stearyl triethanolamine, sodium lauryl sulfate, benzalkonium chloride, polyvinyl alcohol, polyvinylpyrrolidone, hydroxyethyl cellulose, etc., pain-reducing agents, e.g., benzyl alcohol, etc., isotonizing agents, e.g., sodium chloride, glycerin, etc., buffers, e.g., phosphates, acetates, carbonates, citrates, etc., lubricants, e.g., magnesium stearate, calcium stearate, talc, starch, sodium benzoate, etc., colorants, e.g., tar pigments, caramel, iron sesquioxide, titanium oxide, riboflavins, etc., tasting agent, e.g., sweeteners, flavors, etc., stabilizers, e.g., sodium sulfite, ascorbic acid, etc., preservatives, e.g., parabens, sorbic acid, etc., and the like.
Preferred methods of administration of the pharmaceutical compositions described above include oral and parenteral, e.g., i.v. infusion, i.v. bolus and i.m. injection formulated so that a unit dosage comprises a therapeutically effective amount of each active component or some submultiples thereof. The compounds may be employed in powder or crystalline form, in liquid solution, or in suspension. Theses compounds may be formulated by any method well known in the art and may be prepared for administration by any route, including, without limitation, parenteral, oral, sublingual, by inhalation spray, transdermal, topical, intranasal, intra- tracheal, intrarectal via ophthalmic solution or ointment, rectally, nasally, buccally, vaginally or via implanted reservoir. The term parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
Pharmaceutical compositions for injection, a preferred route of delivery according to some embodiments herein, may be prepared in unit dosage form in ampules, or in multidose containers. The composition will generally be sterile and pyrogen- free, when intended for delivery by injection into the subject. The injectable compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain various formulating agents. Alternatively, the active ingredient may be in powder (lyophilized or non-lyophilized) form for reconstitution at the time of delivery with a suitable vehicle, such as sterile water. Carriers suitable for an injectable pharmaceutical composition according to some embodiments are typically comprised sterile water, saline or another injectable liquid, e.g., peanut oil for intramuscular injections. Also, various buffering agents, preservatives and the like can be included. The pharmaceutical composition according to some embodiments may also be administered parenterally in a sterile medium. Depending on the vehicle and concentration used, the drug can either be suspended or dissolved in the vehicle. Advantageously, adjuvants such as local anaesthetic, preservative and buffering agents can be dissolved in the vehicle. The proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. It is also preferred to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatine. Intra-venous infusion is another possible route of administration for the compounds used according to some embodiments herein. Orally administrable pharmaceutical compositions according to some embodiments herein may be in the form of tablets, capsules, powders, granules, lozenges, liquid or gel preparations, such as oral, topical, or sterile parenteral solutions or suspensions. The oral compositions may utilize carriers such as conventional formulating agents, and may include sustained release properties as well as rapid delivery forms. Such compositions and preparations should contain at least 0.1 % of active compounds. The percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 60% of the weight of a given unit dosage form. The amount of active compound in such therapeutically useful compositions is such that an effective dosage level will be obtained.
Tablets and capsules for oral administration may be in unit dose presentation form, and may also contain conventional excipients such as binding agents, for example syrup, acacia, gelatine, sorbitol, tragacanth, or polyvinylpyrrolidone; fillers for example lac- tose, sugar, maize-starch, calcium phosphate, sorbitol or glycine; tabletting lubricant, for example magnesium stearate, talc, polyethylene glycol or silica; disintegrates for example potato starch, or acceptable wetting agents such as sodium lauryl sulphate. The tablets may be coated according to methods well known to a person skilled in the art. Oral liquid preparations may be in the form of, for example, aqueous or oily sus- pensions, solutions, emulsions, syrups or elixirs, or may be presented as a dry product for reconstitution with water or other suitable vehicle before use. Such liquid preparations may contain conventional additives such as suspending agents, for example sorbitol, syrup, methyl cellulose, glucose syrup, gelatine hydrogenated edible fats; emulsifying agents, for example lecithin, sorbitan monooleate, or acacia; non-aqueous vehicles which may include edible oils, for example almond oil, fractionated coconut oil, oily esters such as glycerine, propylene glycol, or ethyl alcohol; preservatives, for example methyl or propyl p-hydroxybenzoate or sorbic acid, and if desired conventional flavouring or colouring agents. Pharmaceutical compositions according to some embodiments herein may also be prepared in suitable forms for absorption through the mucous membranes of the nose and throat or bronchial tissues and may conveniently take the form of powder or liquid sprays or inhalants, lozenges, throat paints, etc. For medication of the eyes or ears, the preparations may be presented as individual capsules, in liquid or semi-solid form, or may be used as drops, etc.
Suitable subjects for the administration of the formulation of some embodiments herein include mammals, primates, humans, and other animals. Typically the animal subject is a mammal, generally a domesticated farm mammal, e.g. horse, pig, cow, sheep, goat etc., or a companion animal, e.g. cat, dog etc.. In vitro antibacterial activity is predictive of in vivo activity when the compositions are administered to a mammal infected with a susceptible bacterial organism. EXAMPLES
In the following examples, unless stated otherwise, the following methods were used: ChoKa expression in RA synovium and RA FLS was evaluated by immunohistochemistry (IHC) and Western blot (WB), respectively. Osteoarthritis (OA) samples were used as controls in some experiments. The metabolic profile of FLS cells was determined by 1H-MRS under conditions of ChoKa inhibition. FLS function using the ChoKa inhibitor MN58b (IC50= 4.2 μΜ) in medium and platelet derived growth factor (PDGF) stimulated cells was evaluated by measuring 1 ) migration into a cleared area in cultured FLS monolayers (scratch assay); 2) proliferation using an MTT assay; and 3) protein expression by WB. Cell survival was determined in H2O2 treated cells by phase contrast light microscopy. For arthritis experiments, mice were injected with K/BxN sera on day 0. MN58b (3mg/kg) was injected daily i.p. beginning on day 0 or day 4 after serum administration. Clinical arthritis scores were serially assessed. Joints were evaluated for inflammation and joint damage using histology and a semiquantitative scoring system.
EXAMPLE 1
ChoKa mRNA and protein were highly expressed in RA synovial tissue and in cultured FLS. Its expression in FLS was increased 2-3-fold after tumor necrosis factor (TNF) and PDGF stimulation, respectively with peak expression within 48 hours. Metabolomic studies of choline-containing compounds in cultured FLS extracts showed increased levels of phosphocholine in RA FLS, confirming activation of this pathway. ChoKa regulates key FLS functions that might contribute to cartilage destruction in RA. For example, ChoKa inhibition with MN58b (5 μΜ) reduced proliferation by 79±3.2% and migration by 54±15% (p<0.05). ChoKa inhibition also markedly increased H2O2-induced apoptosis in FLS. Additionally, Akt phosphorylation in response to PDGF as determined by WB was blocked by ChoKa inhibition Finally, ChoKa inhibition significantly decreased arthritis in pre-treatment protocols (day 0) as well as in established disease (day 4). For example, day 8 scores were 12±1 .6 and 7±2.6 (P<0.05) for vehicle and MN58b-treated mice, respectively, when initiated on day 0; and were 6.6±0.9 and 1 .6±2.5 (PO.05) for PBS and MN58b-treated mice when initiated on day 4. Joint histology scores for vehicle and MN58b-treated mice for inflammation were 3.2±0.5 and 1 .25±1 (pO.05), bone erosion scores were 2.7±0.5 and 0.25±0.5 (p<0.05), and cartilage damage scores were 1 .5+1 and 1 .6±0.5 (p<0.05) respectively. Careful dissection of the metabolic profile in RA FLS suggests that choline metabolism is abnormal and is similar to transformed cells. Blocking this pathway with a selective ChoKa inhibitor suppressed inflammatory arthritis in mice as well as the aggressive behavior of cultured RA FLS, including cell migration and resistance to apoptosis. These data suggest that ChoKa inhibition could be an effective strategy for arthritis. EXAMPLE 2: CHOLINE KINASE-A EXPRESSION IN RA SYNOVIUM
Immunohistochemistry (IHC) was done to determine if ChoKa is expressed in RA synovium. As shown in the Fig. 1 , ChoKa is highly expressed in synovial tissue, it is primarily expressed in the intimal lining with scattered positive cells in the sublining, suggesting the expression of ChoKa in FLS. EXAMPLE 3: CHOLINE KINASE EXPRESSION AND REGULATION IN
CULTURED FLS
ChoKa protein is localized to the intimal lining cells, we evaluated its expression and regulation in cultured RA FLS derived from this region. Protein expression was confirmed with Western blot analysis. To determine whether ChoKa is regulated by inflammatory mediators implicated in RA, we stimulated RA FLS with TNF, IL-1 , PDGF, EGF or LPS for 48 hrs, and protein was assayed by WB. ChoKa increased following TNF, PDGF and EGF stimulation (Fig. 2) while LPS had no effect.
EXAMPLE 4: ACTIVATED CHOLINE METABOLITES ARE DETECTED IN RA
FLS
One-dimensional 1H NMR spectra of aqueous extracts revealed and activated choline profile in RA FLS (Fig. 3A). The relative areas of signal components due to individual PC metabolites (GPC, PCho and Cho) are more similar to tumor than non-tumor cells (Fig. 1 ), PCho becoming the predominant metabolite, which is further increased after PDGF stimulation (GPC:PC ratio <0.1 ). Fig. 3 shows 1H NMR spectra of normal IMR-90 fibroblast cell line, which shows a GPC/PC ratio around 1.
EXAMPLE 5: CHOLINE KINASE REGULATES P-AKT AND P-MAPK
ACTIVATION IN FLS
ChoKa in tumor cells can activate Akt and MAPK. To explore ChoKa function in FLS, we next studied activation of Akt and MAPK in response to growth factor PDGF, a potent activator of these pathways in various cell types. FLS were treated with ChoKa inhibitor or vehicle for 8h then stimulated with PDGF-BB 10 ng/ml or medium for 30 min. Western blot analysis was done for P-Akt (S473) and P-MAPK and total protein to quantify the inhibitory effects of the compound. As shown in Fig. 4, MN58b partially decreased P-Akt and P-ERK in FLS.
EXAMPLE 6: CHOLINE KINASE REGULATES RA FLS GROWTH
We also tested whether ChoKa inhibition interferes with cell growth in vitro in response to PDGF. FLS were cultured in presence of PDGF-BB (10 ng/ml) with MN58b (0.1 , 0.5, 1 and 5 uM) or vehicle for 7 days. Growth was measured on day 4 using MTT assay. As shown, the MN58b decreased cell proliferation dose dependency (Fig. 5). Data is presented as mean absorbance units+/-SEM normalized to Day 0. Treatment with MN58b alone did not affect viability when compared with media (not shown).
EXAMPLE 7: CHOKa REGULATES FLS MIGRATION
Because PDGF is a known chemotactic agent for mesenchymal cells, we evaluated whether ChoKa is required for FLS migration. FLS were grown to a confluent monolayer in a 6-well plates and wounded area was generated with 1 ml micropipette tip. Cells were cultured in low serum Med (1 % FBS) alone or with PDGF-BB 10 ng/ml +/- MN58b (1 μΜ) or vehicle (Fig. 6). Cell migration in response to PDGF was dramatically decreased in presence of CHoKa inhibitor MN58b at 1 μΜ. These results were also confirmed after siRNA knock down of ChoKa (not shown).
EXAMPLE 8: EFFECT OF CHOKa INHIBITOR IN KXB/N MODEL OF
ARTHRITIS
To determine whether choline kinase contributes to arthritis we tested efficacy, tolerability and safety of a choline kinase inhibitor, MN58b, in passive K/BxN animals. WT mice were injected with 150ul serum from adult K/BxN mice on day 0. MN58b significantly decreased paw swelling in this model (p<0.01 for vehicle compared with daily 3 mg/kg) (Fig. 7A). It also decreased histological score (Fig. 7B) and IL-1 b and MMP3 mRNA expression (Fig 7 C and D). These results demonstrate that MN58b is effective in a rodent model of arthritis.
To evaluate the safety of ChoKa inhibition, we treated WT mice with 1.5mg/kg and 3mg/kg of MN58b for 7 days, and tested liver and kidney histology that did not show any significant change (no shown). We also realized cleaved caspase-3 staining that did not show any apoptotic damage in the tissues studied (no shown). Biochemistry and hematologic studies did not show an increase of liver enzymes, worsening of kidney function or anemia. EXAMPLE 9: LACK OF EFFECT OF CHOKa INHIBITOR IN ADAPTIVE IMMUNE
RESPONSE
To test adaptive response, we performed preliminary studies in an antigen- induced arthritis model. Mice were immunized with an intradermal injection on day 0 with methylated BSA (mBSA) in complete Freund's adjuvant. 2 weeks later mice were bled and antibodies against mBSA were analyzed by ELISA. MN58b treatment (daily 3mg/kg) did not change the titer of antibodies against mBSA suggesting that the inhibitor was acting more in the innate than in the adaptive response compartment. lgG1 antibodies levels were 0.54±0.1 and 0.53±0.12 (p=ns) and lgG2b were 0.18±0.04 and 0.17±0.06 (p=ns) for PBS and ChoKa inhibitor-treated mice.
EXAMPLE 10: STATISTICAL ANALYSIS
In vivo studies can be analyzed by ANOVA. For in vitro studies, paired or unpaired Student's t-test will be used when the data have a normal distribution. In other cases, non-parametric tests such as the Wilcoxon signed rank test will be used. Pearson's coefficient will be used for correlation studies. A p value <0.05 will be considered significant. Power analysis shows that 8 mice per group gives a 90% chance of detecting 30% differences in mouse arthritis studies. Statistical support will be provided by the UCSD Biostatistics Core (ctri.ucsd.edu). EXAMPLE 11 : DETERMINATION OF PATTERNS OF CHOLINE-CONTAINING
COMPOUNDS IN RA FLS BY MRS
Preliminary MRS studies in RA FLS shows increased PCho ratios similar to tumor cells. We hypothesize that this switch in choline metabolites is secondary to increased ChoKa activity in RA, which could explain its aggressive phenotype. We will determine the choline kinase expression and functional state of choline kinase from RA, osteoarthritis (OA) and normal FLS by MRS. We will also use tumor fibroblast cell lines derived from pigmented villonodular synovitis and sarcoma that are currently in our repository. We will then evaluate how choline metabolites are modified by cytokines. Although, rheumatoid synoviocytes display certain unique features that are reminiscent of transformed cells, it is not clear whether these features are inherent in RA FLS (transformed aggressors) or are somehow imprinted due to exposure to cytokines in the rheumatoid milieu in vivo (passive responders). Studies using normal and OA FLS after cytokine stimulation will determine if cytokines mimic the change in choline metabolism in RA FLS.
Preparation of synovium and synoviocytes. Synovium and FLS will be obtained from patients undergoing total joint replacement or synovectomy who meet the 1987 revised American College of Rheumatology criteria for seropositive RA or patients with OA as previously described[29]. RA patients will discontinue methotrexate for at least 1 month prior to surgery to minimize the influence of methotrexate on folate metabolism and methyl donors[30]. For FLS lines, tissue is enzymatically dispersed and cells allowed to adhere overnight. Nonadherent cells are washed off, and the adherent FLS are grown in DMEM containing 10% FCS. FLS are used from passage 3 through 8 during which time they are a homogeneous population of cells (<1 % CD1 1 b positive, <1 % phagocytic, and <1 % FcR II and FcR III receptor positive)[31].
Choline metabolites profiling using 1H-MRS: 1H-MRS platform has become an established tool for the comprehensive analysis of the metabolome in biological samples for both polar and lipophilic metabolites. To confirm that RA FLS have a tumor-like metabolite pattern, cells from normal, RA and OA FLS (5 cell lines each), as well as tumor fibroblasts (e.g., PVNS) and dermal fibroblasts will be isolated and prepared for the 1H-MRS analysis as described[21 , 32]. Acquisition of the 1H-MRS metabolic profiles of both the polar and apolar fractions will be performed using a Bruker Avance 700 MHz NMR spectrometer equipped with high throughput robotics. One and two-dimensional NMR spectra will be acquired for improved metabolite identification and quantification. We will look at the 3.20-3.24 ppm region where the PCho, GPC and Cho have been reported. We will analyze the metabolite concentrations of cellular components of fibroblast cell lines. Metabolite quantification will be expressed as nmoles and normalized to the number of extracted cells. Ratio of choline phospholipid metabolites (PCho/total choline and GPC/PCho) will be analyzed.
Is the RA tumor-like choline metabolite profile secondary to ChoKa in RA FLS? Elevated PCho levels are usually attributed to an increased ChoK activity. We will evaluate the functional state of choline kinase by 1H-MRS after incubation of RA, OA, and normal (NL) FLS cells with ChoKa inhibitor MN58b (1 nM-5 uM) to show that the profile is due to a ChoKa. The optimum concentrations of MN58b will be used in Aim 2.
Is choline kinase expression greater in RA FLS and synovium? Expression will be determined by qPCR and WB. We will also determine the expression of ChoKa in OA, RA and normal FLS (5 cell lines each) before and after stimulation with cytokines (IL-1 at 2ng/ml, TNF or PDGF BB at 10ng/ml) by Western blot (WB) and qPCR. RA, OA and normal synovium will be immunostained (5 tissues each) to localize ChoKa. Digital image analysis will determine 1 ) whether intimal lining expression is greater than sublining expression; and 2) whether these proteins are more abundant in RA compared with OA or normal tissues.
Is the RA choline metabolite profile modified by cytokines? To determine if cytokine stimulation further increases ChoKa expression in RA FLS and if cytokine stimulation changes choline metabolism in OA and normal FLS, we will incubate normal, RA and OA FLS with PDGF BB and TNF for 2 to 48 hr as we have shown that these cytokines increase ChoKa expression in RA FLS (see Preliminary Data). We will perform dose response and kinetics to determine optimum time point for ChoKa induction and use these culture conditions to determine choline metabolites by MRS.
Anticipated results: As RA cultured FLS possess a unique tumor like behavior that differentiates them to other fibroblast cell lines, and based on our preliminary results, we expect a choline metabolites profile characterized by high PCho/total choline and low GPC/PCho ratios in RA FLS, similar to the pattern from tumor fibroblasts. In contrast, normal and OA FLS will probably show a different profile characterized by low PCho/total choline and similar GPC/PCho as found in normal fibroblasts. In that case, analysis of other metabolites such as phosphatidylcholine, lysophosphatidylcholine, phosphatidylethanomaline can be addressed to further characterize choline metabolism. As choline phosphorylation is mostly due to ChoKa activity, incubation of the RA FLS with the ChoKa inhibitor should reverse the activated choline metabolism profile. We will compare the expression of ChoKa in both OA and RA synovium and FLS to determine if the FLS profile is mimicked by tissue. Because the RA FLS phenotype is restored by cytokines in vitro, we expect that choline metabolism will also be modified.
EXAMPLE 12: DETERMINING THE EFFECT OF CHOLINE KINASE INHIBITION ON GENE EXPRESSION, SIGNALING AND FUNCTION
OF FLS
Based on the observation of the activated choline metabolism in FLS and its expression in these cells, and with the hypothesis that increased ChoKa activity might contribute to the unique tumor-like behavior or RA cultured FLS, we will explore how ChoKa modulates RA FLS growth, migration and invasion. Our preliminary data show that ChoKa regulates Akt/MAPK phosphorylation, growth and migration of RA FLS suggesting that ChoKa inhibition might be chondroprotective in RA. Cells will be pre-treated with the ChoKa inhibitor at the concentrations determined in Specific Aim 1. Key findings seen with chemical inhibition will be confirmed with siRNA knock down.
Does ChoKa regulate PDGF-mediated FLS activation? Our preliminary data show that ChoKa regulates P-Akt and P-ERK after PDGF stimulation. To examine the role of ChoKa in PDGF function, RA FLS will be stimulated with PDGF for 5 to 30 min in the presence or absence of the MN58b or after ChoKa knockdown. P-Akt, P-JNK, P-ERK and P-p38 will be determined by WB. If confirmed, we will also determine whether ChoKa blockade or deficiency affects other PDGF-mediated functions, such as MMP1 and MMP3 expression by qPCR.
Anticipated results and potential pitfalls: ChoKa will probably decrease P-protein and MMP expression based on our preliminary studies. Cytokines, especially TNF and IL-1 , enhance the destructive properties of RA FLS and their effects will probably be potentiated by growth factors like PDGF.
Does ChoKa blockade or deficiency alter synoviocyte growth through an effect on proliferation and apoptosis? Our data predict that ChoKa inhibition could have chondroprotective effects in RA. To explore this possibility, we will determine if ChoKa regulate proliferation and survival of cultured RA FLS. Human FLS will be cultured with MN58b compound of after ChoKa knockdown
• Apoptosis— Human and murine FLS will be treated with anti-Fas antibody or 100 uM hydrogen peroxide to induce apoptosis. Cells will be evaluated from 4 to 24 hr later using trypan blue dye exclusion. A histone release assay performed by ELISA will be performed to confirm the results.
• Proliferation— Cell proliferation will be evaluated using cultured FLS (human and murine) and stimulating with medium or PDGF (10ng/ml) in the presence of ChoKa inhibitor. Tritiated thymidine will be used to quantify DNA synthesis after 2-7 days.
Anticipated results: We expect that apoptosis will be increased and proliferation will be decreased in cells after blockade or deficiency of ChoKa activity because this kinase contributes to Akt phosphorylation in FLS that is known to play a role in these functions.
Does ChoKa regulate synoviocyte migration and invasion in vitro? Cell migration is an essential process for synoviocytes to reach, invade and destroy articular cartilage in RA. In addition, it was recently proposed that FLS migrating through the blood stream might be responsible for progression of arthritis and that this migratory behavior is unique for RA but not OA FLS. Interfering with key regulators of these functions of synoviocytes has the potential to reduce articular damage, progression of disease and improve outcome in RA. Based on our preliminary results that ChoKaDinhibition decreases synoviocytes migration and MMP expression, we hypothesized that ChoKa is a majorD regulator of FLS invasion. RA FLS will be cultured with MN58b compound of after ChoKa knockdown
• In vitro migration using a scratch assay— .FLS will be grown into a monolayer and an area will be cleared using micropipette. Cells will be pre- incubated with ChoKa inhibitor or ChoKa knockdown. Medium or PDGF (10ng/ml) will then be added to the cultures and migration of cells into the cleared area quantified after 12, 24, and 48 hr by image analysis.
• Invasion using Cytoselect assay— Because invasion is feature of RA synoviocytes, we will test the effect of ChoKa inhibition on FLS invasion using Matrigel coated transwell inserts (Cell Biolabs, San Diego, CA). FLS will be pre-treated with ChoKa inhibitor or vehicle for 8 hr. The FLS are added to the upper chamber and then medium with or without PDGF (25 ng/ml) is added to lower chamber. Invasion is quantified after 24 hr by counting cells on the lower surface of the membrane.
Anticipated results: Our preliminary data suggest that the ChoKa contributes to migration, although we still need to quantify the effects, determine the effect on invasion, and perhaps evaluate other chemoattractants. Given the role of ChoKa in cell movement, the ChoKa inhibitor will probably decrease cell invasion into matrix. While Matrigel, which is mainly comprised of laminin, is an imperfect matrix, it is commonly used as a substrate for invasion assays. If an effect is seen, then we will consider evaluating the effect of ChoKa inhibition on MMP and integrin gene expression.
Does ChoKa regulate migration and MMP expression through activation of Akt/MAPK pathway respectively? Because ChoKa regulates MAPK and Akt signaling after PDGF stimulation in RA FLS, and these signaling have been involved in MMP and migration respectively, we will determine the effect of Akt and MAPK siRNA on those functions together with ChoKa inhibition. We hypothesize that if the effect of ChoKa is through these two pathways, ChoKa inhibitor will not add any further effect after blockade of MAPK and Akt by siRNAs or chemical inhibitors. MAPK (either JNK, p38 or ERK siRNA) will be chosen according to the effect of ChoKa inhibition on those phospho-protein after PDGF stimulation) and Akt will be knocked-down by siRNA in RA FLS and at day 5, MMP expression and migration assays after PDGF stimulation will be realized and assessed as detailed above after chemical ChoKa inhibition.
Anticipated results: Based on our preliminary results that ChoKa inhibition decreases P-Akt and p-MAPK in FLS, we expect that addition of ChoKa inhibition will not add any additional effect in both migration and MMP expression after akt and MAPK siRNA downregulation respectively. We could also consider using chemical MAPK inhibitors, like SP600125, SB203580, and PD98059 if multiple isoform of each MAPK are involved.
EXAMPLE 13: DETERMING EFFECTS OF CHOLINEKINASE INHIBITION IN
K/BXN ARTHRITIS MODEL
The lack of effect of ChoKa inhibitor in adaptive immune response and its effect on FLS, led us evaluate the role of this kinase in the K/BxN arthritis model[33]. Although arthritis models do not represent RA in mouse, in vivo studies allow investigation of pathogenic mechanism in RA. The K/BxN passive serum transfer model is useful because it is FLS dependent and requires only innate immunity. We will use the choline kinase inhibitor MN58b to evaluate choline kinase blockade in vivo. We hypothesized that ChoKa inhibition will ameliorate inflammation and cartilage damage. Preliminary toxicology studies did not show any evident toxicity in major organs such as liver or kidney in mice on this specific choline kinase inhibitor. We will also determine if choline pathway correlates with disease activity in this animal model. What is the effect of chemical inhibition of ChoKa in K/BxN arthritis? Initial experiments will determine optimal doses and efficacy of MN58b compound in vivo. WT mice will be treated daily with several doses of ChoKa inhibitor (1 -3 mg/kg) or vehicle starting on day 0 parallel with K/BxN serum transfer. To minimize potential toxicity, we will also test other dosing options such as administering the drug every day during the first 5 days of arthritis, or giving the drug on alternate days. To examine efficacy in established disease, we will start therapy on day 5. To induce arthritis, WT mice will be injected with 150 ul serum from adult K/BxN mice on day 0. Clinical scoring will be done daily and experiments will be terminated on day 10. The endpoints for our analysis: 1 ) Clinical scores and ankle diameter; 2) Ankle histology, for synovial hyperplasia, cartilage erosion, cartilage proteoglycan, inflammation and bone destruction 3) IHC for infiltrating cell types 4) Inflammatory gene expression (mRNA and protein analysis) and 5) Alteration in synovial signaling in the arthritic joints.
· Synovial proliferation and cartilage erosion. Hind paws of mice are harvested for paraffin sections and are stained with H&E, safranin O-fast green for proteoglycan content. We will determine in situ FLS proliferation by performing IHC for PCNA and Ki67, nuclear antigens used as an index of cell proliferation in tissue. Results will be quantified by image analysis. · Synovial mediator expression— Ankle extracts will be assayed by tissue extract ELISA and qPCR of pro-inflammatory cytokines (TNF and IL-6), prototypical Th1 , Th2, and Th17 cytokines (IFNy, IL-4 and IL-17A), and proteases (MMP3, MMP13) as possible mediators regulated by the Akt/MAPK pathway.
· Synovial signaling— WB will be performed to determine P-Akt, P-JNK, P-p38 and P-ERK in the joints.
Anticipated results: As we shown in our preliminary results, the compound is tolerated by mice and can be used in the animal model of arthritis. We expect that chemical inhibition of ChoKa will ameliorate arthritis. Based on our in vitro studies in FLS, we expect to see decreased synovial lining hyperplasia and significant reduction in cartilage erosion in the mutant mice. There will be decreased synovial inflammation due to possible role of ChoKa on macrophages, and decreased inflammatory mediator production by resident synoviocytes. These benefits will correlate with decreased P-Akt and P-MAPK. These results would make us move forward and test the inhibitor in a antigen-induced arthritis and collagen-induced arthritis model. To confirm the results, we will target ChoKa using anti-sense oligonucleotides. We recently used this technique to demonstrate the role of the kinase MKK7 in passive K/BxN arthritis[34]. Unfortunately, CHKA-deficient mice is lethal and there is not CHKAF F mice available in order to study which cell type confers protection in arthritis. One option is to use the choline inhibitor on cadherin 1 1 -deficient mice[35] to determine if ChoKa inhibition is additive with cadherin deficiency.
Toxicology studies: Although the ChoKa inhibitor is well tolerated by mice, we would like to extend our preliminary toxicology studies. We will treat the mice with several doses of MN58b inhibitor (1 -3mg/kg) for one week (8 mice/group) and we will realize histology of major organs such as liver, kidney, spleen and brain. We will also get basic hematology and biochemistry studies from blood and serum respectively. Apoptotic cells will be determined by cleaved-caspase 3 staining by IHC.
Anticipated results: MN58b inhibitor is well tolerated by mice at the doses tested, and preliminary data already shows no changes in histology of liver and kidney. Do the choline metabolites correlate with disease activity in K/BxN arthritis model? We will analyze choline metabolites by 1H-MRS in joint extracts, peripheral blood cells and serum from WT mice at different time points after K/BxN serum injection (day 2,4,6,8 and 10), to determine when we can better detect choline metabolites concentrations and ratio between levels of choline phospholipid metabolites (PCho/total choline and GPC/PCho). We will then inject K/BxN serum to 10 WT mice that will be sacrificed at that time point. We will correlate choline metabolites with clinical and histological score, IL-6, IL-1 , TNF, MMP3, and MMP13 expression by qPCR from joints, and cytokines assayed in serum by Luminex multiplex technology. PCho and GPC are typically intracellular metabolites but other metabolites such as phosphatidylcholines can be detected extracellularly. We will also determine choline metabolites in joint extracts at the same time point after MN58b administration to prove the effect of the compound.
Anticipated results: We expect to see decreased PCho/total choline ratio in joints after MN58b administration. As our preliminary results using the inhibitor in vivo decreases both inflammation and joint damage, we expect to see correlation of choline metabolites ratio with disease activity and mentioned mediators of inflammation and joint damage.
EXAMPLE 14: DETERMINING SUITABILITY OF CHOLINE METABOLITES AS
BIOMARKER OF INFLAMMATION
Biomarkers associated with clinical response might not be the same biomarkers that predict risk of further joint damage. MRS is a commonly used analytical method to analyze the metabolome of body fluids such as urine and blood serum[36]. Recent studies demonstrate the applicability of NMR-based metabolomics using serum samples for the diagnosis and prognosis[37]. In cancer, increased levels of these compounds provide a non-invasive biomarker of transformation, staging and response to therapy disease. However, few studies have addressed metabolic changes in RA and little information of this metabolic pathway has been addressed in inflammation. We will explore the hypothesis that choline metabolites can be detected in different kind of samples from RA patients, and that correlates with clinical response. We will take advantage of samples that we already have in our department to explore the relationship between choline metabolites and clinical response.
Are choline metabolites present in serum, peripheral blood and synovial tissue of RA patients? Our laboratory has developed a biological sample repository, which include thousands of blood, serum, and synovial tissue samples from patients with a variety of forms of arthritis. We will first determine choline metabolic profiles by 1H MRS of peripheral blood, serum and synovial tissue from RA and OA patients and normal samples (10 samples each). We will determine which metabolites can be detected in those samples and which profile is normal (present in samples from healthy controls) and which one is disease specific (OA vs RA). PCho and GPC and other metabolites such as phosphatidylcholine, lysophosphatidylcholine, phosphatidylethanomaline are typically intracellular metabolites. However, some of these metabolites such as phosphatidylcholines can be detected in serum.
To determine the relationship between the choline metabolites pattern and clinical response to treatment in RA patients. Our group recently conducted a study using the anti-CD20 monoclonal antibody rituximab in patients with active rheumatoid arthritis. In this study, serum, blood and synovial tissue samples were obtained before and following treatment, and detailed clinical information collected (i.e. to define whether or not patients responded to treatment) [38]. Choline- containing compounds will be assayed in serum and synovial tissue before and after treatment. The relationship between choline kinase metabolites in synovial tissue and blood and clinical responses as well as changes in histologic scores and gene expression will be determined. Statistical Analyses: Comparison of pre- treatment to post-treatment values was performed using Wilcoxon's signed rank test for paired data and corrected for multiple comparisons. Associations between changes in various clinical and biomarker results and choline metabolites will be performed using Spearman's rank correlation coefficient.
List of References
All references discussed herein, including the references below, are incorporated herein by reference in their entirety and are hereby made a part of this specification. To the extent publications and patents or patent applications incorporated by reference contradict the disclosure contained in the specification, the specification is intended to supersede and/or take precedence over any such contradictory material.
1 . Firestein, G.S., Evolving concepts of rheumatoid arthritis. Nature, 2003.
423(6937): p. 356-61 .
2. Bartok, B. and G.S. Firestein, Fibroblast-like synoviocytes: key effector cells in rheumatoid arthritis. Immunological reviews, 2010. 233(1 ): p. 233-55.
3. Lefevre, S., et al., Synovial fibroblasts spread rheumatoid arthritis to unaffected joints. Nature medicine, 2009. 15(12): p. 1414-20.
4. Sweeney, S.E. and G.S. Firestein, Primer: signal transduction in rheumatic disease-a clinician's guide. Nature clinical practice. Rheumatology, 2007.
3(1 1 ): p. 651 -60.
5. Weinblatt, M.E., et al., Treatment of rheumatoid arthritis with a Syk kinase inhibitor: a twelve-week, randomized, placebo-controlled trial. Arthritis and rheumatism, 2008. 58(1 1 ): p. 3309-18.
6. Kremer, J.M., et al., The safety and efficacy of a JAK inhibitor in patients with active rheumatoid arthritis: Results of a double-blind, placebo- controlled phase Ha trial of three dosage levels of CP-690, 550 versus placebo. Arthritis and rheumatism, 2009. 60(7): p. 1895-905.
7. Eckhart, A.D., K. Beebe, and M. Milburn, Metabolomics as a key integrator for "omic" advancement of personalized medicine and future therapies.
Clinical and translational science, 2012. 5(3): p. 285-8.
8. Hugle, T., et al., Synovial fluid metabolomics in different forms of arthritis assessed by nuclear magnetic resonance spectroscopy. Clinical and experimental rheumatology, 2012. 30(2): p. 240-5. 9. Lauridsen, M.B., et al., 1H NMR spectroscopy-based interventional metabolic phenotyping: a cohort study of rheumatoid arthritis patients. Journal of proteome research, 2010. 9(9): p. 4545-53.
10. Madsen, R.K., et al., Diagnostic properties of metabolic perturbations in rheumatoid arthritis. Arthritis research & therapy, 201 1 . 13(1 ): p. R19.
1 1 . Seeger, K., Metabolic changes in autoimmune diseases. Current drug discovery technologies, 2009. 6(4): p. 256-61 .
12. Weljie, A.M., et al., An inflammatory arthritis-associated metabolite biomarker pattern revealed by 1H NMR spectroscopy. Journal of proteome research, 2007. 6(9): p. 3456-64.
13. DeBerardinis, R.J., et al., The biology of cancer: Metabolic reprogramming fuels cell growth and proliferation. Cell Metabolism, 2008. 7(1 ): p. 1 1 -20.
14. Hsu, P.P. and D.M. Sabatini, Cancer cell metabolism: Warburg and beyond.
Cell, 2008. 134(5): p. 703-707.
15. Casado-Vela, J., et al., Approaches for the study of cancer: towards the integration of genomics, proteomics and metabolomics. Clinical & translational oncology : official publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico, 201 1 . 13(9): p. 617-28.
16. Glunde, K., C. Jie, and Z.M. Bhujwalla, Molecular causes of the aberrant choline phospholipid metabolism in breast cancer. Cancer Research, 2004. 64(12): p. 4270-6.
17. lorio, E., et al., Alterations of choline phospholipid metabolism in ovarian tumor progression. Cancer Research, 2005. 65(20): p. 9369-76.
18. Glunde, K., Z.M. Bhujwalla, and S.M. Ronen, Choline metabolism in malignant transformation. Nature reviews. Cancer, 201 1 . 11 (12): p. 835-48. 19. Beckonert, O., et al., Metabolic profiling, metabolomic and metabonomic procedures for NMR spectroscopy of urine, plasma, serum and tissue extracts. Nature Protocols, 2007. 2(1 1 ): p. 2692-2703. 20. Asiago, V.M., et al., Early Detection of Recurrent Breast Cancer Using Metabolite Profiling. Cancer Research, 2010. 70(21 ): p. 8309-8318.
21 . Tiziani, S., et al., Metabolomic profiling of drug responses in acute myeloid leukaemia cell lines. PLoS One, 2009. 4(1 ): p. e4251 .
22. Beloueche-Babari, M., et al., Metabolic assessment of the action of targeted cancer therapeutics using magnetic resonance spectroscopy. British journal of cancer, 2010. 102(1 ): p. 1 -7.
23. Glunde, K., et al., MRS and MRSI guidance in molecular medicine: targeting and monitoring of choline and glucose metabolism in cancer. NMR in biomedicine, 201 1 . 24(6): p. 673-90.
24. Gallego-Ortega, D., et al., Involvement of human choline kinase alpha and beta in carcinogenesis: a different role in lipid metabolism and biological functions. Advances in enzyme regulation, 201 1 . 51 (1 ): p. 183-94.
25. Yalcin, A., et al., Selective inhibition of choline kinase simultaneously attenuates MAPK and PI3K/AKT signaling. Oncogene, 2010. 29(1 ): p. 139-
49.
26. Han, Z., et al., c-Jun N-terminal kinase is required for metalloproteinase expression and joint destruction in inflammatory arthritis. The Journal of clinical investigation, 2001 . 108(1 ): p. 73-81 .
27. Bartok, B., et al., PI3 kinase delta is a key regulator of synoviocyte function in rheumatoid arthritis. The American journal of pathology, 2012. 180(5): p. 1906-16.
28. Rodriguez-Gonzalez, A., et al., Inhibition of choline kinase as a specific cytotoxic strategy in oncogene-transformed cells. Oncogene, 2003. 22(55): p. 8803-12.
29. Alvaro-Gracia, J.M., et al., Cytokines in chronic inflammatory arthritis. VI.
Analysis of the synovial cells involved in granulocyte-macrophage colony- stimulating factor production and gene expression in rheumatoid arthritis and its regulation by IL-1 and tumor necrosis factor-alpha. Journal of immunology, 1991 . 146(10): p. 3365-71 .
30. Schipper, R.G., et al., Polyamines and DNA methylation in childhood leukaemia. Biochemical Society transactions, 2007. 35(Pt 2): p. 331 -5.
31 . Alvaro-Gracia, J.M., N.J. Zvaifler, and G.S. Firestein, Cytokines in chronic inflammatory arthritis. V. Mutual antagonism between interferon-gamma and tumor necrosis factor-alpha on HLA-DR expression, proliferation, coiiagenase production, and granulocyte macrophage colony-stimulating factor production by rheumatoid arthritis synoviocytes. The Journal of clinical investigation, 1990. 86(6): p. 1790-8.
32. Tiziani, S., V. Lopes, and U.L. Gunther, Early Stage Diagnosis of Oral Cancer Using 1H NMR-Based Metabolomics. Neoplasia, 2009. 11 (3): p. 269-U69.
33. Kyburz, D. and M. Corr, The KRN mouse model of inflammatory arthritis.
Springer seminars in immunopathology, 2003. 25(1 ): p. 79-90.
34. Lee, S.I., et al., Regulation of inflammatory arthritis by the upstream kinase mitogen activated protein kinase kinase 7 in the c-Jun N-terminal kinase pathway. Arthritis research & therapy, 2012. 14(1 ): p. R38.
35. Lee, D.M., et al., Cadherin-11 in synovial lining formation and pathology in arthritis. Science, 2007. 315(5814): p. 1006-10.
36. Psychogios, N., et al., The Human Serum Metabolome. Plos One, 201 1 .
6(2).
37. Maclntyre, D.A., et al., Serum metabolome analysis by (1)H-NMR reveals differences between chronic lymphocytic leukaemia molecular subgroups. Leukemia, 2010. 24(4): p. 788-797.
38. Kavanaugh, A., et al., Assessment of rituximab's immunomodulatory synovial effects (ARISE trial). 1: clinical and synovial biomarker results. Annals of the rheumatic diseases, 2008. 67(3): p. 402-8. With respect to the use of substantially any plural and/or singular terms herein, those having skill in the art can translate from the plural to the singular and/or from the singular to the plural as is appropriate to the context and/or application. The various singular/plural permutations may be expressly set forth herein for sake of clarity.
It will be understood by those within the art that, in general, terms used herein, and especially in the appended claims (e.g., bodies of the appended claims) are generally intended as "open" terms (e.g., the term "including" should be interpreted as "including but not limited to," the term "having" should be interpreted as "having at least," the term "includes" should be interpreted as "includes but is not limited to," etc.). It will be further understood by those within the art that if a specific number of an introduced claim recitation is intended, such an intent will be explicitly recited in the claim, and in the absence of such recitation no such intent is present. For example, as an aid to understanding, the following appended claims may contain usage of the introductory phrases "at least one" and "one or more" to introduce claim recitations. However, the use of such phrases should not be construed to imply that the introduction of a claim recitation by the indefinite articles "a" or "an" limits any particular claim containing such introduced claim recitation to embodiments containing only one such recitation, even when the same claim includes the introductory phrases "one or more" or "at least one" and indefinite articles such as "a" or "an" (e.g., "a" and/or "an" should be interpreted to mean "at least one" or "one or more"); the same holds true for the use of definite articles used to introduce claim recitations. In addition, even if a specific number of an introduced claim recitation is explicitly recited, those skilled in the art will recognize that such recitation should be interpreted to mean at least the recited number (e.g., the bare recitation of "two recitations," without other modifiers, means at least two recitations, or two or more recitations). Furthermore, in those instances where a convention analogous to "at least one of A, B, and C, etc." is used, in general such a construction is intended in the sense one having skill in the art would understand the convention (e.g., " a system having at least one of A, B, and C" would include but not be limited to systems that have A alone, B alone, C alone, A and B together, A and C together, B and C together, and/or A, B, and C together, etc.). In those instances where a convention analogous to "at least one of A, B, or C, etc." is used, in general such a construction is intended in the sense one having skill in the art would understand the convention (e.g., " a system having at least one of A, B, or C" would include but not be limited to systems that have A alone, B alone, C alone, A and B together, A and C together, B and C together, and/or A, B, and C together, etc.). It will be further understood by those within the art that virtually any disjunctive word and/or phrase presenting two or more alternative terms, whether in the description, claims, or drawings, should be understood to contemplate the possibilities of including one of the terms, either of the terms, or both terms. For example, the phrase "A or B" will be understood to include the possibilities of "A" or "B" or "A and B."
In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group.
As will be understood by one skilled in the art, for any and all purposes, such as in terms of providing a written description, all ranges disclosed herein also encompass any and all possible sub-ranges and combinations of sub-ranges thereof. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as "up to," "at least," "greater than," "less than," and the like include the number recited and refer to ranges which can be subsequently broken down into sub-ranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. Thus, for example, a group having 1 -3 articles refers to groups having 1 , 2, or 3 articles. Similarly, a group having 1 -5 articles refers to groups having 1 , 2, 3, 4, or 5 articles, and so forth.
While various aspects and embodiments have been disclosed herein, other aspects and embodiments will be apparent to those skilled in the art. The various aspects and embodiments disclosed herein are for purposes of illustration and are not intended to be limiting, with the true scope and spirit being indicated by the following claims.
While the present invention has been described in some detail for purposes of clarity and understanding, one skilled in the art will appreciate that various changes in form and detail can be made without departing from the true scope of the invention.
The term "comprising" as used herein is synonymous with "including," "containing," or "characterized by," and is inclusive or open-ended and does not exclude additional, unrecited elements or method steps.
All numbers expressing quantities of ingredients, reaction conditions, and so forth used in the specification are to be understood as being modified in all instances by the term "about." Accordingly, unless indicated to the contrary, the numerical parameters set forth herein are approximations that may vary depending upon the desired properties sought to be obtained. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of any claims in any application claiming priority to the present application, each numerical parameter should be construed in light of the number of significant digits and ordinary rounding approaches.
The above description discloses several methods and materials of the present invention. This invention is susceptible to modifications in the methods and materials, as well as alterations in the fabrication methods and equipment. Such modifications will become apparent to those skilled in the art from a consideration of this disclosure or practice of the invention disclosed herein. Consequently, it is not intended that this invention be limited to the specific embodiments disclosed herein, but that it cover all modifications and alternatives coming within the true scope and spirit of the invention.
The foregoing description and Examples detail certain embodiments. It will be appreciated, however, that no matter how detailed the foregoing may appear in text, the invention may be practiced in many ways and the invention should be construed in accordance with the appended claims and any equivalents thereof.

Claims

1. A method of ameliorating rheumatoid arthritis in a subject in need of such amelioration, the method comprising administering a therapeutically effective dose of a choline kinase a (ChoKa) inhibitor to the subject.
2. The method of claim 1 , wherein the ChoKa inhibitor comprises a small molecule.
3. The method of claim 1 , wherein the ChoKa inhibitor comprises 1 ,4- (4-4'-Bis-((4-(dimethylamine)pyridinium-1 -yl) methyl)diphenyl) butane dibromide (MN58b).
4. The method of claim 1 , wherein the ChoKa inhibitor consists of MN58b.
5. The method of claim 4 or claim 5, wherein the dose of MN58b comprises about 1 mg per kg to about 10mg per kg.
6. The method of claim 4 or claim 5, wherein the dose of MN58b comprises about 3mg per kg.
7. The method of any of claims 1 to 6, wherein administration comprises injecting the ChoKa inhibitor.
8. The method of any of claims 1 to 7, wherein administration of the ChoKa inhibitor inhibits proliferation of at least one resident synovial fibroblast
(FLS) of the subject.
9. The method of any of claims 1 to 8, wherein administration of the ChoKa inhibitor inhibits migration of at least one FLS of the subject.
10. The method of any of claims 1 to 9, wherein administration of the ChoKa inhibitor reduces at least one of IL-1 b expression or MMP3 expression by the subject
1 1. The method of any of claims 1 to 10, further comprising detecting an amount of proliferation or migration of at least one FLS of the subject.
12. The method of any of claims 1 to 1 1 , further comprising detecting a presence or absence of at least one of Akt activation or MAPK activation in at least one FLS of the subject.
13. The method of any of claims 1 to 12, further comprising detecting an amount of expression of at least one of IL-1 b expression or MMP3 expression by the subject.
14. A pharmaceutical composition comprising a dosage of a choline kinase a (ChoKa) inhibitor suitable for treatment of rheumatoid arthritis.
15. The pharmaceutical composition of claim 14, wherein the dosage comprises about 1 mg per kg to about 10mg per kg of body mass of a subject in need of treatment of rheumatoid arthritis.
16. The pharmaceutical composition of claim 14, further comprising an antibody for treatment of rheumatoid arthritis.
17. The pharmaceutical composition of claim 16, wherein the antibody comprises an anti-CD20 antibody.
18. The pharmaceutical composition of claim 16, wherein the antibody comprises rituximab.
19. A method of determining a response to at least one clinical treatment of rheumatoid arthritis administered to a subject, the method comprising measuring the amount of at least one of ChoKa mRNA, ChoKa protein, or a choline metabolite in the subject.
20. The method of claim 19, wherein an amount of ChoKa mRNA or ChoKa protein is measured in an FLS of the subject.
21. The method of claim 19 or 20, wherein the at least one clinical treatment of rheumatoid arthritis comprises treatment with an anti-CD20 antibody.
22. The method of claim 21 , wherein the anti-CD20 antibody comprises rituximab.
23. The method any of claims 19 to 22, wherein the at least one clinical treatment of rheumatoid arthritis comprises treatment with a ChoKa inhibitor.
24. The method of claim 23, wherein the ChoKa inhibitor comprises MN58b.
25. A method of identifying a metabolic profile indicative of osteoarthritis or rheumatoid arthritis comprising comparing the level of at least one of GPC, ChoKa mRNA, ChoKa protein, or a choline metabolite in individuals with osteoarthritis or rheumatoid arthritis to individuals who do not have osteoarthritis or rheumatoid arthritis.
26. A method of diagnosing osteoarthritis or rheumatoid arthritis comprising determining whether a subject has level of at least one of GPC, ChoKa mRNA, ChoKa protein, or a choline metabolite indicative of osteoarthritis or rheumatoid arthritis.
27. The method of Claim 26, further comprising administering a treatment for osteoarthritis or rheumatoid arthritis if said subject has level of at least one of GPC, ChoKa mRNA, ChoKa protein, or a choline metabolite indicative of osteoarthritis or rheumatoid arthritis.
PCT/EP2014/068501 2013-08-30 2014-09-01 Compositions and methods for characterization and amelioration of rheumatoid arthritis WO2015028662A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361872585P 2013-08-30 2013-08-30
US61/872,585 2013-08-30

Publications (1)

Publication Number Publication Date
WO2015028662A1 true WO2015028662A1 (en) 2015-03-05

Family

ID=51454707

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2014/068501 WO2015028662A1 (en) 2013-08-30 2014-09-01 Compositions and methods for characterization and amelioration of rheumatoid arthritis

Country Status (1)

Country Link
WO (1) WO2015028662A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020057549A1 (en) 2018-09-18 2020-03-26 Beijing Innocare Pharma Tech Co., Ltd. Crystalline forms of 6- (1-acryloylpiperidin-4-yl) -2- (4-phenoxyphenyl) nicotinamide

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040202658A1 (en) * 2003-04-09 2004-10-14 Genentech, Inc. Therapy of autoimmune disease in a patient with an inadequate response to TNF-alpha inhibitor
US20070185170A1 (en) 2004-01-14 2007-08-09 Juan Carlos Lacal Sanjuan Derivatives of pyridine and quinoline
WO2007138143A2 (en) 2006-05-29 2007-12-06 Consejo Superior De Investigaciones Cientificas Alpha anticholine kinase monoclonal antibodies and their use in analytical techniques for the diagnosis of cancer and the preparation of medicinal products
EP1889920A1 (en) * 2005-04-13 2008-02-20 Consejo Superior De Investigaciones Cientificas In vitro cancer therapy compound identification method
US20100068302A1 (en) 2008-09-17 2010-03-18 Traslational Cancer Drugs Pharma, S.L. Methods and compositions for the treatment of cancer
US20110212994A1 (en) * 2009-06-26 2011-09-01 Brian Clem Small Molecule Choline Kinase Inhibitors, Screening Assays, and Methods for Safe and Effective Treatment of Neoplastic Disorders

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040202658A1 (en) * 2003-04-09 2004-10-14 Genentech, Inc. Therapy of autoimmune disease in a patient with an inadequate response to TNF-alpha inhibitor
US20070185170A1 (en) 2004-01-14 2007-08-09 Juan Carlos Lacal Sanjuan Derivatives of pyridine and quinoline
EP1889920A1 (en) * 2005-04-13 2008-02-20 Consejo Superior De Investigaciones Cientificas In vitro cancer therapy compound identification method
US8481256B2 (en) 2005-04-13 2013-07-09 Consejo Superior De Investigaciones Cientificas In vitro method for identifying compounds for cancer therapy
WO2007138143A2 (en) 2006-05-29 2007-12-06 Consejo Superior De Investigaciones Cientificas Alpha anticholine kinase monoclonal antibodies and their use in analytical techniques for the diagnosis of cancer and the preparation of medicinal products
US20100068302A1 (en) 2008-09-17 2010-03-18 Traslational Cancer Drugs Pharma, S.L. Methods and compositions for the treatment of cancer
US20110212994A1 (en) * 2009-06-26 2011-09-01 Brian Clem Small Molecule Choline Kinase Inhibitors, Screening Assays, and Methods for Safe and Effective Treatment of Neoplastic Disorders

Non-Patent Citations (41)

* Cited by examiner, † Cited by third party
Title
ALVARO-GRACIA, J.M. ET AL.: "Cytokines in chronic inflammatory arthritis. VI. Analysis of the synovial cells involved in granulocyte-macrophage colony-stimulating factor production and gene expression in rheumatoid arthritis and its regulation by IL-1 and tumor necrosis factor-alpha", JOURNAL OF IMMUNOLOGY, vol. 146, no. 10, 1991, pages 3365 - 71
ALVARO-GRACIA, J.M.; N.J. ZVAIFLER; G.S. FIRESTEIN: "Cytokines in chronic inflammatory arthritis. V. Mutual antagonism between interferon-gamma and tumor necrosis factor-alpha on HLA-DR expression, proliferation, collagenase production, and granulocyte macrophage colony-stimulating factor production by rheumatoid arthritis synoviocytes", THE JOURNAL OF CLINICAL INVESTIGATION, vol. 86, no. 6, 1990, pages 1790 - 8
ASIAGO, V.M. ET AL.: "Early Detection of Recurrent Breast Cancer Using Metabolite Profiling", CANCER RESEARCH, vol. 70, no. 21, 2010, pages 8309 - 8318, XP055060861, DOI: doi:10.1158/0008-5472.CAN-10-1319
BARTOK, B. ET AL.: "P13 kinase delta is a key regulator of synoviocyte function in rheumatoid arthritis", THE AMERICAN JOURNAL OF PATHOLOGY, vol. 180, no. 5, 2012, pages 1906 - 16, XP055162593, DOI: doi:10.1016/j.ajpath.2012.01.030
BARTOK, B.; G.S. FIRESTEIN: "Fibroblast-like synoviocytes: key effector cells in rheumatoid arthritis", IMMUNOLOGICAL REVIEWS, vol. 233, no. 1, 2010, pages 233 - 55, XP055096873, DOI: doi:10.1111/j.0105-2896.2009.00859.x
BEATRIX BARTOK ET AL: "Fibroblast-like synoviocytes: key effector cells in rheumatoid arthritis", IMMUNOLOGICAL REVIEWS, vol. 233, no. 1, 1 January 2010 (2010-01-01), pages 233 - 255, XP055096873, ISSN: 0105-2896, DOI: 10.1111/j.0105-2896.2009.00859.x *
BECKONERT, 0. ET AL.: "Metabolic profiling, metabolomic and metabonomic procedures for NMR spectroscopy of urine, plasma, serum and tissue extracts", NATURE PROTOCOLS, vol. 2, no. 11, 2007, pages 2692 - 2703
BELOUECHE-BABARI, M. ET AL.: "Metabolic assessment of the action of targeted cancer therapeutics using magnetic resonance spectroscopy", BRITISH JOURNAL OF CANCER, vol. 102, no. 1, 2010, pages 1 - 7
CASADO-VELA, J. ET AL.: "Approaches for the study of cancer: towards the integration of genomics, proteomics and metabolomics", CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO, vol. 13, no. 9, 2011, pages 617 - 28, XP019947289, DOI: doi:10.1007/s12094-011-0707-9
DEBERARDINIS, R.J. ET AL.: "The biology of cancer: Metabolic reprogramming fuels cell growth and proliferation", CELL METABOLISM, vol. 7, no. 1, 2008, pages 11 - 20
ECKHART, A.D.; K. BEEBE; M. MILBURN: "Metabolomics as a key integrator for ''omic'' advancement of personalized medicine and future therapies", CLINICAL AND TRANSLATIONAL SCIENCE, vol. 5, no. 3, 2012, pages 285 - 8, XP055194689, DOI: doi:10.1111/j.1752-8062.2011.00388.x
FIRESTEIN, G.S.: "Evolving concepts of rheumatoid arthritis", NATURE, vol. 423, no. 6937, 2003, pages 356 - 61
GALLEGO-ORTEGA, D. ET AL.: "Involvement of human choline kinase alpha and beta in carcinogenesis: a different role in lipid metabolism and biological functions", ADVANCES IN ENZYME REGULATION, vol. 51, no. 1, 2011, pages 183 - 94, XP028192087, DOI: doi:10.1016/j.advenzreg.2010.09.010
GLUNDE, K. ET AL.: "MRS and MRSI guidance in molecular medicine: targeting and monitoring of choline and glucose metabolism in cancer", NMR IN BIOMEDICINE, vol. 24, no. 6, 2011, pages 673 - 90
GLUNDE, K.; C. JIE; Z.M. BHUJWALLA: "Molecular causes of the aberrant choline phospholipid metabolism in breast cancer", CANCER RESEARCH, vol. 64, no. 12, 2004, pages 4270 - 6
GLUNDE, K.; Z.M. BHUJWALLA; S.M. RONEN: "Choline metabolism in malignant transformation", NATURE REVIEWS. CANCER, vol. 11, no. 12, 2011, pages 835 - 48
GUMA MONICA ET AL: "Choline Kinase: A Novel Target For Rheumatoid Arthritis", ARTHRITIS & RHEUMATISM, WILEY, US, vol. 65, no. Suppl.10, 1 October 2013 (2013-10-01), pages S542, XP009182069, ISSN: 0004-3591 *
HAN, Z. ET AL.: "c-Jun N-terminal kinase is required for metalloproteinase expression and joint destruction in inflammatory arthritis", THE JOURNAL OF CLINICAL INVESTIGATION, vol. 108, no. 1, 2001, pages 73 - 81, XP002972691, DOI: doi:10.1172/JCI200112466
HSU, P.P.; D.M. SABATINI: "Cancer cell metabolism: Warburg and beyond", CELL, vol. 134, no. 5, 2008, pages 703 - 707
HUGLE, T. ET AL.: "Synovial fluid metabolomics in different forms of arthritis assessed by nuclear magnetic resonance spectroscopy", CLINICAL AND EXPERIMENTAL RHEUMATOLOGY, vol. 30, no. 2, 2012, pages 240 - 5
INAZU MASATO ET AL: "Functional analysis of choline transporters in rheumatoid arthritis synovial fibroblasts", JOURNAL OF PHARMACOLOGICAL SCIENCES, JAPANESE PHARMACOLOGICAL SOCIETY, TOKYO, JP, vol. 118, no. Suppl. 1, 1 January 2012 (2012-01-01), pages 208P, XP009182080, ISSN: 1347-8613 *
KAVANAUGH, A. ET AL.: "Assessment of rituximab's immunomodulatory synovial effects (ARISE trial). 1: clinical and synovial biomarker results", ANNALS OF THE RHEUMATIC DISEASES, vol. 67, no. 3, 2008, pages 402 - 8
KREMER, J.M. ET AL.: "The safety and efficacy of a JAK inhibitor in patients with active rheumatoid arthritis: Results of a double-blind, placebo-controlled phase IIa trial of three dosage levels of CP-690,550 versus placebo", ARTHRITIS AND RHEUMATISM, vol. 60, no. 7, 2009, pages 1895 - 905
KYBURZ, D.; M. CORR: "The KRN mouse model of inflammatory arthritis", SPRINGER SEMINARS IN IMMUNOPATHOLOGY, vol. 25, no. 1, 2003, pages 79 - 90
LAURIDSEN, M.B. ET AL.: "1H NMR spectroscopy-based interventional metabolic phenotyping: a cohort study of rheumatoid arthritis patients", JOURNAL OF PROTEOME RESEARCH, vol. 9, no. 9, 2010, pages 4545 - 53
LEE, D.M. ET AL.: "Cadherin-11 in synovial lining formation and pathology in arthritis", SCIENCE, vol. 315, no. 5814, 2007, pages 1006 - 10, XP002528086, DOI: doi:10.1126/SCIENCE.1137306
LEE, S.I. ET AL.: "Regulation of inflammatory arthritis by the upstream kinase mitogen activated protein kinase kinase 7 in the c-Jun N-terminal kinase pathway", ARTHRITIS RESEARCH & THERAPY, vol. 14, no. 1, 2012, pages R38, XP021119730, DOI: doi:10.1186/ar3750
LEFEVRE, S. ET AL.: "Synovial fibroblasts spread rheumatoid arthritis to unaffected joints", NATURE MEDICINE, vol. 15, no. 12, 2009, pages 1414 - 20
LORIO, E. ET AL.: "Alterations of choline phospholipid metabolism in ovarian tumor progression", CANCER RESEARCH, vol. 65, no. 20, 2005, pages 9369 - 76, XP055011314, DOI: doi:10.1158/0008-5472.CAN-05-1146
MACINTYRE, D.A. ET AL.: "Serum metabolome analysis by (I )H-NMR reveals differences between chronic lymphocytic leukaemia molecular subgroups", LEUKEMIA, vol. 24, no. 4, 2010, pages 788 - 797
MADSEN, R.K. ET AL.: "Diagnostic properties of metabolic perturbations in rheumatoid arthritis", ARTHRITIS RESEARCH & THERAPY, vol. 13, no. 1, 2011, pages R19, XP021091608, DOI: doi:10.1186/ar3243
PSYCHOGIOS, N. ET AL.: "The Human Serum Metabolome", PLOS ONE, vol. 6, no. 2, 2011
RODRIGUEZ-GONZALEZ, A. ET AL.: "Inhibition of choline kinase as a specific cytotoxic strategy in oncogene-transformed cells", ONCOGENE, vol. 22, no. 55, 2003, pages 8803 - 12
SCHIPPER, R.G. ET AL.: "Polyamines and DNA methylation in childhood leukaemia", BIOCHEMICAL SOCIETY TRANSACTIONS, vol. 35, 2007, pages 331 - 5
SEEGER, K.: "Metabolic changes in autoimmune diseases", CURRENT DRUG DISCOVERY TECHNOLOGIES, vol. 6, no. 4, 2009, pages 256 - 61
SWEENEY, S.E.; G.S. FIRESTEIN: "Primer: signal transduction in rheumatic disease--a clinician's guide", NATURE CLINICAL PRACTICE. RHEUMATOLOGY, vol. 3, no. 11, 2007, pages 651 - 60
TIZIANI, S. ET AL.: "Metabolomic profiling of drug responses in acute myeloid leukaemia cell lines", PLOS ONE, vol. 4, no. 1, 2009, pages E4251
TIZIANI, S.; V. LOPES; U.L. GÜNTHER: "Early Stage Diagnosis of Oral Cancer Using 1H NMR-Based Metabolomics", NEOPLASIA, vol. 11, no. 3, 2009, pages 269 - U69
WEINBLATT, M.E. ET AL.: "Treatment of rheumatoid arthritis with a Syk kinase inhibitor: a twelve-week, randomized, placebo-controlled trial", ARTHRITIS AND RHEUMATISM, vol. 58, no. 11, 2008, pages 3309 - 18
WELJIE, A.M. ET AL.: "An inflammatory arthritis-associated metabolite biomarker pattern revealed by 1H NMR spectroscopy", JOURNAL OF PROTEOME RESEARCH, vol. 6, no. 9, 2007, pages 3456 - 64
YALCIN, A. ET AL.: "Selective inhibition of choline kinase simultaneously attenuates MAPK and PI3K/AKT signaling", ONCOGENE, vol. 29, no. 1, 2010, pages 139 - 49

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020057549A1 (en) 2018-09-18 2020-03-26 Beijing Innocare Pharma Tech Co., Ltd. Crystalline forms of 6- (1-acryloylpiperidin-4-yl) -2- (4-phenoxyphenyl) nicotinamide

Similar Documents

Publication Publication Date Title
Lee et al. Metformin overcomes resistance to cisplatin in triple-negative breast cancer (TNBC) cells by targeting RAD51
Wang et al. Genistein suppresses psoriasis-related inflammation through a STAT3–NF-κB-dependent mechanism in keratinocytes
Liao et al. Inhibition of EGFR signaling with Spautin-1 represents a novel therapeutics for prostate cancer
Kim et al. Hypoxia enhances lysophosphatidic acid responsiveness in ovarian cancer cells and lysophosphatidic acid induces ovarian tumor metastasis in vivo
Li et al. PI3 kinase/Akt/HIF-1α pathway is associated with hypoxia-induced epithelial–mesenchymal transition in fibroblast-like synoviocytes of rheumatoid arthritis
Shi et al. De novo pyrimidine synthesis is a targetable vulnerability in IDH mutant glioma
Zhang et al. PLK4 is a determinant of temozolomide sensitivity through phosphorylation of IKBKE in glioblastoma
Tailor et al. Y box binding protein 1 inhibition as a targeted therapy for ovarian cancer
US20120107323A1 (en) Kinase protein binding inhibitors
WO2007100640A2 (en) Growth hormone receptor antagonist cancer treatment
Sun et al. Sphingosine kinase 1/sphingosine 1-phosphate/sphingosine 1-phosphate receptor 1 pathway: A novel target of geniposide to inhibit angiogenesis
KR20150010793A (en) Methods of treating a metabolic syndrome by modulating heat shock protein (hsp) 90-beta
Lin et al. Targeting positive feedback between BASP1 and EGFR as a therapeutic strategy for lung cancer progression
US20180011102A1 (en) The protein kinase activity of phosphoglycerate kinase 1 as a target for cancer treatment and diagnosis
US20160303137A1 (en) Dual pi3k and wnt pathway inhibition as a treatment for cancer
US20230068698A1 (en) Combination therapy for cancer
KR20110084533A (en) Enzastaurin for the treatment of cancer
Ding et al. SCP2-mediated cholesterol membrane trafficking promotes the growth of pituitary adenomas via Hedgehog signaling activation
de Miguel et al. Mammalian SWI/SNF chromatin remodeling complexes promote tyrosine kinase inhibitor resistance in EGFR-mutant lung cancer
CA3191363A1 (en) Pharmaceutical combination and tumor treatment
MX2012013305A (en) Combination therapy and method for assessing resistance to treatment.
Cui et al. Blockage of EGFR/AKT and mevalonate pathways synergize the antitumor effect of temozolomide by reprogramming energy metabolism in glioblastoma
Zhu et al. The effect of benzyl isothiocyanate and its computer-aided design derivants targeting alkylglycerone phosphate synthase on the inhibition of human glioma U87MG cell line
US10894837B2 (en) MMP9 inhibitors and uses thereof in the prevention or treatment of a depigmenting disorder
He et al. Thyroid carcinoma cells produce PLGF to enhance metastasis

Legal Events

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

Ref country code: DE

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

Ref document number: 14758389

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 14758389

Country of ref document: EP

Kind code of ref document: A1