WO2008090331A1 - Serotonin reuptake inhibitors for treating arthritis - Google Patents

Serotonin reuptake inhibitors for treating arthritis Download PDF

Info

Publication number
WO2008090331A1
WO2008090331A1 PCT/GB2008/000217 GB2008000217W WO2008090331A1 WO 2008090331 A1 WO2008090331 A1 WO 2008090331A1 GB 2008000217 W GB2008000217 W GB 2008000217W WO 2008090331 A1 WO2008090331 A1 WO 2008090331A1
Authority
WO
WIPO (PCT)
Prior art keywords
reuptake inhibitor
serotonin reuptake
serotonin
agent
inflammatory
Prior art date
Application number
PCT/GB2008/000217
Other languages
French (fr)
Inventor
Brian M. Foxwell
Sandra Sacre
Original Assignee
Imperial Innovations Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Imperial Innovations Limited filed Critical Imperial Innovations Limited
Publication of WO2008090331A1 publication Critical patent/WO2008090331A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/138Aryloxyalkylamines, e.g. propranolol, tamoxifen, phenoxybenzamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/343Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide condensed with a carbocyclic ring, e.g. coumaran, bufuralol, befunolol, clobenfurol, amiodarone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics

Definitions

  • the present invention relates to serotonin reuptake inhibitors (SRIs) including selective serotonin reuptake inhibitors (SSRIs) and serotonin norepinephrine reuptake inhibitors (SNRIs) for treating inflammatory arthritis and related conditions.
  • SRIs serotonin reuptake inhibitors
  • SSRIs selective serotonin reuptake inhibitors
  • SNRIs serotonin norepinephrine reuptake inhibitors
  • the present invention also relates to a kit comprising a pharmaceutical composition comprising a serotonin reuptake inhibitor and instructions indicating that the composition is for use in treating inflammatory arthritis and related conditions.
  • the present invention relates to a method of treating inflammatory arthritis and related conditions comprising administration of a serotonin reuptake inhibitor. The method is preferably carried out on a subject in need of treatment.
  • the inflammatory arthritis is preferably rheumatoid arthritis.
  • Rheumatoid arthritis is a chronic inflammatory disease occurring in 0.5%-l% of the population. This disease affects the joints and is characterized by thickening of the synovial membrane. This disease leads to progressive debilitation in joint function which results in pain, disability, loss of man power and shorter life expectancy. In addition there are a number of similar related conditions; psoriatic arthritis, juvenile arthritis ankylosing spondelytis, Crohn's disease and psoriasis.
  • steroids such as dexamethasone and methyl-prednisolone
  • steroids are widely used in the treatment of rheumatoid arthritis. While treatment with steroids can be effective, there are a number of serious side effects. These side effects include hypertension, growth deficiencies in younger patients, osteoporosis, cataracts, psychosis, elevated blood sugar, glaucoma, etc.
  • some patients are resistant to long-term use of steroids or become so with time.
  • New and alternative treatments currently used for rheumatoid arthritis are based on biologicals such as antibodies and soluble receptors.
  • the most widely used of these is based on blocking TNF function with neutralizing antibodies or soluble receptors and has defined TNF as a key pathway in the inflammatory processes in RA.
  • This type of anti-TNF therapy has been successful in the treatment of a number of diseases, with a substantial proportion of patients (approximately 60%) showing significant clinical benefit Besides treating the inflammation, anti-TNF therapy also prevents joint destruction and thus is considered to have disease modifying anti-arth ⁇ tic drug (DMARD) activity
  • DMARD disease modifying anti-arth ⁇ tic drug
  • DARDS disease modifying anti-rheumatic drugs
  • methotrexate an anti-metabolite drug, which is widely used for the treatment of rheumatoid arthritis, pso ⁇ atic arthritis and pso ⁇ asis.
  • Methotrexate has been successful in the treatment of these diseases, but can cause substantial side effects, such as severe skin reaction, infections such as pneumonia, severe damage to liver, kidneys, lungs and gastrointestinal tract. Again a significant number of patients do not respond or become refractory with time.
  • a number of DMARD pharmaceutical agents containing gold are also used in the treatment of rheumatoid arthritis
  • examples of such agents include gold sodium thiomalate and auranofin
  • Potential side effects from being treated with antiinflammatory gold agents are oral ulcers, altered taste, se ⁇ ous skin rashes, renal problems, inflammation of the intestines (enterocolitis), liver injury and lung disease. Furthermore, resistance to gold has been known to develop in patients
  • a further class of drugs are the non-steroidal anti-inflammatory drugs (NSAED' s). These are used to alleviate symptoms but not modify disease progression and includes the Cox 2 inhibitors "VIOXX” ® , (a registered trademark of Merck & Co., Inc) and "CELEBREX” ® , (a registered trademark of G.D. Searle & Co). However there is major concern about the safety of these drugs with evidence of increased cardiovascular risk.
  • NSAED' s non-steroidal anti-inflammatory drugs
  • SRIs are well known for use in the treatment of depression and other related conditions such as OCD. SRIs (including SSRIs and SNRIs) would be expected to promote inflammation as serotonin is associated with inflammation. In addition, other studies have indicated that sertraline, a SSRI, exacerbates inflammation in the carrageenan-induced model of paw inflammation (Abdel-Salam et al., Pharmacol Res 48, 157-165, 2003).
  • the first aspect of the present invention provides a serotonin reuptake inhibitor (SRI) for treating inflammatory arthritis and related conditions.
  • SRI serotonin reuptake inhibitor
  • the inflammatory arthritis is anti-TNF responsive arthritis, as are the related conditions psoriatic arthritis, juvenile arthritis, ankylosing spondelytis, Crohn's disease and psoriasis.
  • These drugs are envisaged to be useful in diseases where treatment with anti-TNF has shown to be beneficial e.g. Crohn's disease and psoriasis, but not useful in diseases where anti-TNF has been shown to be ineffective or to exacerbate disease, e.g. systemic lupus erythematosus, multiple sclerosis and ulcerative colitis.
  • LPS lipopolysaccharide
  • the inflammatory arthritis is rheumatoid arthritis and the related conditions are psoriatic arthritis, juvenile arthritis, ankylosing spondelytis, Crohn's disease and psoriasis.
  • a serotonin reuptake inhibitor of the invention is any compound that inhibits reuptake of serotonin. This includes selective serotonin reuptake inhibitor (SSRI) compounds and serotonin norepinephrine reuptake inhibitor (SNRI) compounds.
  • SSRI selective serotonin reuptake inhibitor
  • SNRI serotonin norepinephrine reuptake inhibitor
  • a SRI is a drug that increases the amount of serotonin (5- hydroxytryptamine or 5-HT) by inhibiting its reuptake into cells.
  • the drug is an inhibitor of the serotonin transporter (5-HTT), a monoamine transporter protein.
  • a SRI of the invention inhibits TNF production.
  • the serotonin reuptake inhibitor of the invention can be amoxapine, amitriptyline ("ELAVIL”, “ENDEP”, “TRYPTANOL”, “TREPILINE”), clomipramine
  • ANAFRANIL desipramine
  • NPRAMIN desipramine
  • PROTfflADEN dothiepin hydrochloride
  • ADAPIN doxepin
  • SINEQUAN imipramine
  • TOFRANIL lofepramine
  • GAMANIL lofepramine
  • PROTAPEL lofepramine
  • PAMELOR nortriptyline
  • VIVACTIL trimipramine
  • trimipramine SURMONTIL
  • the selective serotonin reuptake inhibitor of the invention can be citalopram ("CELEXA”, “CEPRAMIL”, “EMOCAL”, “SEPRAM”, “SEROPRAM”), escitalopram oxalate ("LEXAPRO”, “CIPRALEX”, “ESERTIA”), fluoxetine ("PROZAC”, “FONTEX”, “SEROMEX”, “SERONIL”, “SARAFEM”, “FLUCTIN” (EUR)), fluvoxamine maleate ("LUVOX”, “FAVERIN”), paroxetine ("PAXIL”, “SEROXAT”, “AROPAX”, “DEROXAT”, “PAROXAT”), sertraline (“ZOLOFT”, "LUSTRAL”, “SERLAIN”), dapoxetine or St Johns Wort
  • the serotonin-norepinephnne reuptake inhibitor of the invention can be venlafaxine ("EFFEXOR XR” and “EFEXOR”), nefazodone ("SERZONE”), milnacipran (“DALCIPRAN” and “IXEL”), desipramine (“N0RPRAMINE” and “PERTOFRANEIS”) or duloxetine ("CYMBALTA”).
  • the use may be of combinations of two or more SRIs, which may be for simultaneous, separate or sequential use.
  • the SRI may be used in combination with a further anti-inflammatory agent.
  • Administration of the SRI and other anti-inflammatory agent can be simultaneous, separate and/or sequential
  • the SRI, in combination with another pharmaceutical agent, can act additively or synergistically.
  • the other anti -inflammatory agent may be termed a non-steroidal anti-inflammatory agent (NSAID), a disease modifying anti-rheumatic drug (DMARD), a statin (including HMG-CoA reductase inhibitors such as simvastatin), a biological agent (biologicals), a steroid, an immunosuppressive agent, a salicylate and/or a microbicidal agent.
  • NSAID non-steroidal anti-inflammatory agent
  • DMARD disease modifying anti-rheumatic drug
  • statin including HMG-CoA reductase inhibitors such as simvastatin
  • a biological agent biologicals
  • a steroid an immunosuppressive agent
  • an immunosuppressive agent a salicylate and/or a microbicidal agent.
  • Non-steroidal anti-inflammatory agents include anti-metabolite agents (including methotrexate) and anti-inflammatory gold agents (including gold sodium thiomalate, aurothiomalate or gold
  • Biologicals include anti-TNF agents (including adalimumab, etanercept, infliximab, anti-EL-1 leagents, anti-IL-6 reagents, anti-B cell reagents (retoximab), anti-T cell reagents (anti-CD4 antibodies), anti-IL-15 reagents, anti-CLTA4 reagents, anti-RAGE reagents), antibodies, soluble receptors, receptor binding proteins, cytokine binding proteins, mutant proteins with altered or attenuated functions, RNAi, polynucleotide aptmers, antisense oligonucleotides or omega 3 fatty acids
  • Steroids include cortisone, prednisolone or dexamethasone.
  • Immunosuppressive agents include cylcospo ⁇ n, FK506, rapamycin, mycophenolic acid Salicylates include aspirin, sodium salicylate, choline salicylate and magnesium salicylate. Microbicidal agents include quinine and chloroquine.
  • the SRI may be administered in combination with one or more of an NSAID, DMARD, or immunosuppressant.
  • the further anti-inflammatory agent is administered by any appropriate route, for example oral (including buccal or sublingual), suppositories, topical (including buccal, sublingual or transdermal), or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route.
  • oral including buccal or sublingual
  • suppositories including buccal, sublingual or transdermal
  • parenteral including subcutaneous, intramuscular, intravenous or intradermal
  • the SRIs of the present invention are those SRIs which are non-toxic to living cells.
  • the present invention also provides the use of a serotonin reuptake inhibitor (SRI) in the manufacture of a medicament for treating inflammatory arthritis and related conditions.
  • SRI serotonin reuptake inhibitor
  • the second aspect of the invention is a kit comprising a pharmaceutical composition comprising an SRI and instructions indicating that the composition is for use in treating inflammatory arthritis and related conditions.
  • compositions in accordance with the invention may be supplied as part of a sterile, pharmaceutical composition which will normally include a pharmaceutically acceptable earner.
  • This pharmaceutical composition may be in any suitable form. It may be provided in unit dosage form and will generally be provided in a sealed container.
  • the kit of the invention may comprise a plurality of said unit dosage forms.
  • compositions may be presented as discrete units such as capsules or tablets; as powders or granules; as solutions, syrups or suspensions (in aqueous or non-aqueous liquids; or as edible foams or whips; or as emulsions).
  • Suitable excipients for tablets or hard gelatine capsules include lactose, maize starch or derivatives thereof, stea ⁇ c acid or salts thereof
  • Suitable excipients for use with soft gelatine capsules include for example vegetable oils, waxes, fats, semi-solid, or liquid polyols etc
  • excipients which may be used include for example water, polyols and sugars
  • suspensions oils e g vegetable oils
  • compositions may contain preserving agents, solubilising agents, stabilising agents, wetting agents, emulsifiers, sweeteners, colourants, odourants, salts, buffers, coating agents or antioxidants They may also contain further therapeutically active agents
  • Route of administration may include, parenterally (including subcutaneous, intramuscular, intravenous, by means of, for example a d ⁇ p patch), some further suitable routes of administration include (but are not limited to) oral (including buccal and sublingual), rectal, nasal, topical, infusion, vaginal, intradermal, intrape ⁇ toneally, intracranially, intrathecal and epidural administration or administration via oral or nasal inhalation, by means of, for example a nebuliser or inhaler, or by an implant
  • the may be delivered using a mechanical form including, but not restricted to, an inhaler or nebuliser device
  • administration is by a SPAG (small particulate aerosol generator) may be used
  • Dosages of the substances of the present invention can vary between wide limits, depending upon the condition to be treated, the health of the individual to be treated, etc and a physician may determine approp ⁇ ate dosages to be used The dosage may be repeated as often as approp ⁇ ate
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy
  • the amount of active ingredient which can be combined with a earner matenal to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration
  • the amount of active ingredient which can be combined with a earner material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect
  • compositions and uses described in this application are envisaged to have human, animal and veterinary applications They are preferably applicable to mammals, in particular humans but are also applicable for use in production animals, in particular sheep, cows, pigs, chickens and goats, as well as companion animals, in particular cats and dogs and sporting animals, such as horses
  • the third aspect of the invention is a method of treating inflammatory arthritis and related conditions comprising administration of a serotonin reuptake inhibitor
  • the method is preferably earned out on a subject in need of treatment
  • the term "treatment” includes prophylactic treatment (i e prevention)
  • prophylactic treatment i e prevention
  • prevention of inflammatory arthntis or a related condition is unlikely to be earned out Usually, it is only when the presence of inflammatory arthntis or a related condition is diagnosed in a subject that prevention means are applied
  • prophylactic treatment may be appropnate if there is i) a known family history of significant inflammatory arthntis or a related condition or if tests (e g genetic tests) identify that an individual has a predisposition to inflammatory arthntis or a related condition
  • Figure 1 illustrates inhibition of spontaneous cytokine release from RA synovial membrane cultures by citalopram and fluoxetine hydrochlonde (“PROZAC")
  • Graph (A) illustrates a dose dependent inhibition of TNF levels and IL- l ⁇ levels by fluoxetine hydrochlo ⁇ de (“PROZAC”) as measured by ELISA
  • Graph (B) illustrates a dose dependent inhibition of TNF in cells incubated with citalopram
  • Figure 3 illustrates inhibition of disease progression in the CIA model of rheumatoid arthritis by fluoxetine hydrochlo ⁇ de (“PROZAC").
  • A illustrates clinical score
  • Figure 4 Graph (A) illustrates inhibition by fluoxetine hydrochlonde of TNF production from human macrophages activated by LPS (10ng/ml), Flagelhn (lOng/ml), R848 (l ⁇ g/ml) and malp-2 (lOng/ml)
  • Graph (B) illustrates TNF production in murine RAW264.7 macrophages stimulated with 20 ⁇ g/ml poly IC in the presence of media alone or fluoxetine hydrochlo ⁇ de
  • Graph (C) illustrates inhibition by fluoxetine hydrochlo ⁇ de of TNF production in CpG stimulated mu ⁇ ne RAW264 7 macrophages.
  • FIG. 5 illustrates that inhibition of TNF production by R-848 is common to the serotonin reuptake inhibitors.
  • Graph (A) illustrates TNF production from macrophages stimulated with l ⁇ g/ml R848 or 10ng/ml LPS in the media alone or in the presence of 30 ⁇ g/ml citalopram, lO ⁇ g/ml fluoxetine hydrochlo ⁇ de (“PROZAC") or 5 ⁇ g/ml sertraline (“ZOLOFT”)
  • FIG.B illustrates TNF levels from macrophages stimulated with 10ng/ml Pam3, 10ng/ml LPS, 10ng/ml Flagelhn, l ⁇ g/ml R848 and 10ng/ml malp-2 in media alone or in the presence of 5 ⁇ g/ml sertraline (“ZOLOFT”)
  • Figure 6 illustrates that TLR3, 7, 8 and 9 are expressed in the rheumatoid synovial membrane cultures but only TLR3 and 8 induce TNF production Graph
  • Graph (C) illustrates TNF production in rheumatoid membrane synovial cells after stimulation with 20 ⁇ g/ml Poly IC (TLR3), l ⁇ g/ml imiquimod (TLR7), l ⁇ g/ml R-848 (TLR7/8) and 2 ⁇ M CpG (TLR9).
  • Figure 7 illustrates the effect of serotonin on TNF and EL-I levels in RA membrane cell cultures Graph
  • Cell culture reagents used were Penicillin-Streptomycin, RPMI 1640 and DMEM obtained from Cambrex (Belgium), Indomethacin from Sigma (USA) and FBS from PAA (Aust ⁇ a).
  • the TLR hgands used were chloroform extracted Escherichia coli (E.coh) LPS, resiquimod (R-848), mu ⁇ ne CpG (ODN1826), human CpG (ODN 2006) and imiquimod from Invivogen (USA), Macrophage-activating hpopeptide (Malp-2), Flagellin (pu ⁇ fied) and serotonin (5-HT) hydrochlo ⁇ de were from Alexis (UK) Fluoxetine hydrochlo ⁇ de (“PROZAC”) and chloroquine diphosphate salt, Sertraline (“ZOLOFT”) and citalopram were purchased from Sigma (USA) Pam 3 cys- ser(lys) 4 3HCl (Pam3) was from Alex
  • TTGAAGGCTTGGGACCAAGGCA-S 1 were used with an annealing temperature of
  • the primers 5'- TCTACCTGGGCCAAAACTGTT-3' and 5'- GGCAC ATGCTGAAGAG AGTTA-3' were used with an annealing temperature of 6O 0 C.
  • AATGCTTCATTTGGGATGTGCT-3' were used with an annealing temperature of
  • ELISA enzyme-linked immunosorbent assay
  • Macrophages or RA synovial membrane cell cultures were stimulated with lOng/ml chloroform-extracted LPS, 20 ⁇ g/ml poly IC, 2 ⁇ M CpG (murine or human), 10ng/ml Pam3, 10ng/ml Flagellin, 10ng/ml Malp-2 or l ⁇ g/ml R-848 in complete media for 6h.
  • Supernatants from RA cultures were harvested after 24 hours.
  • AU reagents other than LPS were free from LPS contamination as assessed using the limulus amebocyte lysate (LAL) assay from Cambrex (USA).
  • Sandwich ELISAs were employed to measure TNF, (Pharmingen, UK), IP-10 (R&D, USA) and IL- l ⁇ (Bioscource, USA). Absorbance was read on a spectrophotometric ELISA plate reader (Labsystems Multiscan Biochromic) and analyzed using Ascent software V2.6 (Thermo Labsystems, Cambridge, United Kingdom). All results are expressed as the mean cytokine concentration ⁇ SD obtained from triplicate cultures per condition. Cell viability was not significantly affected over this time period when examined by the 3-[4,5 dimethylthiazol-2-yl]-2,5-diphenyl-tetrazolium bromide (MTT) assay (Sigma).
  • MTT 3-[4,5 dimethylthiazol-2-yl]-2,5-diphenyl-tetrazolium bromide
  • RA synovial membrane cells were isolated from patients undergoing joint replacement surgery as previously desc ⁇ bed (Foxwell et al., Proc. Natl. Acad. Sci. USA 95,8211-8215, 1998). All patients gave w ⁇ tten informed consent and the study was approved by the local ethics committee. Immediately after isolation, cells were used for mRNA analysis, stained by FACS or cultured at IxIO 5 cells/well in 96-well tissue culture plates (Falcon, UK) in RPMI 1640 containing 10% (v/v) FBS and 100 U/ml penicillin/streptomycin. Pe ⁇ pheral blood monocytes were isolated and cultured as previously desc ⁇ bed (Foxwell et al., Proc. Natl.
  • Macrophages were de ⁇ ved from monocytes after differentiation for 4 days with 100 ng/ml M-CSF (PeproTech, UK).
  • the mouse macrophage cell line RAW 264.7 was maintained in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum.
  • Example 1 Rheumatoid arthritis synovial membrane cells were cultured for 24 hours in the presence of media alone or media containing 5 ⁇ g/ml orlO ⁇ g/ml fluoxetine hydrochlo ⁇ de ("PROZAC"). Supernatants were harvested and levels of cytokines (A) TNF or IL- 1 were measured by ELISA Addition of fluoxetine hydrochlo ⁇ de (“PROZAC") at the time of culture led to a dose dependent decrease in the levels of TNF and IL-I ( Figure 1, Graph A).
  • Rheumatoid arthritis synovial membrane cells were also cultured in the presence of media alone or media containing lO ⁇ g/ml, 20 ⁇ g/ml and 30 ⁇ g/ml citalopram and levels of TNF were measured. TNF production was dose dependently inhibited in cells treated with citalopram (see Figure IB).
  • the murine CIA model has been shown to be clinically similar to human rheumatoid arthritis with a comparable synovitis, bone erosion and panus formation.
  • CFA Co Laboratories, West Molesey, U.K.
  • mice were given an interperitoneal injection of vehicle (PBS) or 25mg/kg of citalopram or fluoxetine hydrochlo ⁇ de once a day for 7 days Mice were assessed for ( Figures 2A and 3A) clinical score on a daily basis.
  • PBS vehicle
  • citalopram or fluoxetine hydrochlo ⁇ de
  • mice treated with citalopram at the day of onset of arthritis exhibited an attenuated disease progression over a 7 day pe ⁇ od.
  • Disease pathogenesis was assessed by measuring clinical score
  • the mice treated with citalopram showed a significant improvement to the control group from day 2-3 onwards ( Figure 2, Graph A) Histology samples taken from the hind paws of mice treated with citalopram showed a decreased histology score over the control group ( Figure 2B).
  • mice treated with fluoxetine hydrochlo ⁇ de at the day of onset of arthritis exhibited an attenuated disease progression over a 7 day pe ⁇ od. Disease pathogenesis was assessed by measu ⁇ ng clinical score.
  • the mice treated with fluoxetine hydrochlo ⁇ de showed a significant improvement to the control group ( Figure 3, Graph A, B).
  • This effect of fluoxetine hydrochlo ⁇ de was dose dependent as 10mg/kg showed a significant improvement throughout the study from the vehicle control, whilst the lOmg/kg group showed a small but not significant improvement (Figure 3, Graph A). This was also reflected in the measurement of hind paw swelling that also showed a significant improvement throughout the study for both the 10mg/kg and 20mg/kg treated groups ( Figure 3, Graph B).
  • P ⁇ mary human macrophages and munne RAW 264 7 cells were incubated with fluoxetine hydrochlonde for 30 minutes and then stimulated for 6 hours with TLR ligands (R-848, LPS, flagellin, malp-2, poly IC or CpG).
  • TLR ligands R-848, LPS, flagellin, malp-2, poly IC or CpG.
  • Human macrophages showed a decreased production of TNF in response to R-848 but not LPS, flagellin or Malp-2 in the presence of fluoxetine hydrochloride ( Figure 4, Graph A).
  • Murine RAW 264.7 cells were used to examine poly IC and CpG induced TNF, as unlike human macrophages they respond to poly IC a TLR3 ligand and CpG, a TLR9 ligand to produce TNF. Fluoxetine hydrochloride was able to inhibit TNF production in these cells on stimulation with poly IC ( Figure 4, Graph B) and CPG ( Figure 4, Graph C).
  • TLRs toll-like receptors
  • TLR3, 7, 8 and 9 are expressed in human RA synovial tissue. mRNA from synovial tissue was extracted, DNAase treated and then analysed by RT-PCR. Expression of message for TLR3, 7, 8 and 9 was detected by RT-PCR from the mixed population of cells in human rheumatoid synovial tissue in three unrelated donors 1-3 ( Figure 6, Graph A). TLR3, 8 and 9 were further investigated using FACS. Cells were washed, then blocked with 10% human serum in PBS containing 0.01% azide for 30 minutes at 4°C or for intracellular staining cells were fixed in 2% paraformaldehyde and permeabilised with 0.1% saponin (Sigma, USA) before blocking.
  • TLR3, 8 and 9 protein expression was detected at low levels on the cell surface but was more readily detected upon intracellular staining ( Figure 6, Graph B).
  • the main mechanism of action of the antidepressants (SRIs) used in this study is to increase the amount of serotonin by inhibiting serotonin reuptake receptors.
  • SRIs antidepressants
  • Fluoxetine hydrochlo ⁇ de (“PROZAC"), citalopram and sertraline have been demonstrated as being able to inhibit selective TLRs TLR3, 7, 8 and 9 are classically viewed as having an endosomal localization. Chloroquine has been reported to inhibit the function of TLR3, 7, 8 and 9, by increasing the pH inside vesicles, which is essential for functional signaling from TLR3, 7, 8 and 9.
  • chloroquine inhibits spontaneous TNF and EL-I release but serotonin does not

Abstract

The present invention relates to serotonin reuptake inhibitors (SRIs) including selective serotonin reuptake inhibitors (SSRIs) and serotonin norepinephrine reuptake inhibitors (SNRIs) for treating inflammatory arthritis and related conditions. The present invention also relates to a kit comprising a pharmaceutical composition comprising a serotonin reuptake inhibitor and instructions indicating that the composition is for use in treating inflammatory arthritis and related conditions. In addition, the present invention relates to a method of treating inflammatory arthritis and related conditions comprising administration of a serotonin reuptake inhibitor. The method is preferably carried out on a subject in need of treatment. The inflammatory arthritis is preferably rheumatoid arthritis.

Description

Serotonin reuptake inhibitors for treating arthritis
The present invention relates to serotonin reuptake inhibitors (SRIs) including selective serotonin reuptake inhibitors (SSRIs) and serotonin norepinephrine reuptake inhibitors (SNRIs) for treating inflammatory arthritis and related conditions. The present invention also relates to a kit comprising a pharmaceutical composition comprising a serotonin reuptake inhibitor and instructions indicating that the composition is for use in treating inflammatory arthritis and related conditions. In addition, the present invention relates to a method of treating inflammatory arthritis and related conditions comprising administration of a serotonin reuptake inhibitor. The method is preferably carried out on a subject in need of treatment. The inflammatory arthritis is preferably rheumatoid arthritis.
Rheumatoid arthritis is a chronic inflammatory disease occurring in 0.5%-l% of the population. This disease affects the joints and is characterized by thickening of the synovial membrane. This disease leads to progressive debilitation in joint function which results in pain, disability, loss of man power and shorter life expectancy. In addition there are a number of similar related conditions; psoriatic arthritis, juvenile arthritis ankylosing spondelytis, Crohn's disease and psoriasis.
Current treatments for rheumatoid arthritis have a number of disadvantages, including expense and/or severe side effects. At present, steroids such as dexamethasone and methyl-prednisolone, are widely used in the treatment of rheumatoid arthritis. While treatment with steroids can be effective, there are a number of serious side effects. These side effects include hypertension, growth deficiencies in younger patients, osteoporosis, cataracts, psychosis, elevated blood sugar, glaucoma, etc. In addition, some patients are resistant to long-term use of steroids or become so with time.
New and alternative treatments currently used for rheumatoid arthritis are based on biologicals such as antibodies and soluble receptors. The most widely used of these is based on blocking TNF function with neutralizing antibodies or soluble receptors and has defined TNF as a key pathway in the inflammatory processes in RA. This type of anti-TNF therapy has been successful in the treatment of a number of diseases, with a substantial proportion of patients (approximately 60%) showing significant clinical benefit Besides treating the inflammation, anti-TNF therapy also prevents joint destruction and thus is considered to have disease modifying anti-arthπtic drug (DMARD) activity However, it is extremely expensive and this places a heavy financial burden either on the patient or the healthcare system or both and this seriously limits availability. Many patients in the developed world and the majoπty in the developing world are not able to afford this treatment. In addition, possible side effects of anti-TNF therapy include anaphylaxis and cytopenia Moreover systemic neutralisation of TNF leads to increased susceptibility to infection and the long-term effects are still unknown Currently it is not possible to take anti-TNF drugs orally, which is a disadvantage Moreover of those patients that do respond approximately 50% will become refractory to treatment in 2 years due to the development of immunity to the biological treatment
Another class of drugs are the disease modifying anti-rheumatic drugs (DMARDS). An example of these is methotrexate, an anti-metabolite drug, which is widely used for the treatment of rheumatoid arthritis, psoπatic arthritis and psoπasis. Methotrexate has been successful in the treatment of these diseases, but can cause substantial side effects, such as severe skin reaction, infections such as pneumonia, severe damage to liver, kidneys, lungs and gastrointestinal tract. Again a significant number of patients do not respond or become refractory with time.
A number of DMARD pharmaceutical agents containing gold are also used in the treatment of rheumatoid arthritis Examples of such agents include gold sodium thiomalate and auranofin Potential side effects from being treated with antiinflammatory gold agents are oral ulcers, altered taste, seπous skin rashes, renal problems, inflammation of the intestines (enterocolitis), liver injury and lung disease. Furthermore, resistance to gold has been known to develop in patients
A further class of drugs are the non-steroidal anti-inflammatory drugs (NSAED' s). These are used to alleviate symptoms but not modify disease progression and includes the Cox 2 inhibitors "VIOXX"®, (a registered trademark of Merck & Co., Inc) and "CELEBREX" ®, (a registered trademark of G.D. Searle & Co). However there is major concern about the safety of these drugs with evidence of increased cardiovascular risk.
As a result of lack of efficacy, development of resistance, unacceptable side-effects and expense of existing treatments and route of administration, it is hugely desirable to find alternative treatments for rheumatoid arthritis. Thus there is a huge unmet medical need for an orally-available, well tolerated, inexpensive drug that could selectively block the production of TNF associated with pathological inflammation found in RA and related conditions.
SRIs (including SSRIs and SNRIs) are well known for use in the treatment of depression and other related conditions such as OCD. SRIs (including SSRIs and SNRIs) would be expected to promote inflammation as serotonin is associated with inflammation. In addition, other studies have indicated that sertraline, a SSRI, exacerbates inflammation in the carrageenan-induced model of paw inflammation (Abdel-Salam et al., Pharmacol Res 48, 157-165, 2003).
The first aspect of the present invention provides a serotonin reuptake inhibitor (SRI) for treating inflammatory arthritis and related conditions.
The inflammatory arthritis is anti-TNF responsive arthritis, as are the related conditions psoriatic arthritis, juvenile arthritis, ankylosing spondelytis, Crohn's disease and psoriasis. These drugs are envisaged to be useful in diseases where treatment with anti-TNF has shown to be beneficial e.g. Crohn's disease and psoriasis, but not useful in diseases where anti-TNF has been shown to be ineffective or to exacerbate disease, e.g. systemic lupus erythematosus, multiple sclerosis and ulcerative colitis. These drugs would not be expected to be beneficial for the treatment of sepsis where TNF is produced by lipopolysaccharide (LPS) stimulation. These drugs are unable to inhibit LPS induced TNF. This indicates that these drugs do not possess general anti-inflammatory properties. In a preferred embodiment the inflammatory arthritis is rheumatoid arthritis and the related conditions are psoriatic arthritis, juvenile arthritis, ankylosing spondelytis, Crohn's disease and psoriasis.
A serotonin reuptake inhibitor of the invention is any compound that inhibits reuptake of serotonin. This includes selective serotonin reuptake inhibitor (SSRI) compounds and serotonin norepinephrine reuptake inhibitor (SNRI) compounds.
As is known in the art, a SRI is a drug that increases the amount of serotonin (5- hydroxytryptamine or 5-HT) by inhibiting its reuptake into cells. The drug is an inhibitor of the serotonin transporter (5-HTT), a monoamine transporter protein.
A SRI of the invention inhibits TNF production.
Information on SRIs can be found in Wilson et al., J. Med. Chem. 43, 3103-3110, 2000 and Tatsumi et al., European Journal of Pharmacology 340, 249-258, 1997.
The serotonin reuptake inhibitor of the invention can be amoxapine, amitriptyline ("ELAVIL", "ENDEP", "TRYPTANOL", "TREPILINE"), clomipramine
("ANAFRANIL"), desipramine ("NORPRAMIN", "PERTOFRANE"), dothiepin hydrochloride ("PROTfflADEN", "THADEN"), doxepin ("ADAPIN", "SINEQUAN"), imipramine ("TOFRANIL"), lofepramine ("GAMANIL", "LOMONT"), nortriptyline ("PAMELOR"), protriptyline ("VIVACTIL") or trimipramine ("SURMONTIL").
The selective serotonin reuptake inhibitor of the invention can be citalopram ("CELEXA", "CEPRAMIL", "EMOCAL", "SEPRAM", "SEROPRAM"), escitalopram oxalate ("LEXAPRO", "CIPRALEX", "ESERTIA"), fluoxetine ("PROZAC", "FONTEX", "SEROMEX", "SERONIL", "SARAFEM", "FLUCTIN" (EUR)), fluvoxamine maleate ("LUVOX", "FAVERIN"), paroxetine ("PAXIL", "SEROXAT", "AROPAX", "DEROXAT", "PAROXAT"), sertraline ("ZOLOFT", "LUSTRAL", "SERLAIN"), dapoxetine or St Johns Wort
The serotonin-norepinephnne reuptake inhibitor of the invention can be venlafaxine ("EFFEXOR XR" and "EFEXOR"), nefazodone ("SERZONE"), milnacipran ("DALCIPRAN" and "IXEL"), desipramine ("N0RPRAMINE" and "PERTOFRANEIS") or duloxetine ("CYMBALTA").
The use may be of combinations of two or more SRIs, which may be for simultaneous, separate or sequential use.
The SRI may be used in combination with a further anti-inflammatory agent. Administration of the SRI and other anti-inflammatory agent can be simultaneous, separate and/or sequential The SRI, in combination with another pharmaceutical agent, can act additively or synergistically.
The other anti -inflammatory agent may be termed a non-steroidal anti-inflammatory agent (NSAID), a disease modifying anti-rheumatic drug (DMARD), a statin (including HMG-CoA reductase inhibitors such as simvastatin), a biological agent (biologicals), a steroid, an immunosuppressive agent, a salicylate and/or a microbicidal agent. Non-steroidal anti-inflammatory agents include anti-metabolite agents (including methotrexate) and anti-inflammatory gold agents (including gold sodium thiomalate, aurothiomalate or gold salts, such as auranofin). Biologicals include anti-TNF agents (including adalimumab, etanercept, infliximab, anti-EL-1 leagents, anti-IL-6 reagents, anti-B cell reagents (retoximab), anti-T cell reagents (anti-CD4 antibodies), anti-IL-15 reagents, anti-CLTA4 reagents, anti-RAGE reagents), antibodies, soluble receptors, receptor binding proteins, cytokine binding proteins, mutant proteins with altered or attenuated functions, RNAi, polynucleotide aptemers, antisense oligonucleotides or omega 3 fatty acids Steroids include cortisone, prednisolone or dexamethasone. Immunosuppressive agents include cylcospoπn, FK506, rapamycin, mycophenolic acid Salicylates include aspirin, sodium salicylate, choline salicylate and magnesium salicylate. Microbicidal agents include quinine and chloroquine. For example, the SRI may be administered in combination with one or more of an NSAID, DMARD, or immunosuppressant.
The further anti-inflammatory agent is administered by any appropriate route, for example oral (including buccal or sublingual), suppositories, topical (including buccal, sublingual or transdermal), or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route. Where the further anti-inflammatory agent is administered orally, it may be administered as part of the same composition as the SRI.
The SRIs of the present invention are those SRIs which are non-toxic to living cells.
The present invention also provides the use of a serotonin reuptake inhibitor (SRI) in the manufacture of a medicament for treating inflammatory arthritis and related conditions.
The second aspect of the invention is a kit comprising a pharmaceutical composition comprising an SRI and instructions indicating that the composition is for use in treating inflammatory arthritis and related conditions.
Compositions in accordance with the invention may be supplied as part of a sterile, pharmaceutical composition which will normally include a pharmaceutically acceptable earner. This pharmaceutical composition may be in any suitable form. It may be provided in unit dosage form and will generally be provided in a sealed container. The kit of the invention may comprise a plurality of said unit dosage forms.
The oral pharmaceutical compositions may be presented as discrete units such as capsules or tablets; as powders or granules; as solutions, syrups or suspensions (in aqueous or non-aqueous liquids; or as edible foams or whips; or as emulsions).
Suitable excipients for tablets or hard gelatine capsules include lactose, maize starch or derivatives thereof, steaπc acid or salts thereof Suitable excipients for use with soft gelatine capsules include for example vegetable oils, waxes, fats, semi-solid, or liquid polyols etc
For the preparation of solutions and syrups, excipients which may be used include for example water, polyols and sugars For the preparation of suspensions oils (e g vegetable oils) may be used to provide oil-in-water or water in oil suspensions
The pharmaceutical compositions may contain preserving agents, solubilising agents, stabilising agents, wetting agents, emulsifiers, sweeteners, colourants, odourants, salts, buffers, coating agents or antioxidants They may also contain further therapeutically active agents
Route of administration may include, parenterally (including subcutaneous, intramuscular, intravenous, by means of, for example a dπp patch), some further suitable routes of administration include (but are not limited to) oral (including buccal and sublingual), rectal, nasal, topical, infusion, vaginal, intradermal, intrapeπtoneally, intracranially, intrathecal and epidural administration or administration via oral or nasal inhalation, by means of, for example a nebuliser or inhaler, or by an implant
For administration via the oral or nasal inhalation routes the may be delivered using a mechanical form including, but not restricted to, an inhaler or nebuliser device
Further, where the oral or nasal inhalation routes are used, administration is by a SPAG (small particulate aerosol generator) may be used
Dosages of the substances of the present invention can vary between wide limits, depending upon the condition to be treated, the health of the individual to be treated, etc and a physician may determine appropπate dosages to be used The dosage may be repeated as often as appropπate The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy The amount of active ingredient which can be combined with a earner matenal to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration The amount of active ingredient which can be combined with a earner material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect
The compositions and uses described in this application are envisaged to have human, animal and veterinary applications They are preferably applicable to mammals, in particular humans but are also applicable for use in production animals, in particular sheep, cows, pigs, chickens and goats, as well as companion animals, in particular cats and dogs and sporting animals, such as horses
The third aspect of the invention is a method of treating inflammatory arthritis and related conditions comprising administration of a serotonin reuptake inhibitor In the third aspect of the invention, the method is preferably earned out on a subject in need of treatment
In the present invention, the term "treatment" includes prophylactic treatment (i e prevention) In most circumstances, prevention of inflammatory arthntis or a related condition is unlikely to be earned out Usually, it is only when the presence of inflammatory arthntis or a related condition is diagnosed in a subject that prevention means are applied However, prophylactic treatment may be appropnate if there is i) a known family history of significant inflammatory arthntis or a related condition or if tests (e g genetic tests) identify that an individual has a predisposition to inflammatory arthntis or a related condition
The preferred embodiments, as descnbed for the first aspect of the invention, are the same for other aspects of the invention mutatis mutandis
The present invention is descnbed with references to the drawings, in which
Figure 1 illustrates inhibition of spontaneous cytokine release from RA synovial membrane cultures by citalopram and fluoxetine hydrochlonde ("PROZAC") Graph (A) illustrates a dose dependent inhibition of TNF levels and IL- lβ levels by fluoxetine hydrochloπde ("PROZAC") as measured by ELISA Graph (B) illustrates a dose dependent inhibition of TNF in cells incubated with citalopram
Figure 2 illustrates inhibition of disease progression in the CIA model of rheumatoid arthritis by citalopram Graph (A) illustrates clinical score, Graph (B) illustrates histology scoπng. All statistics were calculated using a Mann- Whitney one tailed test, **=P<0.01, *=P<0.05, (n=6).
Figure 3 illustrates inhibition of disease progression in the CIA model of rheumatoid arthritis by fluoxetine hydrochloπde ("PROZAC"). (A) illustrates clinical score (B) illustrates paw swelling. All statistics were calculated using a Mann-Whitney one tailed test **=P<0.01, *=P<0.05.
Figure 4 Graph (A) illustrates inhibition by fluoxetine hydrochlonde of TNF production from human macrophages activated by LPS (10ng/ml), Flagelhn (lOng/ml), R848 (lμg/ml) and malp-2 (lOng/ml) Graph (B) illustrates TNF production in murine RAW264.7 macrophages stimulated with 20μg/ml poly IC in the presence of media alone or fluoxetine hydrochloπde Graph (C) illustrates inhibition by fluoxetine hydrochloπde of TNF production in CpG stimulated muπne RAW264 7 macrophages.
Figure 5 illustrates that inhibition of TNF production by R-848 is common to the serotonin reuptake inhibitors. Graph (A) illustrates TNF production from macrophages stimulated with lμg/ml R848 or 10ng/ml LPS in the media alone or in the presence of 30μg/ml citalopram, lOμg/ml fluoxetine hydrochloπde ("PROZAC") or 5μg/ml sertraline ("ZOLOFT") Graph (B) illustrates TNF levels from macrophages stimulated with 10ng/ml Pam3, 10ng/ml LPS, 10ng/ml Flagelhn, lμg/ml R848 and 10ng/ml malp-2 in media alone or in the presence of 5μg/ml sertraline ("ZOLOFT") Figure 6 illustrates that TLR3, 7, 8 and 9 are expressed in the rheumatoid synovial membrane cultures but only TLR3 and 8 induce TNF production Graph (A) RT-PCR illustrates the presence of TLR3, TLR7, TLR8 and TLR9 mRNA in 3 separate donors Graph (B) illustrates FACS analysis of cell surface and intracellular expression of TLR3, TLR8 and TLR9. Representative histogram plots of 3 independent donors are shown Graph (C) illustrates TNF production in rheumatoid membrane synovial cells after stimulation with 20μg/ml Poly IC (TLR3), lμg/ml imiquimod (TLR7), lμg/ml R-848 (TLR7/8) and 2μM CpG (TLR9).
Figure 7 illustrates the effect of serotonin on TNF and EL-I levels in RA membrane cell cultures Graph (A) illustrates addition of serotonin to RA membrane cell cultures for 24 hours (n=5-6). (B) Addition of chloroquine to RA membrane cell cultures for 24 hours (n=4). *=P<0.05
The present invention is descπbed with reference to the following non-limiting examples:
Examples
General Materials and Methods
Reagents
Cell culture reagents used were Penicillin-Streptomycin, RPMI 1640 and DMEM obtained from Cambrex (Belgium), Indomethacin from Sigma (USA) and FBS from PAA (Austπa). The TLR hgands used were chloroform extracted Escherichia coli (E.coh) LPS, resiquimod (R-848), muπne CpG (ODN1826), human CpG (ODN 2006) and imiquimod from Invivogen (USA), Macrophage-activating hpopeptide (Malp-2), Flagellin (puπfied) and serotonin (5-HT) hydrochloπde were from Alexis (UK) Fluoxetine hydrochloπde ("PROZAC") and chloroquine diphosphate salt, Sertraline ("ZOLOFT") and citalopram were purchased from Sigma (USA) Pam3cys- ser(lys)4 3HCl (Pam3) was from Alexis (UK) The antibodies used for FACS were FITC conjugated α-TLR3, α-TLR8 and α-TLR9 from Imgenex (USA) and a IgGl- FITC control from BD Pharmingen. Reverse transcriptase-polymerase chain reaction (RT-PCR)
RNA was isolated using the Qiagen RNA Blood isolation kit (Qiagen, Valencia, CA) according to the manufacturer's instructions. Total RNA was reverse transcribed with Superscript II RNase H" reverse transcriptase (Gibco Life Technologies, Carlsbad,
CA) and oligo (dT) primer. For human TLR3 amplification, the primers 5'-
GCAAACACAAGCATTCGGAATCT-3' and 5'-
TTGAAGGCTTGGGACCAAGGCA-S1 were used with an annealing temperature of
62°C. For human TLR7 amplification, the primers 5'- TCTACCTGGGCCAAAACTGTT-3' and 5'- GGCAC ATGCTGAAGAG AGTTA-3' were used with an annealing temperature of 6O0C. For human TLR8 amplification, the primers 5'- CCG ACTTGG A AGTTCTAG ATC-31 and 51-
AATGCTTCATTTGGGATGTGCT-3' were used with an annealing temperature of
600C. For human TLR9 amplification, the primers 5'- GTGCCCCACTTCTCCATG- 31 and 5'- GGCACAGTC ATGATGTTGTTG-31 were used with an annealing temperature of 550C. Amplification was performed in a Dyad PCR machine (MJ
Instruments). Subsequent PCR amplification consisted of 35 cycles.
ELISA (enzyme-linked immunosorbent assay) Macrophages or RA synovial membrane cell cultures were stimulated with lOng/ml chloroform-extracted LPS, 20μg/ml poly IC, 2μM CpG (murine or human), 10ng/ml Pam3, 10ng/ml Flagellin, 10ng/ml Malp-2 or lμg/ml R-848 in complete media for 6h. Supernatants from RA cultures were harvested after 24 hours. AU reagents other than LPS were free from LPS contamination as assessed using the limulus amebocyte lysate (LAL) assay from Cambrex (USA).
Sandwich ELISAs were employed to measure TNF, (Pharmingen, UK), IP-10 (R&D, USA) and IL- lβ (Bioscource, USA). Absorbance was read on a spectrophotometric ELISA plate reader (Labsystems Multiscan Biochromic) and analyzed using Ascent software V2.6 (Thermo Labsystems, Cambridge, United Kingdom). All results are expressed as the mean cytokine concentration ± SD obtained from triplicate cultures per condition. Cell viability was not significantly affected over this time period when examined by the 3-[4,5 dimethylthiazol-2-yl]-2,5-diphenyl-tetrazolium bromide (MTT) assay (Sigma).
Statistical methods Mean, standard deviation (SD) and statistical significance were calculated using GraphPad version 3 (GraphPad Software Inc., USA). For statistical analysis, a one tailed student's t-test of paired data was used with a 95% confidence interval and the Mann-Whitney test.
Cell culture
RA synovial membrane cells were isolated from patients undergoing joint replacement surgery as previously descπbed (Foxwell et al., Proc. Natl. Acad. Sci. USA 95,8211-8215, 1998). All patients gave wπtten informed consent and the study was approved by the local ethics committee. Immediately after isolation, cells were used for mRNA analysis, stained by FACS or cultured at IxIO5 cells/well in 96-well tissue culture plates (Falcon, UK) in RPMI 1640 containing 10% (v/v) FBS and 100 U/ml penicillin/streptomycin. Peπpheral blood monocytes were isolated and cultured as previously descπbed (Foxwell et al., Proc. Natl. Acad. Sci. USA 95,821 1-8215, 1998). Macrophages were deπved from monocytes after differentiation for 4 days with 100 ng/ml M-CSF (PeproTech, UK). The mouse macrophage cell line RAW 264.7 was maintained in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum.
Example 1 Rheumatoid arthritis synovial membrane cells were cultured for 24 hours in the presence of media alone or media containing 5μg/ml orlOμg/ml fluoxetine hydrochloπde ("PROZAC"). Supernatants were harvested and levels of cytokines (A) TNF or IL- 1 were measured by ELISA Addition of fluoxetine hydrochloπde ("PROZAC") at the time of culture led to a dose dependent decrease in the levels of TNF and IL-I (Figure 1, Graph A). Rheumatoid arthritis synovial membrane cells were also cultured in the presence of media alone or media containing lOμg/ml, 20μg/ml and 30μg/ml citalopram and levels of TNF were measured. TNF production was dose dependently inhibited in cells treated with citalopram (see Figure IB).
To check if the ability of citalopram and fluoxetine hydrochloride to inhibit these cytokines was due to increased serotonin in the cultures, the cells were incubated with 10 or lOOμM serotonin for 24 hours. After 24 hours supernatants were collected and TNF and IL-I levels determined (n=5). The serotonin had no effect on the amount of TNF (Figure 7, Graph A) produced from these cultures, but increased the amount of IL-I being produced at the higher dose of serotonin (Figure 7, graph A).
These results illustrate that citalopram inhibits the spontaneous release of TNF and fluoxetine hydrochloride inhibits the spontaneous release of TNF and EL- lβ from rheumatoid synovial membrane cultures.
Example 2
The murine CIA model has been shown to be clinically similar to human rheumatoid arthritis with a comparable synovitis, bone erosion and panus formation.
Male DBA mice (8-10 weeks of age) were immunized at the base of the tail intradermally with 200μg type II collagen emulsified in CFA (Difco Laboratories, West Molesey, U.K.). The onset of arthritis was considered to be the day that erythema and/or swelling were first observed, and arthritic mice were given a daily clinical score per limb from 0 to 3, with 0 = normal, 1 = slight erythema and swelling, 2 = pronounced edematous swelling and 3 = joint deformity with ankylosis, resulting in a maximum score of 12 per animal. Citalopram or fluoxetine hydrochloride treatment started on the day of onset and was administered daily for 7 days.
The experiment was started at the onset of arthritis (day 1). Mice were given an interperitoneal injection of vehicle (PBS) or 25mg/kg of citalopram or fluoxetine hydrochloπde once a day for 7 days Mice were assessed for (Figures 2A and 3A) clinical score on a daily basis.
Results after treatment with citalopram Mice treated with citalopram at the day of onset of arthritis (day 1) exhibited an attenuated disease progression over a 7 day peπod. Disease pathogenesis was assessed by measuring clinical score The mice treated with citalopram showed a significant improvement to the control group from day 2-3 onwards (Figure 2, Graph A) Histology samples taken from the hind paws of mice treated with citalopram showed a decreased histology score over the control group (Figure 2B).
These results illustrate that citalopram inhibits disease progression in the CIA model of rheumatoid arthritis.
Results after treatment with fluoxetine hydrochloride
Mice treated with fluoxetine hydrochloπde at the day of onset of arthritis (day 1) exhibited an attenuated disease progression over a 7 day peπod. Disease pathogenesis was assessed by measuπng clinical score. The mice treated with fluoxetine hydrochloπde showed a significant improvement to the control group (Figure 3, Graph A, B). This effect of fluoxetine hydrochloπde was dose dependent as 10mg/kg showed a significant improvement throughout the study from the vehicle control, whilst the lOmg/kg group showed a small but not significant improvement (Figure 3, Graph A). This was also reflected in the measurement of hind paw swelling that also showed a significant improvement throughout the study for both the 10mg/kg and 20mg/kg treated groups (Figure 3, Graph B).
These results illustrate that fluoxetine hydrochloπde inhibits disease progression in the CIA model of rheumatoid arthπtis
Example 3
Pπmary human macrophages and munne RAW 264 7 cells were incubated with fluoxetine hydrochlonde for 30 minutes and then stimulated for 6 hours with TLR ligands (R-848, LPS, flagellin, malp-2, poly IC or CpG). Human macrophages showed a decreased production of TNF in response to R-848 but not LPS, flagellin or Malp-2 in the presence of fluoxetine hydrochloride (Figure 4, Graph A).
Murine RAW 264.7 cells were used to examine poly IC and CpG induced TNF, as unlike human macrophages they respond to poly IC a TLR3 ligand and CpG, a TLR9 ligand to produce TNF. Fluoxetine hydrochloride was able to inhibit TNF production in these cells on stimulation with poly IC (Figure 4, Graph B) and CPG (Figure 4, Graph C).
This shows that signaling from toll-like receptors (TLRs) 3, 8 and 9 are selectively inhibited by fluoxetine hydrochloride and that it does not have general antiinflammatory properties as it was unable to inhibit TNF stimulated by activation of the other TLRs.
Example 4
Selective inhibition of TLRs by fluoxetine hydrochloride is also observed for other anti-depressants (SRIs). Citalopram, fluoxetine hydrochloride ("PROZAC") and sertraline ("ZOLOFT") all decreased TNF production in human macrophages stimulated by R848, but did not inhibit LPS induced TNF in the same cells (Figure 5, Graph A). Figure 5B illustrates that sertraline ("ZOLOFT"), like fluoxetine hydrochloride selectively inhibits TLR8 induced TNF in human macrophages (Figure 5, Graph B).
Example 5
TLR3, 7, 8 and 9 are expressed in human RA synovial tissue. mRNA from synovial tissue was extracted, DNAase treated and then analysed by RT-PCR. Expression of message for TLR3, 7, 8 and 9 was detected by RT-PCR from the mixed population of cells in human rheumatoid synovial tissue in three unrelated donors 1-3 (Figure 6, Graph A). TLR3, 8 and 9 were further investigated using FACS. Cells were washed, then blocked with 10% human serum in PBS containing 0.01% azide for 30 minutes at 4°C or for intracellular staining cells were fixed in 2% paraformaldehyde and permeabilised with 0.1% saponin (Sigma, USA) before blocking. Cells were then incubated with FITC conjugated α-TLR3, α-TLR8, α-TLR9 or isotype controls for lhr at 40C and then washed before analysis on a Becton-Dickinson LSR flow cytometer. A directly conjugated antibody was not available to examine TLR7 expression.
TLR3, 8 and 9 protein expression was detected at low levels on the cell surface but was more readily detected upon intracellular staining (Figure 6, Graph B).
To check if these TLRs were functional in synovial membrane cultures, cells were stimulated with the appropriate TLR ligands at the time of seeding the cells (Figure 6, Graph C). These cells spontaneously release cytokines into the media. TNF production was increased on stimulation of cells with Poly IC for TLR3 (334% ±60 P=0.0002, n=12) and R-848 for TLR7/8 (515% ±76 P<0.0001, n=l l), but imiquimod, a TLR 7 ligand and CpG a TLR9 ligand (see Figure 6, Graph C) were unable to stimulate further release of cytokines over the spontaneous levels. This demonstrates that TLR 3 and 8 are both functional in rheumatoid synovial membrane cultures.
In summary, these results shows that TLRs 3, 7, 8 and 9 are expressed in rheumatoid membrane synovial cells but only TLRs 3 and 8 functionally induce increased TNF production in rheumatoid membrane synovial cells.
Example 6
The main mechanism of action of the antidepressants (SRIs) used in this study is to increase the amount of serotonin by inhibiting serotonin reuptake receptors. As we have shown that two serotonin reuptake inhibitors suppress TNF production from human rheumatoid synovial membrane cultures, serotonin was tested in these cultures. At the time of seeding the cells 10 or lOOμM serotonin was added to the culture media Supernatants were collected after 24 hours and the levels of TNfF and IL-I were compared with the spontaneously produced levels in the culture Serotonin had no effect on TNF levels (n=6) (Figure 7, Graph A) but significantly increased the levels of IL-I (n=5) (Figure 7, Graph A).
Fluoxetine hydrochloπde ("PROZAC"), citalopram and sertraline have been demonstrated as being able to inhibit selective TLRs TLR3, 7, 8 and 9 are classically viewed as having an endosomal localization. Chloroquine has been reported to inhibit the function of TLR3, 7, 8 and 9, by increasing the pH inside vesicles, which is essential for functional signaling from TLR3, 7, 8 and 9. To confirm that the inhibition of cytokine release from RA cultures may be through the ability of these SRIs to inhibit signaling through endosomal TLRs, we also measured cytokine release in cells incubated with chloroquine There was a dose dependant effect of chloroquine with the best inhibition being observed at lOOμM, both TNF and EL-I release were inhibited (Figure 7, Graph B).
Thus, chloroquine inhibits spontaneous TNF and EL-I release but serotonin does not

Claims

Claims
1 A serotonin reuptake inhibitor (SRI) for treating inflammatory arthritis or a related condition
2 The serotonin reuptake inhibitor according to claim 1, wherein the serotonin reuptake inhibitor is a selective serotonin reuptake inhibitor or a serotonin norepinephπne reuptake inhibitor
3 The serotonin reuptake inhibitor according to claim 1, wherein the serotonin reuptake inhibitor is fluoxetine, citalopram or sertraline
4 The serotonin reuptake inhibitor according to any one of claims 1 to 3, wherein the inflammatory arthritis is rheumatoid arthritis
5 The serotonin reuptake inhibitor according to any one of claims 1 to 4, wherein the serotonin reuptake inhibitor is administered in combination with a further anti-inflammatory agent
6 The serotonin reuptake inhibitor according to claim 5, wherein the further anti-inflammatory agent is a non-steroidal anti-inflammatory agent (NSADD), a statin, a disease modifying anti-rheumatic drug (DMARD), a biological agent, a steroid, RNAi, aptemers, an immunosuppressive agent, a salicylate and/or a microbicidal agent
7 Use of a serotonin reuptake inhibitor (SRI) in the manufacture of a medicament for treating inflammatory arthritis or a ielated condition
8 A kit composing a pharmaceutical composition compπsing a serotonin reuptake inhibitor (SRI) and instructions indicating that the composition is for use in treating inflammatory arthritis or a related condition
9. The kit of claim 8, wherein the serotonin reuptake inhibitor is a selective serotonin reuptake inhibitor or a serotonin norepinephπne reuptake inhibitor
10 The kit of claim 8, wherein the serotonin reuptake inhibitor is fluoxetine, citalopram or sertraline
11 This kit according to any one of claims 8 to 10, wherein the inflammatory arthritis is rheumatoid arthritis.
12 The kit according to any one of claims 8 to 11 wherein the serotonin reuptake inhibitor is administered in combination with a further anti-inflammatory agent.
13. The kit according to claim 12, wherein the further anti-inflammatory agent is a non-steroidal anti-inflammatory agent (NSAED), a statin, a disease modifying anti- rheumatic drug (DMARD), a biological agent, a steroid, an immunosuppressive agent, a salicylate and/or a microbicidal agent
14. A method of treating inflammatory arthritis or a related condition in a subject, comprising administration of a serotonin reuptake inhibitor to the subject.
15. The method of claim 14, wherein the serotonin reuptake inhibitor is a selective serotonin ieuptake inhibitor or a serotonin norepinephπne reuptake inhibitor
16 The method of claim 14, wherein the serotonin reuptake inhibitor is fluoxetine, citalopram or sertraline
17 The method of any one of claims 14 to 16 wherein the subject is a mammal.
18 The method of claim 17, wherein the mammal is a human.
19 The method of any one of claims 14 to 18, wherein the inflammatory arthritis is rheumatoid arthritis. 20 The method according to any one of claims 14 to 19, wherein the serotonin reuptake inhibitor is administered in combination with a further anti-inflammatory agent
21 The method according to claim 20, wherein the further anti-inflammatory agent is a non-steroidal anti -inflammatory agent (NSAID), a statin, a disease modifying anti-rheumatic drug (DMARD), a biological agent, a steroid, an immunosuppressive agent, a salicylate and/or a microbicidal agent
PCT/GB2008/000217 2007-01-22 2008-01-22 Serotonin reuptake inhibitors for treating arthritis WO2008090331A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB0701171.1A GB0701171D0 (en) 2007-01-22 2007-01-22 Compositions and uses thereof
GB0701171.1 2007-01-22

Publications (1)

Publication Number Publication Date
WO2008090331A1 true WO2008090331A1 (en) 2008-07-31

Family

ID=37846742

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2008/000217 WO2008090331A1 (en) 2007-01-22 2008-01-22 Serotonin reuptake inhibitors for treating arthritis

Country Status (2)

Country Link
GB (1) GB0701171D0 (en)
WO (1) WO2008090331A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009019473A1 (en) * 2007-08-08 2009-02-12 Imperial Innovations Limited Treatments for inflammatory arthritis
WO2010029958A1 (en) 2008-09-11 2010-03-18 アステラス製薬株式会社 Novel pharmaceutical composition for treatment of nociceptive pain
RU2703723C1 (en) * 2018-08-06 2019-10-22 Федеральное Государственное бюджетное образовательное учреждение высшего образования Дагестанский государственный медицинский университет Министерства здравоохранения Российской Федерации Даггосмедуниверситет Method of correction of anxiety-depressive disorders in patients with systemic connective tissue diseases

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004030618A2 (en) * 2002-09-24 2004-04-15 Combinatorx, Incorporated Methods and reagents for the treatment of diseases and disorders associated with increased levels of proinflammatory cytokines
US20050014829A1 (en) * 2003-05-23 2005-01-20 Julius Remenar Sertraline compositions
WO2005027839A2 (en) * 2003-09-15 2005-03-31 Combinatorx, Incorporated Methods and reagents for the treatment of immunoinflammatory disorders
US20060270713A1 (en) * 2003-06-11 2006-11-30 Beadle Christopher D 3-Aminopyrrolidines as inhibitors of monoamine uptake
WO2007000465A1 (en) * 2005-06-28 2007-01-04 Neurosearch A/S Novel 3-aza-spiro[5.5]undecane derivatives and their use as monoamine neurotransmitter re-uptake inhibitors

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004030618A2 (en) * 2002-09-24 2004-04-15 Combinatorx, Incorporated Methods and reagents for the treatment of diseases and disorders associated with increased levels of proinflammatory cytokines
US20050014829A1 (en) * 2003-05-23 2005-01-20 Julius Remenar Sertraline compositions
US20060270713A1 (en) * 2003-06-11 2006-11-30 Beadle Christopher D 3-Aminopyrrolidines as inhibitors of monoamine uptake
WO2005027839A2 (en) * 2003-09-15 2005-03-31 Combinatorx, Incorporated Methods and reagents for the treatment of immunoinflammatory disorders
WO2007000465A1 (en) * 2005-06-28 2007-01-04 Neurosearch A/S Novel 3-aza-spiro[5.5]undecane derivatives and their use as monoamine neurotransmitter re-uptake inhibitors

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
KUBERA MARTA ET AL: "Effect of repeated desipramine and fluoxetine administration on post-adjuvant arthritis", POLISH JOURNAL OF PHARMACOLOGY, vol. 52, no. 3, May 2000 (2000-05-01), pages 229 - 235, XP009098348, ISSN: 1230-6002 *
MITRA A ET AL: "Role of anti-depressant fluoxetine in the puva treatment of psoriasis vulgaris.", INDIAN JOURNAL OF DERMATOLOGY, VENEREOLOGY AND LEPROLOGY 2003 MAR-APR, vol. 69, no. 2, March 2003 (2003-03-01), pages 168 - 169, XP009098350, ISSN: 0973-3922 *
PETITTO J M ET AL: "Improvement of arthritis with fluoxetine.", PSYCHOSOMATICS SUMMER 1992, vol. 33, no. 3, July 1992 (1992-07-01), pages 338 - 341, XP009098357, ISSN: 0033-3182 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009019473A1 (en) * 2007-08-08 2009-02-12 Imperial Innovations Limited Treatments for inflammatory arthritis
WO2010029958A1 (en) 2008-09-11 2010-03-18 アステラス製薬株式会社 Novel pharmaceutical composition for treatment of nociceptive pain
RU2703723C1 (en) * 2018-08-06 2019-10-22 Федеральное Государственное бюджетное образовательное учреждение высшего образования Дагестанский государственный медицинский университет Министерства здравоохранения Российской Федерации Даггосмедуниверситет Method of correction of anxiety-depressive disorders in patients with systemic connective tissue diseases

Also Published As

Publication number Publication date
GB0701171D0 (en) 2007-02-28

Similar Documents

Publication Publication Date Title
TWI387452B (en) Methods for treating sexual dysfunction
US20180110796A1 (en) Compositions and methods for the treatment of hbv infection
US10307401B2 (en) Compositions and methods for treatment of prostate cancer
US10695341B2 (en) Compositions and methods for treating endometriosis
WO2008090331A1 (en) Serotonin reuptake inhibitors for treating arthritis
US20100273750A1 (en) Serotonin receptor antagonists for treating arthritis
WO2009019473A1 (en) Treatments for inflammatory arthritis
US11622954B2 (en) Estrogen receptor ligands, compositions and methods related thereto
AU2016308704B2 (en) MDM2 inhibitors for treating uveal melanoma
EP2735316B1 (en) New therapeutic approaches for treating neuroinflammatory conditions
US20130289023A1 (en) Method for treating brain tumor
US20140378502A1 (en) Par1 Inhibitors for Use in the Treatment or Prevention of Paramyxoviridae Infections
US20230310408A1 (en) Tlr7 inhibitor in combination with prednisolone or hydroxychloroquine for treating cutaneous lupus erythematosus
US20210338685A1 (en) Agents and methods for modulating pathogen activity
WO2007010946A1 (en) Synovial cell proliferation inhibitor
WO2014137978A1 (en) Treatment of lymphangioleiomyomatosis
CN115364102A (en) Application of pyrrolopyrimidine compound
JP2007326824A (en) Synoviocyte-growth inhibitor, pharmaceutical composition and therapy using the same

Legal Events

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

Ref document number: 08701891

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08701891

Country of ref document: EP

Kind code of ref document: A1