WO2015197193A2 - Nouvelle utilisation de molécules inhibitrices de jnk pour le traitement de diverses maladies - Google Patents

Nouvelle utilisation de molécules inhibitrices de jnk pour le traitement de diverses maladies Download PDF

Info

Publication number
WO2015197193A2
WO2015197193A2 PCT/EP2015/001293 EP2015001293W WO2015197193A2 WO 2015197193 A2 WO2015197193 A2 WO 2015197193A2 EP 2015001293 W EP2015001293 W EP 2015001293W WO 2015197193 A2 WO2015197193 A2 WO 2015197193A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
jnk inhibitor
sequence
disease
syndrome
Prior art date
Application number
PCT/EP2015/001293
Other languages
English (en)
Other versions
WO2015197193A3 (fr
Inventor
Jean-Marc Combette
Catherine Deloche
Original Assignee
Xigen Inflammation Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/EP2014/001737 external-priority patent/WO2014206564A1/fr
Priority claimed from PCT/EP2014/002723 external-priority patent/WO2015197097A1/fr
Application filed by Xigen Inflammation Ltd. filed Critical Xigen Inflammation Ltd.
Priority to JP2016575150A priority Critical patent/JP6843625B2/ja
Priority to EP15734295.7A priority patent/EP3160989A2/fr
Priority to US15/321,904 priority patent/US20170128516A1/en
Publication of WO2015197193A2 publication Critical patent/WO2015197193A2/fr
Publication of WO2015197193A3 publication Critical patent/WO2015197193A3/fr
Priority to US16/430,697 priority patent/US11331364B2/en
Priority to US17/719,068 priority patent/US20220280591A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/55Protease inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/03Peptides having up to 20 amino acids in an undefined or only partially defined sequence; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4703Inhibitors; Suppressors
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to the field of enzyme inhibition, in particular to (poly-)peptide inhibitors of c-Jun amino terminal kinase (JNK).
  • JNK c-Jun amino terminal kinase
  • the present invention relates to using these JNK inhibitors in the treatment of various diseases.
  • the c-Jun amino terminal kinase (JNK) is a member of the stress-activated group of mitogen-activated protein (MAP) kinases. These kinases have been implicated in the control of cell growth and differentiation, and, more generally, in the response of cells to environmental stimuli.
  • MAP mitogen-activated protein
  • the JNK signal transduction pathway is activated in response to environmental stress and by the engagement of several classes of cell surface receptors.
  • JNK cytokine receptors
  • serpentine receptors receptor tyrosine kinases.
  • receptors can include cytokine receptors, serpentine receptors and receptor tyrosine kinases.
  • JNK has been implicated in biological processes such as oncogenic transformation and mediating adaptive responses to environmental stress. JNK has also been associated with modulating immune responses, including maturation and differentiation of immune cells, as well as effecting programmed cell death in cells identified for destruction by the immune system. This unique property makes JNK signaling a promising target for developing pharmacological intervention. Among several neurological disorders, JNK signaling is particularly implicated in ischemic stroke and Parkinson's disease, but also in other diseases as mentioned further below.
  • mitogen-activated protein kinase (MAPK) p38alpha was shown to negatively regulate the cell proliferation by antagonizing the JNK-c-Jun-pathway.
  • the mitogen-activated protein kinase (MAPK) p38alpha therefore appears to be active in suppression of normal and cancer cell proliferation and, as a further, demonstrates the involvement of JNK in cancer diseases (see e.g. Hui et al., Nature Genetics, Vol 39, No. 6, June 2007).
  • JNK c-Jun N-terminal Kinase
  • JNK signalling in diseases such as the involvement in excitotoxicity of - - hi ppocampal neurons, liver ischemia, reperfusion, neurodegenerative diseases, hearing loss, deafness, neural tube birth defects, cancer, chronic inflammatory diseases, obesity, diabetes, in particular insulin-resistant diabetes, and proposed that it is likely that selective JNK inhibitors are needed for treatment of various diseases with a high degree of specificity and lack of toxicity.
  • In hibition or interruption of the JNK signalling pathway is thus a promising approach in combating di sorders strongly related to JNK signalling.
  • In hibitors of the JNK signaling pathway include e.g. upstream kinase inhibitors (for example, CEP-1 347), small chemical inhibitors of JNK (SP600125 and AS601245), which directly affect kinase activity e.g. by competing with the ATP-binding site of the protein kinase, and peptide inhibitors of the interaction between JNK and its substrates (see e.g.
  • WO 2007/031280 discloses small cell permeable fusion peptides, comprising a so-called TAT transporter sequence derived from the basic trafficking sequence of the HIV-TAT protein and an amino acid inhibitory sequence of IB1 .
  • WO 2007/031280 discloses in particular two specific sequences, L-TAT-IB1 (GRKKRRQRRRPPRPKRPTTLNLFPQVPRSQD, herein SEQ ID NO: 196) and D-TAT-IB1 (dqsrpvqpflnlttprkprpprrrqrrkkrg; herein SEQ ID NO: 1 97), the latter being the retro-inverso sequence of L-TAT-IB1 . Due to the HIV TAT derived transporter sequence, these fusion peptides are more efficiently transported into the target cells, where they remain effective until proteolytic degradation.
  • ATP independent peptide inhibitors of JNK are usually more specific inhibitors, they are frequently the first choice if it comes to inhibiting JNK.
  • peptide inhibitors disclosed in WO 2007/031280 are not optimal for all purposes.
  • compound L-TAT-IB1 (herein SEQ ID NO: 1 96) which consists of L amino acids only, is quickly proteolytically degraded.
  • D-TAT-IB1 (herein SEQ ID NO: 1 97), which comprises D amino acids.
  • D-TAT-IB1 exhibits the retro-inverso sequence of L-TAT-IB1 .
  • JNK inhibitors have been discussed, proposed and successfully tested in the art as treatment for a variety of disease states.
  • i n 1 997 Dickens et al. described the c-Jun amino terminal kinase i n hibitor JI P-1 and proposed JI P-1 as candidate compounds for therapeutic strategies for the treatment of for example chronic myeloid leukaemia, i n particular, in the context of Bcr-Abl caused transformation of pre-B-cel ls (Science; 1 997; 277(5326):693-696).
  • JIP-1 derived i nhibitors of JNK signal ling are proposed for the treatment of neurodegenerative diseases, such as Parki nson's disease or Alzheimer's disease; stroke and associated memory loss, autoimmune diseases such as arthritis; other conditions characterized by inflammation; malignancies, such as leukemias, e.g. chronic myelogenous leukemia (CML); oxidative damage to organs such as the l iver and kidney; heart diseases; and transplant rejections.
  • neurodegenerative diseases such as Parki nson's disease or Alzheimer's disease
  • stroke and associated memory loss autoimmune diseases such as arthritis
  • other conditions characterized by inflammation malignancies, such as leukemias, e.g. chronic myelogenous leukemia (CML); oxidative damage to organs such as the l iver and kidney; heart diseases; and transplant rejections.
  • CML chronic myelogenous leukemia
  • JNK c-Jun N-termi nal ki nase
  • the authors of said study used a peptide i nhibitor, D-J NKI-I, a two domain peptide contai ning a 20 amino acid sequence of the minimal J NK-binding domain of islet-brain- 1 /JNK-i nteracti ng protein-1 , l inked to a 1 0 amino acid TAT sequence of the human immuno deficiency virus TAT protein that mediates intracellular translocation.
  • the authors have concluded that a reduction i n JNK activity and phosphorylation due to the presence of said inhibitor is i mportant i n the preservation of cardiac function i n rats in the phase of ischemia and apoptosis.
  • Pirvola et al. (The Journal of Neuroscience, 2000, 20(1 ); 43 - 50) described the rescue of hearing, auditory hair cel ls and neurons by CEP-1 347/KT751 5, an inhibitor of c-Jun-N- terminal kinase activation.
  • the authors suggested in general that therapeutic intervention i n the JNK signal ling cascade may offer opportunities to treat i nner ear i njuries.
  • Treatment of hearing loss by means of administering JNK-inhibitory peptides is also disclosed for example in WO 03/1 03698.
  • Roduit et al . (Apoptosis, 2008, 1 3(3), p. 343 - 353) have l ikewise suggested to use JNK-i nhibition as therapeutic approach.
  • Si mi lar considerations relying on JNK-inhibition are disclosed for example in WO 201 0/1 1 3753 for the treatment of age-related macular degeneration, diabetic macular edema, diabetic retinopathy, central exudative chorioreti nopathy, angioid streaks, retinal pigment epithel ium detachment, multifocal choroiditis, neovascular maculopathy, retinopathy of prematurity, retinitis pigmentosa, Leber's disease, retinal artery occlusion, retinal vei n occlusion, central serous chorioretinopathy, retinal macroaneurysm, retinal detachment, proliferative vitreoretinopathy, Stargardt's disease, - - choroidal sclerosis, chorioderemia, vitelliform macular dystrophy, Oguchi's disease, fundus albipunctatus, retinitis punctata alb
  • the problem to be solved by the present invention was to provide further (peptide) inhibitors of JNK for the treatment of specific diseases.
  • the object of the present invention is solved by the inventor by means of the subject-matter set out below and in the appended claims.
  • Fig.1 Illustration of the inhibitory efficacy of several JNK inhibitors according to the present invention, which was investigated by in vitro AlphaScreen assay (Amplified Luminescence Proximity Homogeneous-Screen Assay).
  • Fig.l A Inhibition of JNK1 by SEQ ID NOs: 193, 2, 3, 5, 6, and 7.
  • Fig.lB Inhibition of JNK2 by SEQ ID NOs: 193, 2, 3, 5, 6, and 7.
  • Fig.lC Inhibition of JNK3 by SEQ ID NOs: 193, 2, 3, 5, 6, and 7.
  • Fig.2 Table illustrating the inhibitory efficacy of several JNK inhibitors (SEQ ID NOs: 193, 2, 3, 5, 6, and 7) according to the present invention. Given are the IC50 values in the nM range, the respective standard error of the mean and the number of experiments performed (n).
  • Fig.3 Illustration of the inhibitory efficacy of several JNK inhibitors according to the present invention, which are fusion proteins of a JNK inhibitory (poly-)peptide sequence and a transporter sequence.
  • the inhibitory efficacy was determined by means of in vitro AlphaScreen assay (Amplified Luminescence Proximity Homogeneous-Screen Assay).
  • Fig.3A Inhibition of JNK1 by SEQ ID NOs: 194, 195, 172, 200, 46, 173, 174, 175, 176, 177, 178, 179, 180, 181 and 197.
  • Fig.3B Inhibition of JNK2 by SEQ ID NOs: 194, 195, 172, 200, 46, 173, 174, 175, 176, 177,
  • Fig.3C Inhibition of JNK3 by SEQ ID NOs: 194, 195, 172, 200, 46, 173, 174, 175, 176,
  • Fig.3D Inhibition of JNK1 by SEQ ID NOs: 194, 195, 172, 200, 46, 182, 183, 184, 185, 186, 187, 188, 189, 190 and 197.
  • Fig.3E Inhibition of JNK2 by SEQ ID NOs: 194, 195, 172, 200, 46, 182, 183, 184, 185, 186,
  • Fig.3F Inhibition of JNK3 by SEQ ID NOs: 194, 195, 172, 200, 46, 182, 183, 184, 185, 186, 187, 188, 189, 190 and 197. . .
  • Fig. 4 Table illustrating the inhibitory efficacy of several JNK inhibitors according to the present invention, which are fusion proteins of a JNK inhibitory (poly-)peptide sequence and a transporter sequence. Given are the IC50 values in the nM range, the respective standard error of the mean (SEM) and the number of experiments performed (n).
  • Fig. 5 Stability of JNK inhibitors with SEQ ID NOs: 1 72, 1 96 and 197 in 50% human serum.
  • the JNK inhibitor with SEQ ID NO: 1 96 was totally degraded into amino acids residues within 6 hours (A).
  • the JNK inhibitor with SEQ ID NO: 1 72 was completely degraded only after 14 days (B).
  • the JNK inhibitor with SEQ ID NO: 1 97 was stable at least up to 30 days (B).
  • Fig. 6 shows internalization experiments using TAT derived transporter constructs with D-amino acid/L-amino acid pattern as denoted in SEQ ID NO: 30.
  • the transporter sequences analyzed correspond to SEQ ID NOs: 52-94 plus SEQ ID NOs: 45, 47, 46, 43 and 99 (Fig 6a) and SEQ ID NOs: 100-147 (Fig. 6b).
  • SEQ ID NO: 31 all transporters with the consensus sequence rXXXrXXXr (SEQ ID NO: 31 ) showed a higher internalization capability than the L-TAT transporter (SEQ ID NO: 43).
  • Hela cells were incubated 24 hours in 96well plate with 1 0mM of the respective transporters.
  • the cells were then washed twice with an acidic buffer (0.2M Glycin, 0.1 5M NaCI, pH 3.0) and twice with PBS. Cells were broken by the addition of RIPA lysis buffer. The relative amount of internalized peptide was then determined by reading the fluorescence intensity (Fusion Alpha plate reader; PerkinElmer) of each extract followed by background subtraction.
  • Fig. 7 The JNK inhibitor with the sequence of SEQ ID NO: 1 72 blocks LPS-induced cytokine and chemokine release in THP1 -PMA-differentiated macrophages.
  • Fig. 7A TNF release (THP1 pma 6h 3ng/ml LPS);
  • Fig. 7B TNF-a release (THP1 pma 6h 1 0ng/ml LPS);
  • Fig. 7C IL 6 release (THPI pma 6h 1 0ng/ml LPS);
  • Fig. 7D MCP1 release (THPI pma 6h 3ng/ml LPS).
  • Fig. 8 The JNK inhibitor of SEQ ID NO: 1 72 blocks LPS-induced IL6 release in THP1 differentiated macrophages with higher potency than D-TAT-IB1 (SEQ ID NO: 1 97), dTAT (SEQ ID NO: 45) and SP 600125. LPS was added for 6h (10 ng/ml).
  • the JNK inhibitor of SEQ ID NO: 1 72 blocks LPS-induced TNFa release in THP1 differentiated macrophages with higher potency than D-TAT-IB1 (SEQ ID NO: 1 97), dTAT (SEQ ID NO: 45) and SP 600125. LPS was added for 6h (10 ng/ml). - -
  • the JNK inhibitor of SEQ ID NO: 1 72 blocks LPS-induced IL-6 release in PMA differentiated macrophages with higher potency than D-TAT-IB1 (SEQ ID NO: 197) and L-TAT-IB1 (SEQ ID NO: 1 96). LPS was added for 6h.
  • Fig. 1 1 The JNK inhibitor of SEQ ID NO: 1 72 blocks LPS-induced TNFa release in PMA differentiated macrophages with higher potency than D-TAT-IB1 (SEQ ID NO: 197) and L- TAT-IB1 (SEQ ID NO: 196).
  • Fig. 12 The JNK inhibitor of SEQ ID NO: 1 72 blocks LPS-induced TNFa release in Primary Rat Whole Blood Cells at 3 ng/ml. Given are the results for the control, 1 ⁇ of SEQ ID NO:
  • Fig. 13 The JNK inhibitor of SEQ ID NO: 1 72 blocks IL-2 secretion by primary human T-cells in response to PMA/lonomycin.
  • Fig. 14 The JNK inhibitor of SEQ ID NO: 1 72 blocks IL-2 secretion by primary human T-cells in response to CD3/CD28 stimulation.
  • the JNK inhibitors used are indicated by their SEQ ID NO: 1 72 and 1 97.
  • Fig. 1 5 Dose-dependent inhibition by JNK inhibitor with SEQ ID NO: 1 72 of CD3/CD28-induced IL-2 release in primary rat lymph-nodes purified T cells. Control rat were sacrificed and lymph-nodes were harvested. T cells further were purified (using magnetic negative selection) and plated into 96-well plates at 200.000 cells/well. Cells were treated with anti- rat CD3 and anti-rat CD28 antibodies (2 g/mL). JNK inhibitor with SEQ ID NO: 1 72 was added to the cultures 1 h before CD3/CD28 treatment and IL-2 release was assessed in supernatant 24h after treatment.
  • Fig. 1 6 Dose-dependent inhibition of CD3/CD28-induced IL-2 release in primary rat lymph nodes purified T cells: Comparison of several JNK inhibitors, namely SEQ ID NOs: 1 72, 1 97 and
  • Fig. 1 7 Dose dependent inhibition of IL-2 release in rat whole blood stimulated with PMA + ionomycin.
  • JNK inhibitor with SEQ ID NO: 1 72 was added at three different concentrations, namely 1 , 3 and 10 ⁇ 1 h before stimulation with PMA + ionomycin.
  • Three doses of activators were added (25/500 ng/mL, 50/750 ng/mL and 50/1000 ng/mL) for 4h.
  • IL-2 release - - was assessed in supernatant.
  • JNK inhibitor with SEQ ID NO: 172 at 10 ⁇ did efficiently reduce PMA-iono-induced IL-2 release at the three tested activator concentrations.
  • the JNK inhibitor with SEQ ID NO: 172 did reduce the LPS-induced IL-6 release in a dose-dependent manner.
  • JNK inhibition and IL-2 release in human whole blood The JNK inhibitor with SEQ ID NO:
  • the JNK inhibitor with SEQ ID NO: 172 did reduce the PMA+ionomycin- induced IL-2 release in a dose-dependent manner.
  • the JNK inhibitor with SEQ ID NO: 172 did reduce the PMA+ionomycin- induced IFN- ⁇ release in a dose-dependent manner.
  • Fig.21 JNK inhibition and TNF-a release in human whole blood The JNK inhibitor with SEQ ID NO: 172 was added at three different concentrations, namely 1, 3 and 10 ⁇ 1h before whole blood stimulation with PMA+ionomycin (25/700 ng/mL, 50/800 ng/ml and 50/1000 ng/mL) for 4 hours.
  • the JNK inhibitor with SEQ ID NO: 172 did reduce the PMA+ionomycin -induced TNF-a release in a dose-dependent manner.
  • Fig.22 JNK inhibition and TNF-a release in human whole blood The JNK inhibitor with SEQ ID NO: 172 was added at three different concentrations, namely 1, 3 and 10 ⁇ 1h before whole blood stimulation with PHA-L (5 ⁇ g/mL) for 3 days.
  • the JNK inhibitor with SEQ ID NO: 172 did reduce the PHA-L-induced TNF-a release in a dose-dependent manner.
  • JNK inhibition and IL-2 release in human whole blood The JNK inhibitor with SEQ ID NO:
  • the JNK inhibitor with SEQ ID NO: 172 did reduce the PHA-L-induced IL-2 release in a dose-dependent manner. - -
  • Fig.24 JNK inhibition and TNF-a release in human whole blood The JNK inhibitor with SEQ ID NO: 172 was added at three different concentrations, namely 1, 3 and 10 ⁇ 1h before whole blood stimulation with CD3 +/- CD28 antibodies (2 ⁇ g/mL) for 3 days.
  • the JNK inhibitor with SEQ ID NO: 172 did reduce the CD3/CD28-induced TNF-a release in a dose- dependent manner.
  • FIG.25 Photograhic illustration of in vivo anti-inflammatory properties of the JNK inhibitors with SEQ ID NO: 197 (10 pg/kg) and SEQ ID NO: 172 (10 pg/kg) after CFA (complete Freund's adjuvant) induced paw swelling. Paw swelling was induced in the left hind paw, the right hind paw was not treated.
  • FIG.27 Graphical representation of in vivo anti-inflammatory properties of the JNK inhibitors with SEQ ID NO: 197 (10 pg/kg) and SEQ ID NO: 172 (10 pg/kg) after CFA (complete Freund's adjuvant) induced paw swelling. Indicated is the measured in vivo cytokine release one hour after CFA induced paw swelling.
  • Fig.29 Responsive effects of the JNK inhibitor of SEQ ID NO: 172 after daily intravenous administration in 14 day rat chronic established Type II collagen arthritis (RTTC/SOL-1 ).
  • Fig. 33 Clinical scoring by slit lamp 24 hours after ElU induction and administration of JNK inhibitor according to SEQ ID NO: 1 72 (1 mg/kg i.v.) at different times prior to ElU induction. From left to right: Vehicle (0 hours); SEQ ID NO: 1 72 4 weeks prior to ElU induction; SEQ ID NO: 1 72 2 weeks prior to ElU induction; SEQ ID NO: 1 72 1 week prior to ElU induction; SEQ ID NO: 1 72 48 hours prior to ElU induction; SEQ ID NO: 1 72 24 hours prior to ElU induction; SEQ ID NO: 1 72 0 hours prior to ElU induction; Dexamethasone (2 mg/kg i.v.) 0 hours prior to ElU induction. Mean ⁇ SEM. *p ⁇ 0.05 versus vehicle, **p ⁇ 0.01 versus vehicle.
  • Fig. 34 Number of PMN cells per section quantified 24 hours after ElU induction and administration of JNK inhibitor according to SEQ ID NO: 1 72 (1 mg/kg i.v.) at different times prior to ElU induction. From left to right: Vehicle (0 hours); SEQ ID NO: 1 72 4 weeks prior to ElU induction; SEQ ID NO: 1 72 2 weeks prior to ElU induction; SEQ ID NO: 1 72 1 week prior to ElU induction; SEQ ID NO: 1 72 48 hours prior to ElU induction; SEQ ID NO: 1 72 24 hours prior to ElU induction; SEQ ID NO: 1 72 0 hours prior to ElU induction; Dexamethasone (2 mg/kg i.v.) 0 hours prior to ElU induction. Mean ⁇ SEM. *p ⁇ 0.05 versus vehicle, **p ⁇ 0.01 versus vehicle. - -
  • Fig. 35 shows the mean calculated TBUT AUC values for animals with scopolamine-induced dry eye syndrome. Shown are the results for animals treated with vehicle, 3 different concentrations of an all-D-retro-inverso JNK-inhibitor (poly-)peptide with the sequence of SEQ ID NO: 1 97, 3 different concentrations of a JNK-inhibitor (poly-)peptide with the sequence of SEQ ID NO: 1 72, and the results for animals treated with cyclosporine.
  • Fig. 36 shows the mean calculated PRTT AUCs for animals with scopolamine induced Dry Eye (Day 7-21 ). Shown are the results for animals treated with vehicle, 3 different concentrations of an all-D-retro-inverso JNK-inhibitor (poly-)peptide with the sequence of SEQ ID NO: 1 97, 3 different concentrations of a JNK-inhibitor (poly-)peptide with the sequence of SEQ ID NO:
  • Fig. 37 shows the mean histological Cornea Lesion Scores for animals with scopolamine induced dry eye syndrome. Shown are the results for animals treated with vehicle, 3 different concentrations of an all-D-retro-inverso JNK-inhibitor (poly-)peptide with the sequence of
  • SEQ ID NO: 1 97 3 different concentrations of a JNK-inhibitor (poly-)peptide with the sequence of SEQ ID NO: 1 72, and the results for animals treated with cyclosporine.
  • Fig. 38 shows the renal function assessed by protidemia (A) and urea level (B) of rats in an Adriamycin (ADR)-induced nephropathy model on Days 8, 14, 29, 41 and 56 after ADR administration.
  • Groups No. 1 (“ADR") and No. 2 (“ADR + JNK inhibitor SEQ Id NO: 1 72") have been treated on Day 0 with ADR to induce necropathy, whereas group No. 3 (“NaCI”) received 0.9% NaCL.
  • group No. 2 (“ADR + JNK inhibitor SEQ Id NO: 1 72") has been treated on Day 0 with the JNK inhibitor SEQ ID NO: 1 72, whereas groups No. 1 and 3 received vehicle (0.9% NaCI).
  • ADR Adriamycin
  • PAS periodic acid-Schiff
  • FIG. 40 shows the kidney fibrosis in ADR nephropathy evaluated with Masson's trichrome (blue) on Days 8 (left four panels) and 56 (right four panels) following ADR administration for the group "ADR" (upper panel), which has been treated with ADR and vehicle at Day 0 and for the group "ADR + XG104" (lower panel), which has been treated with ADR and the JNK inhibitor SEQ ID NO: 1 72 at Day 0.
  • the original magnification x10 is depicted in the left panels for the respective day and the original magnification x40 is depicted in the right panels for the respective day.
  • Rats of group G3 received a single dose of 2 mg/kg of the JNK inhibitor according to SEQ ID NO. 1 72 ("XG-1 04") (in 0.9% NaCI as vehicle) and rats of groups G1 and G2 received vehicle, respectively, by IV injection in the tail vein on Day 0, one hour after clamping period (after reperfusion) both renal pedicles with atraumatic clamp.
  • Serum creatinine and urea were increased in vehicle-treated ischemic rats (G2) 24h following ischemia, as compared to vehicle-treated controls rats without ischemia (G1 ).
  • XG-104-treated-ischemic rats G3 exhibited lower serum creatinine and lower urea, relatively to untreated ischemic rats (G2).
  • Fig. 43 shows for Example 20 that XG-104 blocks the development of a psoriatic phenotype in vivo in the K5.STAT3c model for psoriasis.
  • Wild type or K5STAT3c mice were treated with either vehicle (Saline) or XG-104 compound prior to tape stripping, followed by daily treatments of vehicle or XG-1 04.
  • mice were sacrificed and biopsies taken from the lesion sites in order to quantify acanthosis (thickening of the epidermis). Quantification of acanthosis is shown for two independent experiments separately (A) and combined (B). Dots represent mean values from individual mice (A&B).
  • a one-way ANOVA test was performed to analyze significance. - -
  • Example 44 shows for Example 20 representative histologies of K5.STAT3c mice treated with either Saline or XG-1 04, indicating that XG-104 blocks the development of a psoriatic phenotype in vivo in the K5.STAT3c model for psoriasis.
  • Example 45 shows for Example 1 9 the results of the determination of of the cytotoxic activity of XG-104 against HepG2 (A) and PLC/PRF/5 (B) tumour cell lines using MTS assay.
  • Fig. 46 shows for Example 23 the study design.
  • Fig. 47 shows for Example 23 the effects of vehicle and XG-104 (2 mg/kg, i. v.) on tubular damages in a rat model of bilateral IR. ***P ⁇ 0.001 versus Group 1 (Sham/Vehicle) by a Student Mest ns; +P ⁇ 0.05 versus Group 2 (IR/Vehicle) by a one way ANOVA followed by a Bonferroni's post test.
  • Fig. 48 shows for Example 23 the effects of of vehicle and XG-1 04 (2 mg/kg, i. v.) on total tubular histological scores in a rat model of bilateral IR.
  • Total tubular score represents al l tubular changes including degeneration and necrosis, tubular cast, tubular epithelial vacuolation and regeneration (basophil tubules).
  • Fig. 49 shows for Example 23 representative images of hematoxylin/eosin stained kidney sections: comparison between Groups 2 (IR/Vehicle) and 3 (IR/XG-104). Animal 53 (Top Left), Animal 15 (Top Right), Animal 46 (Bottom left), and Animal 1 8 (Bottom right): 1 0x. Representative photomicrographs of tubular degeneration/necrosis and tubular casts in Group 2 (vehicle) and 3 (XG-1 04). Animals having scores from 1 to 4 are represented. The main difference between groups is that the severity of tubular necrosis and cast in Group 2 is generally higher than that observed in Group 3. In Group 2, lesions are extended partially or to the majority of the cortex. Comparatively, in Group 3, lesions are limited to the cortico-medullary junction. Lesions consist of a mixture of active necrosis, cellular tubular casts, hyaline casts, and occasional basophi lic tubules.
  • Fig. 50 shows for Example 24 the study design (A) and the AUCs method to assess allodynia and hyperalgesia (B).
  • - - shows for Example 24 the effect of XG-1 04 (50 mg/mL, i .ves.) and ibuprofen (50 mg/mL, i .ves.) treatments on nociceptive parameters 24h post-CYP injection.
  • Nociceptive threshold (A), nociceptive scores (B), AUC 1 -8 g (C) or AUC 8-60 g (D) 24h after CYP i njection. Results are expressed as mean + s.e.m. (n 1 0).
  • Example 24 shows for Example 24 the effect of XG-1 04 (50 mg/mL, i.ves.) and ibuprofen (50 mg/mL, i.ves.) treatments on urinary bladder wal l thickness as wel l as on oedema and haemorrhage scores 24h post-CYP i njection.
  • Fig. 55 shows for Example 26 the decrease of neuronal apoptosis after PKR down-regulation and/or JNK inhibition with XG-104, referred to as "JNKi" (in Fig. 55).
  • JNKi neuronal apoptosis after PKR down-regulation and/or JNK inhibition with XG-104
  • Fig. 55 shows for Example 26 the decrease of neuronal apoptosis after PKR down-regulation and/or JNK inhibition with XG-104, referred to as "JNKi” (in Fig. 55).
  • A Immunoblot results of the levels of JNK and c-Jun activation, caspase 3 and PARP cleaved activated fragments in primary neuronal cultures of WT and PKR ' mice, treated by 2 ⁇ of ⁇ 42 after or not pre-inhibition of JNK with 10 ⁇ JNKi compound.
  • B-D Corresponding histograms of JNK activity (B), phospho
  • E-G Apoptosis is measured by the level of cleaved caspase 3 (E), caspase 3 activity measured in the cell culture supernatant (F) and cleaved PARP (G). Data are means ⁇ SEM (ri> 3 per condition). * ⁇ 0.05, ** ⁇ 0.01 , and ***/° ⁇ 0.001 .
  • the present invention relates to a JNK inhibitor, which comprises an inhibitory (polypeptide sequence according to the following general formula:
  • X1 is an amino acid selected from amino acids R, P, Q and r
  • X2 is an amino acid selected from amino acids R, P, G and r
  • X3 is an amino acid selected from amino acids K, R, k and r
  • X4 is an amino acid selected from amino acids P and K
  • X5 is an amino acid selected from amino acids T, a, s, q, k or is absent
  • X6 is an amino acid selected from amino acids T, D and A
  • X7 is an amino acid selected from amino acids N, n, r and K
  • X8 is an amino acid selected from F, f and w, with the proviso that at least one, at least two, at least three, at least four, at least five or six of the amino acids selected from the group consisting of X1 , X2, X3, X5, X7 and X8 is/are a
  • D-amino acid(s) preferably with the proviso that at least one, at least two, at least three or four of the amino acids selected from the group consisting of X3, X5, X7 and X8 is/are a D- amino acid(s), - - for use in a method for treatment of the human or animal body by therapy, in particular for the treatment of the diseases/disorders disclosed herein.
  • the inhibitory (poly-)peptide sequence of the JNK inhibitor according to the present invention comprises L-amino acids and in most embodiments D-amino acids. Unless specified otherwise, L- amino acid residues are indicated herein in capital letters, while D amino acid residues are indicated in small letters. Glycine may be indicated in capital or small letters (since there is no D- or L-glycine).
  • the amino acid sequences disclosed herein are always given from N- to C-terminus (left to right) unless specified otherwise. The given amino acid sequence may be modified or unmodified at the C- and/or N-terminus, e.g. acetylation at the C-terminus and/or amidation or modification with cysteamide at the N-terminus.
  • the JNK inhibitors of the present invention are (poly-)peptide inhibitors of the c-Jun N-terminal kinase (JNK). Said inhibitors inhibit the kinase activity of c-Jun N-terminal kinase (JNK), i.e. prevent or reduce the extent of phosphorylation of JNK substrates, such as c-Jun, ATF2 and/or Elk-1 by e.g. blocking the JNK activity.
  • the term "inhibitor”, as used herein, does not comprise compounds which irreversibly destroy the c-Jun N-terminal kinase (JNK) molecule and/or kinase activity. Accordingly, the JNK inhibitory activity of the inhibitors of the present invention typically refers to compounds which bind in a competitive or non-competitive manner to JNK. Furthermore, the term “inhibiting JNK activity” as used herein, refers to the inhibition of the kinase activity of c-Jun N-terminal kinase (JNK). Furthermore, as used herein, a JNK inhibitor comprises at least one functional unit of a polymer of amino acids, i.e.
  • a (poly-)peptide sequence Moreover, this at least one functional polymer of amino acids provides for inhibition of JNK activity.
  • the amino acid monomers of said inhibitory (polypeptide sequence are usually linked to each other via peptide bonds, but (chemical) modifications of said peptide bond(s) or of side chain residues may be tolerable, provided the inhibitory activity (inhibition of JNK activity) is not totally lost, i.e. the resulting chemical entity still qualifies as JNK inhibitor as functionally defined herein.
  • the term "(poly-)peptide” shall not be construed as limiting the length of the (poly-)peptide unit.
  • the inhibitory (poly-)peptide sequence of the JNK inhibitors of the present invention is less than 500, 490, 480, 470, 460, 450, 440, 430, 420, 410, 400, 390, 380, 370, 360, 350, 340, 330, 320, 310, 300, 290, 280, 270, 260, 250, 240, 230, 220, 21 0, 200, 1 90, 1 80, 1 70, 1 60, 1 50, 140, 1 30, 120, 1 10, 1 00, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 49, 48, 47, 46, 45, 44, 43, 42, 41 , 40, 39, 38, 37, 36, 35, 34, 33, 32, 31 , 30, 29, 28, 27, 26, 25, 24, - -
  • the inhibitory (poly-)peptide sequence does not have less than 10 amino acid residues, more preferably not less than 1 1 amino acid residues.
  • a "JNK inhibitor" of the present invention inhibits JNK activity, e.g. exhibits with regard to the inhibition of human JNK mediated phosphorylation of a c-Jun substrate (SEQ ID NO: 1 98) an IC 50 value of: a) less than 3000 nM, more preferably less than 2000 nM, even more preferably less than 1 000 nM, even more preferably less than 500 nM, even more preferably less than 250 nM, even more preferably less than 200 nM, even more preferably less than 1 50 nM, most preferably less than 1 00 nM with regard to inhibition of human JNK1 , b) less than 3000 nM, more preferably less than 2000 nM, even more preferably less than 1 000 nM, even more preferably less than 500 nM, even more preferably less than 250 nM, even more preferably less than 200 nM, even more preferably less than 1 50 nM, most preferably less than 100 nM with
  • nM less than 3000 nM, more preferably less than 2000 nM, even more preferably less than 1 000 nM, even more preferably less than 500 nM, even more preferably less than 250 nM, even more preferably less than 200 nM, even more preferably less than 1 50 nM, most preferably less than 100 nM with regard to inhibition of human JNK3.
  • the inhibitor inhibits human JNK2 and/or human JNK3 according to the above definition, but not JNK1 according to the above definition.
  • JNK activity is inhibited or not, may easily be assessed by a person skilled in the art.
  • a radioactive kinase assay or a non-radioactive ki nase assay e.g. Alpha screen test; see for example Guenat et al. J Biomol Screen, 2006; 1 1 : pages 101 5-1 026).
  • a JNK inhibitor according to the present invention may thus for example comprise an inhibitory (poly-)peptide sequence according to any of SEQ ID NOs: 2 to 27 (see table 1 ).
  • the JNK inhibitor according to the present invention may also be a JNK inhibitor (variant) which comprises an inhibitory (poly-)peptide sequence sharing at least 50%, more preferably at least 55%, more preferably at least 60%, more preferably at least 65%, more preferably at least 70%, more preferably at least 75%, more preferably at least 80%, more preferably at least 85%, most preferably at least 90%, more preferably at least 95% sequence identity with a sequence selected from SEQ ID NOs: 1 -27, in particular with SEQ ID NO: 8, - - preferably with the proviso that with regard to the respective sequence selected from SEQ ID NOs: 1 -27, such inhibitory (poly-)peptide sequence sharing sequence identity
  • c) exhibits one, two, three, four, five or six D-amino acid(s) at the respective positions corresponding to the amino acids selected from the group consisting of X1 , X2, X3, X5, X7 and X8 of SEQ ID NO: 1 and respective positions in SEQ ID NOs: 2-27, more preferably exhibits one, two, three or four D-amino acid(s) at the positions corresponding to the amino acids selected from the group consisting of X3, X5, X7 and X8 of SEQ ID NO: 1 and respective positions in SEQ ID NOs: 2-27, and d) stil l inhibits JNK activity (i.e. is a JNK inhibitor as defined herein).
  • variants disclosed herein (in particular JNK inhibitor variants comprising an inhibitory (poly-)peptide sequence sharing - within the above definition - a certain degree of sequence identity with a sequence selected from SEQ ID NOs: 1 -27), share preferably less than 1 00% sequence identity with the respective reference sequence.
  • the non-identical amino acids are preferably the result of conservative amino acid substitutions.
  • Conservative amino acid substitutions may include amino acid residues within a group which have sufficiently similar physicochemical properties, so that a substitution between members of the group will preserve the biological activity of the molecule (see e.g. Grantham, R. (1 974), Science 185, 862-864).
  • conservative amino acid substitutions are preferably substitutions in which the amino acids originate from the same class of amino acids (e.g. basic amino acids, acidic amino acids, polar amino acids, amino acids with aliphatic side chains, amino acids with positively or negatively charged side chains, amino acids with aromatic groups in the side chains, amino acids the side chains of which can enter into hydrogen bridges, e.g. side chains which have a hydroxy! function, etc.).
  • Conservative substitutions are in the present case for example substituting a basic amino acid residue (Lys, Arg, His) for another basic amino acid residue (Lys, Arg, His), substituting an aliphatic amino acid residue (Gly, Ala, Val, Leu, lie) for another aliphatic amino acid residue, substituting an aromatic amino acid residue (Phe, Tyr, Trp) for another aromatic - - amino acid residue, substituting threonine by serine or leucine by isoleuci ne.
  • the isomer form should preferably be maintai ned, e.g. K is preferably substituted for R or H, whi le k is preferably substituted for r and h.
  • JNK inhibitor variants are for example: a) one, two or more of X1 , X2, X3, X4, X5, X6, X7 and/or X8 of SEQ I D NO: 1 or the corresponding positions within the respective sequence selected from SEQ ID NOs: 2-27 are substituted for A or a,
  • X5 of SEQ I D NO: 1 or the corresponding position within the respective sequence selected from SEQ I D NOs: 2-27 is E, Y, L, V, F or K;
  • X5 of SEQ ID NO: 1 or the corresponding position within the respective sequence selected from SEQ ID NOs: 2-27 is E, L, V, F or K; or
  • n one, two or three of X1 , X2, X3 of SEQ ID NO: 1 or the correspondi ng positions within the respective sequence selected from SEQ ID NOs: 2-27 are neutral amino acids.
  • % sequence identity has to be understood as fol lows: Two sequences to be compared are aligned to give a maximum correlation between the sequences. This may i nclude inserti ng "gaps" i n either one or both sequences, to enhance the degree of alignment.
  • a % identity may then be determined over the whole length of each of the sequences being compared (so-called global alignment), that is particularly suitable for sequences of the same or similar length, or over shorter, defined lengths (so-called local alignment), that is more suitable for sequences of unequal length.
  • an amino acid sequence having a "sequence identity" of at least, for example, 95% to a query ami no acid sequence is intended to mean that the sequence of the subject ami no acid sequence is identical to the query sequence except that the subject ami no acid sequence may include up to five amino acid alterations per each 1 00 amino acids of the query amino acid sequence.
  • an amino acid sequence having a sequence of at least 95% identity to a query amino acid sequence up to 5% (5 of 1 00) of the amino acid residues in the subject sequence may be inserted or substituted with another amino acid or deleted.
  • sequence identity the substitution of an L-amino acid for a D-amino acid (and vice versa) is considered to yield a non-identical residue, even if it is merely the D- (or L-isomer) of the very same amino acid.
  • BESTFIT uses the "local homology” algorithm of (Smith and Waterman (1981 ), J. Mol. Biol. 747, 1 95-1 97.) and finds the best single region of similarity between two sequences.
  • the JNK inhibitor according to the present invention may comprise - in addition to the inhibitory (poly-)peptide sequence mentioned above - additional sequences or sequence elements, domains, labels (e.g. fluorescent or radioactive labels), epitopes etc., as long as the ability to inhibit JNK activity as defined herein is not lost.
  • the JNK inhibitor according to the present invention may also comprise a transporter sequence.
  • a "transporter sequence” as used herein, is a (poly-)peptide sequence providing for translocation of the molecule it is attached to across biological membranes.
  • a JNK inhibitor according to the present invention comprising a transporter sequence is preferably capable of translocating (e.g. the conjugated cargo compound) across biological membranes.
  • translocating e.g. the conjugated cargo compound
  • Said transporter sequence may be joined for example (e.g. directly) N-terminally or (e.g. directly) C- terminally to the inhibitory (poly-)peptide sequence of the JNK inhibitor, preferably by a covalent linkage.
  • the transporter sequence and the inhibitory (poly-)peptide sequence may also be spaced apart, e.g. may be separated by intermediate or linker sequences.
  • the transporter sequence may be positioned entirely elsewhere in the JNK inhibitor molecule than the inhibitory (poly-)peptide sequence, in particular if the JNK inhibitor is a more complex molecule (e.g. comprising several domains, is a multimeric conjugate etc.).
  • the transporter sequence and the inhibitory (poly-)peptide sequence may overlap. However, the JNK inhibitory activity of the JNK inhibitory portion needs to be maintained. Examples for such overlapping instances are given further below.
  • Transporter sequences for use with the JNK inhibitor of the present invention may be selected from, without being limited thereto, transporter sequences derived from HIV TAT (HIV), e.g. native proteins such as e.g. the TAT protein (e.g. as described in U.S. Patent Nos. 5,804,604 and 5,674,980, each of these references being incorporated herein by reference), HSV VP22 ⁇ Herpes simplex) (described in e.g. WO 97/05265; Elliott and O'Hare, Cell 88 : 223-233 (1 997)), non-viral proteins (Jackson et al, Proc. Natl. Acad. Sci.
  • HIV TAT HIV TAT
  • native proteins such as e.g. the TAT protein (e.g. as described in U.S. Patent Nos. 5,804,604 and 5,674,980, each of these references being incorporated herein by reference)
  • HSV VP22 ⁇ Herpes simplex described in
  • transporter sequences derived from Antennapedia, particularly from Drosophila antennapedia (e.g. the antennapedia carrier sequence thereof), FGF, lactoferrin, etc. or derived from basic peptides, e.g. peptides having a length of at least 5 or at least 1 0 or at least 1 5 amino acids, e.g. 5 to 1 5 amino acids, preferably 10 to 12 amino acids,
  • Such transporter sequences preferably comprise at least 50%, more preferably at least 80 %, more preferably 85 % or even 90 % basic amino acids, such as e.g. arginine, lysine and/or hi stidine, or may be selected from e.g.
  • arginine rich peptide sequences such as RRRRRRRRR (R 9 ; SEQ ID NO: 1 52), RRRRRRRR (R 8 ; SEQ ID NO: 1 53), RRRRRRR (R 7 ; SEQ ID NO: 1 54), RRRRRR (R 6 , SEQ ID NO: 1 55), RRRRR (R 5 , SEQ ID NO: 1 56) etc., from VP22, from PTD-4 proteins or peptides, from RGD-K16, from PEPT1 /2 or PEPT1/2 proteins or peptides, from SynB3 or SynB3 proteins or peptides, from PC inhibitors, from P21 derived proteins or peptides, or from JNKI proteins or peptides.
  • transporter sequences for use in the JNK inhibitor of the present invention are in particular, without being limited thereto, basic transporter sequences derived from the HIV-1 TAT protein.
  • the basic transporter sequence of the HIV-1 TAT protein may include sequences from the human immunodeficiency virus HIV-1 TAT protein, e.g. as described in, e.g., U.S. Patent Nos. 5,804,604 and 5,674,980, each incorporated herein by reference.
  • the full- length HIV-1 TAT protein has 86 amino acid residues encoded by two exons of the HIV TAT gene. TAT amino acids 1 -72 are encoded by exon 1 , whereas amino acids 73-86 are encoded by exon 2.
  • the full-length TAT protein is characterized by a basic region which contains two lysines and six arginines (amino acids 49-57) and a cysteine-rich region which contains seven cysteine residues (amino acids 22-37).
  • the basic region i.e., amino acids 49-57 was thought to be important for nuclear localization. Ruben, S. et a/., J. Virol. 63 : 1 -8 (1 989); Hauber, J. et a/., J. Virol. 63 1 1 81 - 1 1 87 (1 989).
  • the cysteine-rich region mediates the formation of metal-linked dimers in vitro (Frankel, A. D.
  • TAT transporter sequences for use in the JNK inhibitor of the present - - invention are preferably characterized by the presence of the TAT basic region amino acid sequence (amino acids 49-57 of naturally-occurring TAT protein); the absence of the TAT cysteine-rich region amino acid sequence (amino acids 22-36 of naturally-occurring TAT protein) and the absence of the TAT exon 2-encoded carboxy-terminal domain (amino acids 73-86 of naturally-occurring TAT protein). More preferably, the transporter sequence in the JNK inhibitor of the present invention may be selected from an amino acid sequence containing TAT residues 48-57 or 49 to 57 or variants thereof.
  • the transporter sequence in a given JNK inhibitor of the present invention also exhibits D-amino acids, for example in order to improve stability towards proteases.
  • Particularly preferred are transporter sequences which exhibit a specific order of alternating D- and L-amino acids.
  • Such order of alternating D- and L-amino acids may follow -without being limited thereto - the pattern of any one of SEQ ID NOs: 28-30: diLLL x d m LLL y dn (SEQ ID NO: 28); dLLLd(LLLd) a (SEQ ID NO: 29); and/or dLLLdLLLd (SEQ ID NO: 30); wherein: d is a D-amino acid;
  • L is a L-amino acid
  • a is 0 - 3, preferably 0-2, more preferably 0, 1 , 2 or 3, even more preferably 0, 1 , or 2 and most preferably 1 ;
  • I, m and n are independently from each other 1 or 2, preferably 1 ;
  • x and y are independently from each other 0, 1 or 2, preferably 1 .
  • Said order of D- and L-amino acids becomes relevant when the transporter sequence is synthesized, i.e. while the amino acid sequence (i.e. the type of side chain residues) remains unaltered, the respective isomers alternate.
  • a known transporter sequence derived from HIV TAT is RKKRRQRRR (SEQ ID NO: 43). Applying the D-/L amino acid order of SEQ ID NO: 30 thereto would yield rKKRrQRRr (SEQ ID NO: 46).
  • the transporter sequence of the JNK inhibitor of the present invention may comprise at least one sequence according to rXXXrXXr (SEQ ID NO: 31 ), wherein:
  • r represents an D-enantiomeric arginine
  • X is any L-amino acid (including glycine); - - and wherein each X may be selected individually and independently of any other X within SEQ ID NO: 31.
  • the JNK inhibitor according to the present invention comprises the transporter sequence rXiX 2 X 3 rX 4 X 5 X6r (SEQ ID NO: 32), wherein Xi is K, X 2 is K, X 3 is R and X 4 , X 5 , and X 6 are any L-amino acid (including glycine) selected independently from each other.
  • the transporter sequence of the JNK inhibitor according to the present invention may comprise the sequence rXiX 2 X 3 rX 4 X 5 X6r (SEQ ID NO: 33), wherein X 4 is Q, X 5 is R, X 6 is R and ⁇ ⁇ , X 2 , and X 3 are any L-amino acid (including glycine) selected independently from each other.
  • the inventive JNK inhibitor may also comprise the sequence rXiX 2 X3rX 4 X5X6r (SEQ ID NO: 34), wherein one, two, three, four, five or six X amino acid residues are chosen from the group consisting of: Xi is K, X 2 is K, X 3 is R, X 4 is Q, Xs is R, ⁇ is R, while the remaining X amino acid residues not selected from above group may be any L-amino acid (including glycine) and are selected independently from each other.
  • Xi is then preferably Y and/or X 4 is preferably K or R.
  • transporter sequences for use in the inventive JNK inhibitor molecule may be selected, without being limited thereto, from sequences as given in table 2 below, (SEQ ID NOs: 31-170) or from any fragment or variant or chemically modified derivative thereof (preferably it retains the function of translocating across a biological membrane).
  • PTD-4 (variant 2) 162 11 WARAQRAAARA
  • transporter sequences may also be selected from fragments or variants of the above sequences of table 2 (with the proviso that such fragment or variant retain preferably the fu nction to provide for translocation across biological membranes).
  • variants and/or fragments of those transporter sequences preferably comprise a peptide sequence sharing at least 1 0%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80% or at least 85%, preferably at least 90%, more preferably at least 95% and most preferably at least 99% sequence identity over the whole length of the sequence with such a transporter sequence as defined in Table 2.
  • a "fragment" of a transporter sequence as defined in Table 2 is preferably to be understood as a truncated sequence thereof, i.e. an amino acid sequence, which is N-terminally, C-terminally and/or intrasequentially truncated compared to the amino acid sequence of the original sequence.
  • a "variant" of a transporter sequence or its fragment as defined above is preferably to be understood as a sequence wherein the amino acid sequence of the variant differs from the original transporter sequence or a fragment thereof as defined herein in one or more mutation(s), such as one or more substituted, (or, if necessary, inserted and/or deleted) amino acid(s).
  • variants of such a transporter sequence as defined above have the same biological function or specific activity compared to the respective original sequence, i.e. provide for transport, e.g. into cells or the nucleus.
  • a variant of such a transporter sequence as defined above may for example comprise about 1 to 50, 1 to 20, more preferably 1 to 1 0 and most preferably 1 to 5, 4, 3, 2 or 1 amino acid alterations.
  • Variants of such a transporter sequence as defined above may preferably comprise conservative amino acid substitutions. The concept of conservative amino acid substitutions is known in the art and has already been set out above for the JNK inhibitory (poly-)peptide sequence and applies here accordingly.
  • the length of a transporter sequence incorporated in the JNK inhibitor of the present invention may vary. It is contemplated that in some embodiments the transporter sequence of the JNK inhibitor according to the present invention is less than 1 50, less than 140, less than 130, less than 120, less than 1 10, less than 100, less than 90, less than 80, less than 70, less than 60, less than 50, less than 40, less than 30, less than 20, and/or less than 1 0 amino acids in length. - -
  • the JNK inhibitor comprising a transporter domain may be fused to a label, e.g. a fluorescent protein such as GFP, a radioactive label, an enzyme, a fluorophore, an epitope etc. which can be readily detected in a cell.
  • a label e.g. a fluorescent protein such as GFP, a radioactive label, an enzyme, a fluorophore, an epitope etc. which can be readily detected in a cell.
  • the JNK inhibitor comprising the transporter sequence and the label is transfected into a cell or added to a culture supernatant and permeation of cell membranes can be monitored by using biophysical and biochemical standard methods (for example flow cytometry, (immuno)fluorescence microscopy etc.).
  • JNK inhibitors according to the present invention comprising a transporter sequence are given in table 3:
  • JNK inhibitors comprising an inhibitory
  • the transporter sequence and the inhibitory (poly-)peptide sequence may overlap.
  • the N-terminus of the transporter sequence may overlap with the C-terminus of the inhibitory (poly-)peptide sequence or the C-terminus of the transporter sequence may overlap with the N-terminus of the inhibitory (polypeptide sequence.
  • the transporter sequence overlaps by one, two or three amino acid residues with the inhibitory (poly-)peptide sequence.
  • a given transporter sequence may overlap with SEQ ID NO:1 or the respective variants thereof at position 1 (X1 ), position 1 and 2 (X1 , X2), positions 1 , 2 and 3 (X1 , X2, X3).
  • SEQ ID NOs: 1 74, 1 75, 1 78, 1 79, 1 80, 181 , 1 82, 183, 1 84, 1 88, 189 and 1 90 are examples for JNK in hibitors according to the present invention, wherein transporter sequence and the inhibitory (polypeptide sequence overlap, e.g. rKKRrO RRrRPTTLNLf (SEP ID NO: 1 74) is an overlap of SEQ ID NO: 46 (underlined) and SEQ ID NO: 1 1 (italics).
  • the JNK inhibitor according to the present invention may also be selected from JNK inhibitors, which are a variant of any one of the JNK inhibitors according to SEQ ID NOs: 1 71 -1 90.
  • such variant shares at least 50%, more preferably at least 55%, more preferably at least 60%, more preferably at least 65%, more preferably at least 70%, more preferably at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, most preferably at least 95% sequence identity with the sequence of SEQ ID NOs: 1 71 -1 90, in particular with SEQ ID NO: 1 72,
  • b) maintains the two L-leucine (L) residues at position 8 and 1 0 (positions 7 and 9 with regard to SEQ ID NOs: 25-27) within the inhibitory (poly-)peptide sequence
  • c) exhibits at least one, at least two, at least three, at least four, at least five or six D- amino acid(s) at the respective positions corresponding to the amino acids selected from the group consisting of X1 , X2, X3, X5, X7 and or X8 of SEQ ID NO: 1 and respective positions in SEQ ID NOs: 2-27, more preferably exhibits at least one, at least two, at least three or four D-amino acid(s) at the positions corresponding to the amino acids selected from the group consisting of X3, X5, X7 and X8 of SEQ ID NO: 1 and respective positions in SEQ ID NOs: 2-27, and - - d) inhibits JNK activity (i.e. is a JNK inhibitor as defined herein).
  • non-identical amino acids in the variants of JNK inhibitors comprising SEQ ID NOs: 1 71 -190 are preferably the result of conservative amino acid substitutions (see above).
  • substitutions mentioned above are also contemplated for variants of JNK inhibitors comprising SEQ ID NOs: 1 71 -1 90.
  • the present invention certainly also contemplates variants of any one of the JNK inhibitors according to SEQ ID NOs: 1 71 -190, which deviate from the original sequence not or not exclusively in the inhibitory (poly-)peptide sequence, but exhibits variant residues in the transporter sequence.
  • the respective disclosure herein is pertinent.
  • the transporter sequence and the JNK inhibitory (poly)-peptide sequence of the JNK inhibitors according to the present invention need not necessari ly be directly linked to each other. They may also be linked by e.g. an intermediate or linking (poly-)peptide sequences.
  • Preferred intermediate or linking sequences separating the inhibitory (poly-)peptide sequences and other (functional) sequences such as transporter sequences consist of short peptide sequences of less than 10 amino acids in length, like a hexamer, a pentamer, a tetramer, a tripeptide or a dipeptide or a single amino acid residue.
  • Particularly preferred intermediate sequence are one, two or more copies of di-proline, di-glycine, di-arginine and/or di-lysine, all either in L-amino acid form only, or in D-amino acid form only, or with mixed D- and L-amino acids.
  • other known peptide spacer or linker sequences may be employed as well.
  • a particularly preferred JNK inhibitor according to the present invention comprises SEQ ID NO: 8 (or a sequence sharing sequence identity with SEQ ID NO: 8 with the scope and limitations defined further above) and a transporter sequence.
  • the transporter sequence is preferably selected from any one of SEQ ID Nos: 31 -1 70 or variants thereof as defined herein, even more preferably from any one of SEQ ID NOs: 31 -34 and 46-1 51 .
  • a particularly preferred embodiment of a JNK inhibitor according to the present invention is a JNK inhibitor comprising SEQ ID NO: 8 and SEQ ID NO: 46 (or sequences sharing respective sequence identity thereto within the scope and limitations defined above).
  • a preferred example is a JNK inhibitor comprising the sequence of SEQ ID NO: 1 72 or respective variants thereof varying in the transporter sequence and/or the inhibitory (poly-)peptide sequence as defined herein. - -
  • the present invention relates to a JNK inhibitor comprising
  • an inhibitory (poly-)peptide comprising a sequence from the group of sequences consisting of RPTTLNLF (SEQ ID NO: 1 91 ), KRPTTLNLF (SEQ ID NO: 1 92), RRPTTLNLF and/or RPKRPTTLNLF (SEQ ID NO: 1 93), and
  • a transporter sequence preferably a transporter sequence selected from the transporter sequences disclosed in table 2 or variants/fragments thereof, even more preferably selected from SEQ ID NOs: 31 -34 and 46-1 51 or respective variants or fragments thereof.
  • the transporter sequence and the inhibitory (poly-)peptide sequence may overlap.
  • Preferred transporter sequences for said embodiment of the invention are particularly the transporter sequence of SEQ ID NO: 46, preferably (covalently) linked (e.g. directly) to the N-terminus of the inhibitory (poly-)peptide sequence.
  • a JNK inhibitor of the present invention may also be a JNK inhibitor comprising or consisting of the sequence GRKKRRQRRRPPKRPTTLNLFPQVPRSQD (SEQ ID NO: 194), or the sequence GRKKRRQRRRPTTLNLFPQVPRSQD (SEQ ID NO: 1 95).
  • the present invention relates to a (poly-)peptide comprising a transporter sequence selected from the group of sequences consisting of rKKRrQRr (SEQ ID NO: 148), r KRrQRrK (SEQ ID NO: 1 49), and/or rKKRrQRrR (SEQ ID NO: 1 50).
  • a sequence or a given SEQ ID NO as disclosed herein usually implies that (at least) one copy of said sequence is present, e g. in the JNK inhibitor molecule.
  • one inhibitory (poly-)peptide sequence will usually suffice to achieve sufficient inhibition of JNK activity.
  • it is contemplated according to the invention to use two or more copies of the respective sequence e.g. two or more copies of an inhibitory (poly-)peptide sequence of different or same type and/or two or more copies of a transporter sequence of different or the same type
  • inventive (poly)peptide may also employed for the inventive (poly)peptide, as long as the overall ability of the resulting molecule to inhibit JNK activity is not abolished (i.e. the respective molecule is still a JNK inhibitor as defined herein).
  • inventive JNK inhibitors may be obtained or produced by methods well-known in the art, e.g. by chemical synthesis via solid-phase peptide synthesis using Fmoc (9-fluorenylmethyloxycarbonyl) strategy, i.e. by successive rounds of Fmoc deprotection and Fmoc-amino acid coupling cycles.
  • Fmoc (9-fluorenylmethyloxycarbonyl) strategy i.e. by successive rounds of Fmoc deprotection and Fmoc-amino acid coupling cycles.
  • a - - commercial service offering such peptide synthesis is provided by many companies, for example the company Polypeptide (St ⁇ bourg, France).
  • JNK inhibitors for use according to the present invention may optionally be further modified, in particular at the amino acid residues of the inhibitory (poly-peptide) sequence. Possible modifications may for example be selected from one or more of items (i) to (xiii) of the group consisting of:
  • radioactive labels i.e. radioactive phosphorylation or a radioactive label with sulphur, hydrogen, carbon, nitrogen, etc.
  • colored dyes e.g. digoxygenin, etc.
  • fluorescent groups e.g. fluorescein, etc.
  • (v) groups for immobilization on a solid phase e.g. His-tag, biotin, strep-tag, flag-tag, antibodies, epitopes, etc.
  • a solid phase e.g. His-tag, biotin, strep-tag, flag-tag, antibodies, epitopes, etc.
  • the present invention relates in a further aspect to a JNK inhibitor as disclosed herein modified with modifications selected from (i) to (xi) or modified with a combination of two or more of the elements mentioned under (i) to (xi), and a pharmaceutical composition (see below) comprising such modified JNK inhibitor.
  • JNK inhibitors as defined according to the invention can be formulated in a pharmaceutical composition, which may be applied in the prevention or treatment of any of the diseases as defined herein.
  • a pharmaceutical composition used according to the present invention includes as an active component a JNK inhibitor as defined herein, in particular a JNK inhibitor - comprising or consisting of an inhibitory (poly-)peptide sequence according to SEQ ID NO: 1 , as defined herein.
  • the active compound is a JNK inhibitor comprising or consisting of an in hibitory (poly-)peptide sequence according to any one of SEQ ID NOs: 2-27, optionally in (covalent) conjugation (via or without a linker sequence) with any suitable transporter sequence; if a transporter sequence is attached, any of the sequences according to any one of SEQ ID NOs: 1 71 - 1 90, or a variant thereof as defined herein, preferably a sequence according to SEQ ID NOs: 1 72, or a variant thereof as defined herein, may be selected.
  • a JNK inhibitor comprising or consisting of an in hibitory (poly-)peptide sequence according to any one of SEQ ID NOs: 2-27, optionally in (covalent) conjugation (via or without a linker sequence) with any suitable transporter sequence; if a transporter sequence is attached, any of the sequences according to any one of SEQ ID NOs: 1 71 - 1 90, or a variant thereof as defined herein, preferably
  • the inventors of the present invention additionally found that the JNK-inhibitors as defined herein, in particular if fused to a transporter sequence; exhibit a particularly pronounced uptake rate into cells involved in the diseases of the present invention. Therefore, the amount of a JNK-inhibitor inhibitor in the pharmaceutical composition to be administered to a subject, may - without being limited thereto - be employed on the basis of a low dose within that composition. Thus, the dose to be administered may be much lower than for peptide drugs known in the art, such as DTS-108 (Florence Meyer-Losic et al., Clin Cancer Res., 2008, 2145-53). Thereby, for example a reduction of potential side reactions and a reduction in costs is achieved by the inventive (poly)peptides.
  • the dose (per kg body weight), e.g. to be administered on a daily basis to the subject is in the range of up to about 10 mmol/kg, preferably up to about 1 mmol/kg, more preferably up to about 100 pmol/kg, even more preferably up to about 1 0 ⁇ /kg, even more preferably up to about 1 pmol/kg, even more preferably up to about 100 nmol/kg, most preferably up to about 50 nmol/kg.
  • the dose range may preferably be from about 0,01 pmol/kg to about 1 mmol/kg, from about 0, 1 pmol/kg to about 0, 1 mmol/kg, from about 1 ,0 pmol/kg to about 0,01 mmol/kg, from about 10 pmol/kg to about 1 pmol/kg, from about 50 pmol/kg to about 500 nmol/kg, from about 100 pmol/kg to about 300 nmol/kg, from about 200 pmol/kg to about 1 00 nmol/kg, from about 300 pmol/kg to about 50 nmol/kg, from about 500 pmol/kg to about 30 nmol/kg, from about 250 pmol/kg to about 5 nmol/kg, from about 750 pmol/kg to about 1 0 nmol/kg, from about 1 nmol/kg to about 50 nmol/kg, or a combination of any two of said values.
  • a "safe and effective amount" for components of the pharmaceutical compositions as used according to the present invention means an amount of each or all of these components, that is sufficient to significantly induce a positive modification of diseases or disorders strongly related to J NK signal ling as defined herein.
  • a "safe and effective amount” is small enough to avoid serious side-effects, that is to say to permit a sensible relationship between advantage and risk.
  • the determination of these limits typically lies within the scope of sensible medical judgment.
  • a "safe and effective amount" of such a component wi l l vary in connection with the particular condition to be treated and also with the age and physical condition of the patient to be treated, the severity of th e condition, the duration of the treatment, the nature of the accompanying therapy, of the particular pharmaceutically acceptable carrier used, and simi lar factors, within the knowledge and experience of the accompanying doctor.
  • the pharmaceutical compositions according to the invention can be used accordi ng to the invention for human and also for veterinary medical purposes.
  • the pharmaceutical composition as used according to the present invention may furthermore comprise, in addition to one or more of the JNK i nhibitors, a (compatible) pharmaceutically acceptable carrier, excipient, buffer, stabi lizer or other materials well known to those ski lled in the art.
  • the expression "(compatible) pharmaceutical ly acceptable carrier” preferably i n cludes the l iquid or non-liquid basis of the composition.
  • the term "compatible” means that the constituents of the pharmaceutical composition as used herei n are capable of bei ng mixed with the pharmaceutical ly active component as defi ned above and with one another component in such a manner that no interaction occurs which would substantial ly reduce the pharmaceutical effectiveness of the composition under usual use conditions.
  • Pharmaceutical ly acceptable carriers must, of course, have sufficiently high purity and sufficiently low toxicity to make them suitable for admi nistration to a person to be treated.
  • the pharmaceutical ly acceptable carrier wil l typically comprise one or more (compatible) pharmaceutical ly acceptable l iquid carriers.
  • the composition may comprise as (compatible) pharmaceutical ly acceptable l iquid carriers e.g. pyrogen-free water; isotonic sali ne, i.e. a solution of 0.9 % NaCl, or buffered (aqueous) solutions, e.g. phosphate, citrate etc.
  • a buffer preferably an aqueous buffer, and/or 0.9 % NaCI may be used.
  • the pharmaceutically acceptable carrier will typically comprise one or more (compatible) pharmaceutically acceptable solid carriers.
  • the composition may comprise as (compatible) pharmaceutically acceptable solid carriers e.g. one or more compatible solid or liquid fillers or diluents or encapsulating compounds may be used as well, which are suitable for administration to a person.
  • suitable pharmaceutically acceptable solid carriers are e.g.
  • sugars such as, for example, lactose, glucose and sucrose
  • starches such as, for example, corn starch or potato starch
  • cellulose and its derivatives such as, for example, sodium carboxymethylcellulose, ethylcellulose, cellulose acetate
  • powdered tragacanth malt
  • gelatin gelatin
  • tallow solid glidants, such as, for example, stearic acid, magnesium stearate; calcium sulphate, etc.
  • the precise nature of the (compatible) pharmaceutically acceptable carrier or other material may depend on the route of administration.
  • the choice of a (compatible) pharmaceutically acceptable carrier may thus be determined in principle by the manner in which the pharmaceutical composition as used according to the invention is administered.
  • Various possible routes of administration are listed in the list "Route of Administration” of the FDA (cf. FDA: Data Standards Manual - Drug Nomenclature Monographs - Monograph Number: C-DRG-00301 ; Version Number 004), which is in corporated by reference herein. Further guidance for selecting an appropriate route of administration, in particular for non-human animals, can be found in Turner PV et al. (201 1 ) Journal of the American Association for Laboratory Animal Science, Vol. 50, No 5, p.
  • routes for administration include, for example, parenteral routes (e.g. via injection), such as intravenous, intramuscular, subcutaneous, intradermal, or transdermal routes, etc., enteral routes, such as oral, or rectal routes, etc., topical routes, such as nasal, or intranasal routes, etc., or other routes, such as epidermal routes or patch delivery.
  • parenteral routes e.g. via injection
  • enteral routes such as oral, or rectal routes, etc.
  • topical routes such as nasal, or intranasal routes, etc.
  • other routes such as epidermal routes or patch delivery.
  • routes for administration include, for example, parenteral routes (e.g. via injection), such as intravenous, intramuscular, subcutaneous, intradermal, or transdermal routes, etc., enteral routes, such as oral, or rectal routes, etc., topical routes, such as nasal, or intranasal routes, etc., or other routes, such as epidermal routes or patch delivery.
  • routes for systemic administration include, for example, parenteral routes (e.g. via injection and/or infusion), such as intravenous, intra-arterial, intraosseous, intramuscular, subcutaneous, intradermal, transdermal, or transmucosal routes, etc., and enteral - - routes (e.g. as tablets, capsules, suppositories, via feeding tubes, gastrostomy), such as oral, gastrointestinal or rectal routes, etc..
  • parenteral routes e.g. via injection and/or infusion
  • enteral - - routes e.g. as tablets, capsules, suppositories, via feeding tubes, gastrostomy
  • oral, gastrointestinal or rectal routes etc.
  • Systemic administration a system-wide action can be achieved and systemic administration is often very convenient, however, depending on the circumstances it may also trigger unwanted "side-effects" and/or higher concentrations of the JNK inhibitor according to the i nvention may be necessary as compared to local administration.
  • Systemic admi nistration is in general applicable for the prevention and/or treatment of the diseases/disorders mentioned herei n due to its system-wide action.
  • Preferred routes of systemic administration are intravenous, intramuscular, subcutaneous, oral and rectal admi nistration, whereby intravenous and oral administration are particularly preferred.
  • Topical administration typically refers to application to body su rfaces such as the skin or mucous membranes, whereas the more general term
  • Warlocal administration additional ly comprises application i n and/or into specific parts of the body.
  • Topical appl ication is particularly preferred for the treatment and/or prevention of diseases and/or disorders of the skin and/or subcutaneous tissue as defined herein as wel l as for certain diseases of the mouth and/or diseases relating to or are accessible by mucous membranes.
  • Routes for local administration include, for example, inhalational routes, such as nasal, or intranasal routes, ophtalamic and otic drugs, e.g. eye drops and ear drops, administration through the mucous membranes in the body, etc., or other routes, such as epidermal routes, epicutaneous routes (appl ication to the ski n) or patch delivery and other local appl ication, e.g. i njection and/or infusion, i nto the organ or tissue to be treated etc..
  • inhalational routes such as nasal, or intranasal routes, ophtalamic and otic drugs, e.g. eye drops and ear drops, administration through the mucous membranes in the body, etc.
  • other routes such as epidermal routes, epicutaneous routes (appl ication to the ski n) or patch delivery and other local appl ication, e.g. i njection and/or infusion, i nto the organ or tissue to be
  • Routes for administration for the pharmaceutical composition as used according to the invention can be chosen according to the desired location of the application dependi ng on the disorder/disease to be prevented or treated.
  • an enteral administration refers to the gastrointestinal tract as appl ication location and includes oral (p.o.), gastroinstestinal and rectal administration, whereby these are typical ly systemic administration routes, which are applicable to the prevention/treatment of the diseases mentioned herein in general.
  • enteral administration is preferred to prevent and/or treat diseases/disorders of the gastrointestinal tract as mentioned herei n, for example inflammatory diseases of the gastrointestinal tract, metabol ic diseases, cancer and tumor diseases, i n particular of - - the gastrointestinal tract etc.
  • the oral route is usual ly the most convenient for a patient and carries the lowest cost. Therefore, oral administration is preferred for convenient systemic administration, if applicable.
  • compositions for oral admi nistration may be in tablet, capsule, powder or l iquid form.
  • a tablet may include a solid carrier as defined above, such as gelatin, and optionally an adjuvant.
  • Liquid pharmaceutical compositions for oral administration general ly may include a l iquid carrier as defined above, such as water, petroleum, animal or vegetable oi ls, mineral oil or synthetic oi l.
  • Physiological saline solution, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included.
  • enteral administration also i ncludes application locations in the proximal gastroi ntesti nal tract without reaching the intestines, for example subli ngual, sublabial, buccal or intragigi ngval application.
  • Such routes of admi nistration are preferred for applications in stomatology, i.e.
  • JNK i nhibitors as disclosed herein, for example pulpitis in general, in particular acute pulpitis, chronic pulpitis, hyperplastic pulpitis, ulcerative pulpitis, irreversible pulpitis and/or reversible pulpitis; periimplantitis; periodontitis i n general, in particular chronic periodontitis, complex periodontitis, simplex periodontitis, aggressive periodontitis, and/or apical periodontitis, e.g.
  • gingivitis in general, in particular acute gingivitis, chronic gingivitis, plaque-induced gingivitis, and/or non-plaque-induced gi ngivitis; pericoronitis, in particu lar acute and chronic pericoronitis; sialadenitis (sialadenitis); parotitis, in particular infectious parotitis and autoimmune parotitis; stomatitis in general, in particular aphthous stomatitis (e.g., minor or major), Bednar's aphthae, periadenitis mucosa necrotica recurrens, recurrent aphthous ulcer, stomatitis herpetiformis, gangrenous stomatitis, denture stomatitis, ulcerative stomatitis, vesicular stomatitis and/or ging
  • Particularly preferred diseases to be treated and/or prevented accordi ng to the invention by these routes of administration are selected from periodontitis, in particular chronic periodontitis, mucositis, oral desquamative disorders, oral liquen planus, pemphigus vulgaris, pulpitis, stomatitis, temporomandibular joint disorder, and peri-implantitis.
  • periodontitis in particular chronic periodontitis, mucositis, oral desquamative disorders, oral liquen planus, pemphigus vulgaris, pulpitis, stomatitis, temporomandibular joint disorder, and peri-implantitis.
  • intragingival administration e.g. by injection into the gums (gingiva) is preferred in stomatology applications, for example for preventing and/or treating periodontitis.
  • disorders/diseases of the mouth, in particular periodontitis may be prevented or treated by sublingual, sublabial, buccal or intragingival application, in particular intragingival application, of the pharmaceutical composition as defined above comprising a dose (per kg body weight) of 100 ng/kg to 100 mg/kg, preferably 10 pg/kg to 10 mg/kg, more preferably of the JNK inhibitor according to the present invention.
  • the diseases of the mouth mentioned herein may also be treated and/or prevented by systemic and, preferably, topical administration of the JNK inhibitor as disclosed herein or the respective pharmaceutical composition.
  • enteral administration also includes strictly enteral administration, i.e. directly into the intestines, which can be used for systemic as well as for local administration.
  • the JNK inhibitor according to the present invention used in the preventention and/or treatment of diseases and/or disorders according to the present invention may be administered to the central nervous system (CNS).
  • CNS central nervous system
  • routes of administration include in particular epidural (peridural), intra-CSF (intra-cerebrospinal fluid), intracerebroventricular (intraventricular), intrathecal and intracerebral administration, for example administration into specific brain regions, whereby problems relating to the blood-brain-barrier can be avoided.
  • CNS routes of administration are preferred if the disease/disorder to be treated is a neural, a neurological and/or a neurodegenerative disease as specified herein.
  • the JNK inhibitor according to the present invention used in the preventention and/or treatment of diseases and/or disorders according to the present invention may be administered at, in or onto the eye.
  • routes of administration include eye drops applied topically, for example onto the conjunctiva, and, intravitreous (IVT), subconjunctival, and posterior juxtascleral administration, e.g. by injection, infusion and/or instillation and/or localized, sustained-release drug delivery (for example in case of the subconjunctival route), whereby eyedrops (for topical application), intravitreous (IVT) and subconjunctival routes of administration are particularly preferred.
  • IVTT intravitreous
  • subconjunctival subconjunctival routes of administration are particularly preferred.
  • the subconjunctival route is safer and less invasive than the intravitreal route, however, the intravitreal route involves less systemic exposure than the subconjunctival route due to the presence of conjunctival and orbital blood vessels and tissue.
  • Eye-related diseases/disorders to be treated and/or prevented as disclosed herei n, for example age-related macu lar degeneration (AMD), i n particular i n the wet and dry form; angioid streaks; anterior ischemic optic neuropathy; anterior uveitis; cataract, in particular age related cataract; central exudative chorioretinopathy; central serous chorioretinopathy; chalazion; chorioderemia; chorioiditis; choroidal sclerosis; conjunctivitis; cyclitis; diabetic reti nopathy; dry eye syndrome; endophthalmitis; episcleritis; eye infection; fundus albipunctatus; gyrate atrophy of choroid and retina; hordeolum; inflammatory diseases of the blephara; inflammatory diseases of the choroid; inflammatory diseases of the ciliary body; inflammatory diseases of the conjunctiva;
  • AMD age-related macu lar
  • Aqueous tear-deficient dry eye may refer to Sjogren syndrome dry eye or Non-Sjogren syndrome dry eye.
  • Non-Sjogren syndrome dry eye may be caused by primary or secondary lacrimal gland dysfunction or obstruction of the lacrimal gland ducts.
  • Evaporative dry eye may have intrinsic, e.g. Meibomian gland dysfunction, low bl ink rate or disorders of lid aperture, or extri nsic causes, e.g. ocular surface disorder, lens wear or al legergic rhi nitis.
  • Sjogrens or non-Sjogrens dry eye syndrome are to be treated by the present i nventiuon.
  • instillation e.g. eyedrops, and/or subconjunctival administration, e.g
  • the respective pharmaceutical composition according to the present invention preferably comprises a dose per eye in the range of 10 ng to 100 mg, more preferably in the range of 1 00 ng to 10 mg, even more preferably in the range of 1 pg to 5 mg, and particularly preferable in the range of 100 pg to 1 mg, for example 0.1 , 0.2, or 0.4 mg, of the JNK inhibitor according to the present invention, preferably of the JNK inhibitor according to a sequence of SEQ ID NO. 1 72.
  • One single administration or more administrations, in particular two, three, four or five, administrations of such dose(s) are preferred, whereby subsequent dose(s) may be administered on different days of the treatment schedule.
  • a single dose (per eye) of the JNK inhibitor is preferably in the range of 1 g to 5 mg, preferably 50 pg to 1 ,5 mg, more preferably 500 pg to 1 pg, most preferably 800 pg to 1 mg.
  • the injection volume, in particular for subconjunctival injection may be for example 1 00 ⁇ to 500 ⁇ , e.g. 250 ⁇ .
  • a single dose (per eye) of the JNK inhibitor is preferably in the range of 1 pg to 5 mg, preferably 1 0 pg to 1 ,5 mg, more preferably 50 pg to 1 mg, most preferably 100 pg to 600 pg.
  • a single dose or repeated doses may be administered, preferably daily, for example daily 2 to 4 times per day, preferably daily 3 times a day, for several weeks, preferably 2 to 4 weeks, more preferably 3 weeks.
  • Such an administration is for example particularly useful to treat and/or prevent dry eye syndrome.
  • the pharmaceutical composition comprising the JNK inhibitor according to the invention is typically a solution, preferably an ophthalamic solution, e.g. comprising (sterile) 0.9 % NaCI.
  • a pharmaceutical composition comprises in particular 0.001 % - 1 0 % of the JNK inhibitor as described herein, preferably 0.01 % - 5 % of the JNK inhibitor as described herein, more preferably 0.05 % - 2 % of the JNK inhibitor as described herein, even more preferably 0.1 % - 1 % of the JNK inhibitor as described herein.
  • the eyedrops may be administered once or repeatedly, whereby repeated administration is preferred.
  • the administration - - depends on the need and may for example be on demand.
  • subsequent dose(s) may be admi nistered on different days of the treatment schedule, whereby on the same day a single dose or more than one si ngle doses, i n particular two, three, four or five, preferably two or three doses may be administered, whereby such repeated administration is preferably spaced by intervals of one or more hour(s), e.g. two, three, four, five, six, seven or eight hours.
  • eye diseases as described herei n may of course also be treated and/or prevented by systemic application of the J NK inhibitor according to the i nvention (which also applies to the other di seases/disorders as described herein).
  • the dose for systemic admi nistration in eye diseases ranges preferably from 0.001 mg/kg to 1 0 mg/kg, more preferably from 0.01 mg/kg to 5 mg/kg, even more preferably from 0.1 mg/kg to 2 mg/kg.
  • Such doses are for example particularly useful to treat and/or prevent uveitis, whereby the treatment schedule may comprises a single dose or repeated doses, whereby subsequent dose(s) may be administered on different days of the treatment schedule.
  • the doses are typical ly spaced by intervals of at least one day, preferably by intervals of at least two days, more preferably by intervals of at least three days, even more preferably by i ntervals of at least four days, at least five days, or at least six days, particularly preferably by intervals of at least a week, most preferably by intervals of at least ten days.
  • Other routes of admi nistration for the use of the JNK i nhibitor according to the present invention include - but are not limited to - epicutaneous appl ication (onto the skin) and/or i ntralesional appl ication (i nto a skin lesion), for example for skin diseases as defined herein (mentioned herein), in particular selected from psoriasis, eczema, dermatitis, acne, mouth ulcers, erythema, lichen plan, sarcoidose, vascularitis, and adult linear IgA disease; nasal administration, for example for diseases of the respiratory system and in particular lung diseases, for example acute respiratory distress syndrome (ARDS), asthma, chronic il lnesses involving the respiratory system, chronic obstructive pulmonary disease (COPD), cystic fibrosis, inflammatory lung diseases, pneumonia, and pulmonary fibros
  • ARDS acute respiratory distress syndrome
  • COPD chronic obstructive pulmonary disease
  • i nto the uri nary bladder for example for diseases of the uri nary system, i n particular the uri nary bladder; intracardiac administration, intracavernous admi nistration, intravaginal administration, and intradermal administration.
  • the method of administration depends on various factors as mentioned above, for example the selected pharmaceutical carrier and the nature of the pharmaceutical preparation (e.g. as a liquid, tablet etc.) as well as the route of administration.
  • the pharmaceutical composition comprising the JNK inhibitor according to the invention may be prepared as a liquid, for example as a solution of the JNK inhibitor according to the invention in 0.9 % NaCI.
  • a liquid pharmaceutical composition can be administered by various methods, for example as a spray (e.g., for inhalational, intranasal etc. routes), as a fluid for topical application, by injection, including bolus injection, by infusion, for example by using a pump, by instillation, but also p.o., e.g.
  • a syringe including a pre-filled syringe
  • an injection device e.g. the INJECT-EASETTM and GENJECTTTM device
  • an infusion pump such as e.g. Accu-ChekTM
  • an injector pen such as the CENPENTTM
  • a needleless device e.g. M EDDECTO TM and BIOJECTORTM
  • an autoinjector e.g. M EDDECTO TM and BIOJECTORTM
  • the suitable amount of the pharmaceutical composition to be used can be determined by routine experiments with animal models. Such models include, without implying any limitation, for example rabbit, sheep, mouse, rat, dog, gerbil, pig, and non-human primate models.
  • Preferred unit dose forms for administration, in particular for injection and/or infusion include sterile solutions of water, physiological saline or mixtures thereof. The pH of such solutions should be adjusted to about 7.4.
  • Suitable carriers for administration, in particular for injection and/or infusion include hydrogels, devices for controlled or delayed release, polylactic acid and collagen matrices.
  • Suitable pharmaceutically acceptable carriers for topical application include those, which are suitable for use in lotions, creams, gels and the like.
  • tablets, capsules and the like are the preferred unit dose form.
  • the pharmaceutically acceptable carriers for the preparation of unit dose forms, which can be used for oral administration are well known in the prior art. The choice thereof will depend on secondary considerations such as taste, costs and storability, which are not critical for the purposes of the present invention, and can be made without difficulty by a person skilled in the art.
  • the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability.
  • a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability.
  • isotonic vehicles such as Sodium Chloride Injection, in particular 0.9 % NaCI, Ringer's Injection, Lactated Ringer's Injection.
  • Preservatives, stabilizers, - - buffers, antioxidants and/or other additives may be included, as required.
  • administration is preferably in a "prophylactically effective amount or a "therapeutically effective amount” (as the case may be), this being sufficient to show benefit to the individual.
  • a proliferatively effective amount or a "therapeutically effective amount” (as the case may be)
  • the actual amount administered, and rate and time-course of administration will depend on the nature and severity of what is being treated. For example, for i.v.
  • single doses of up to 1 mg per kg body weight are preferred, more preferably up to 500 pg per kg body weight, even more preferably up to 1 00 pg per kg body weight, for example in the range of 100 ng to 1 mg per kg body weight, more specifically in the range of 1 pg to 500 pg per kg body weight, even more specifically in the range of 5 pg to 1 00 pg per kg body weight.
  • Such doses may be administered for example as injection and/or infusion, in particular as infusion, whereby the duration of the infusion varies for example between 1 to 90 min, preferably 10 to 70 min, more preferably 30 to 60 min.
  • the pharmaceutical composition as used according to the present invention may additionally - i.e.
  • nucleic acids, cells or cells transfected with a vector and/or nucleic acids as defined above also comprise optionally a further "active component", which is also useful in the respective disease.
  • the pharmaceutical composition according to the present invention may also combined in the therapy of the diseases according to the present invention with a further pharmaceutical composition comprising a further "active component".
  • a pharmaceutical composition comprising a JNK inhibitor according to the present invention may be used in post-surgery intraocular inflammation as stand-alone therapy or in combination with corticosteroids, preferably glucocorticoids, e.g. dexamethasone.
  • a pharmaceutical composition comprising a JNK inhibitor and/or chimeric peptide according to the present invention may preferably be used in the prevention and/or treatment of Alzheimer's Disease and/or Mild Cognitive Impairment, in particular MCI due to Alzheimer's disease, as stand-alone therapy or in combination with PKR inhibitors and, optionally, in addition to the JNK inhibitor according to the present invention and the PKR inhibitor with a amyloid lowering agent.
  • PKR inhibitors are in particular peptides, e.g. "SO 481 " by Polypeptide Group.
  • Amyloid lowering agents include ⁇ -secretase (BACE1 ) inhibitors, y-secretase inhibitors (GSI) and modulators (GSM).
  • Non- limiting examples of such amyloid lowering agents which are currently in clinical trials may be retrieved from Vassar R. (2014) BACE1 inhibitor drugs in clinical trials for Alzheimer's disease. Alzheimers Res Ther.;6(9):89 and/or from Jia Q, Deng Y, Qing H (2014) Potential therapeutic strategies for Alzheimer's disease targeting or beyond ⁇ -amyloid: insights from clinical trials. Biomed Res Int. 2014;2014:8371 57; for example Pioglitazone, CTS-21 1 66, MK8931 , LY2886721 , - -
  • the administration of the JNK inhibitor according to the present invention may be before, during (concomitant or overlapping administration) or after the administration of the other active component comprised in a separate pharmaceutical composition, for example the PKR inhibitor, the amyloid lowering agent or the glucocorticoid.
  • Administration "before” the administration of the JNK inhibitor preferably means within 24 h, more preferably within 12 h, even more preferably within 3 h, particularly preferably within 1 h and most preferably within 30 min before the administration of the JNK inhibitor starts.
  • Administration "after” the administration of the JNK inhibitor preferably means within 24 h, more preferably within 12 h, even more preferably within 3 h, particularly preferably within 1 h and most preferably within 30 min after the administration of the JNK inhibitor is finished.
  • JNK inhibitor for example a JNK inhibitor comprising or consisting of an inhibitory (poly)peptide sequence according to any of sequences of SEQ ID NOs: 2 to 27, potentially comprising an additional transporter sequence, whereby any of the sequences according to any one of SEQ ID NOs: 1 71 - 1 90, or a variant thereof as defined herein, are preferred and the sequence according to SEQ ID NO: 1 72, or a variant thereof as defined herein, are particularly preferred - include (but are not limited to) the prevention and/or treatment of the following diseases/disorders:
  • diseases of the mouth and/or the jaw bone in particular inflammatory diseases of the mouth and/or the jaw bone selected from (i) pulpitis in general, in particular acute pulpitis, chronic pulpitis, hyperplastic pulpitis, ulcerative pulpitis, irreversible pulpitis and/or reversible pulpitis; (ii) periimplantitis; (iii) periodontitis in general, in particular chronic periodontitis, complex periodontitis, simplex periodontitis, aggressive periodontitis, and/or apical periodontitis, e.g.
  • gingivitis in general, in particular acute gingivitis, chronic gingivitis, plaque-induced gingivitis, and/or non-plaque-induced gingivitis;
  • pericoronitis in particular acute and chronic pericoronitis; sialadenitis (sialoadenitis); parotitis, in particular infectious parotitis and autoimmune parotitis;
  • stomatitis in general, in particular aphthous stomatitis (e.g., minor or major), Bednar's aphthae, periadenitis mucosa necrotica recurrens, recurrent aphthous ulcer, stomatitis herpetiformis, gangrenous stomatitis, - - denture stomatitis, ulcerative stomatitis, vesicular stomatit
  • nephrological diseases in particular selected from (i) glomerulonephritis, for example nonproliferative glomerulonephritis, in particular minimal change disease, focal segmental glomerulosclerosis, focal segmental glomerular hyalinosis and/or sclerosis, focal glomerulonephritis, membranous glomerulonephritis, and/or thin basement membrane disease, and proliferative glomerulonephritis, in particular membrano-proliferative glomerulonephritis, mesangio-proliferative glomerulonephritis, endocapillary proliferative glomerulonephritis, mesangiocapillary proliferative glomerulonephritis, dense deposit disease (membranoproliferative glomerulonephritis type II), extracapillary glomerulonephritis (crescentic glomerulonephritis), rapidly
  • (i i i) diseases of the eye in particular (i) dry eye syndrome; (i i) uveitis, i n particular anterior, intermediate and/or posterior uveitis, sympathetic uveitis and/or panuveitis, preferably anterior and/or posterior uveitis; (iii) age-related macular degeneration (AMD), i ncluding exudative and/or non-exudative age-related macular degeneration, preferably the wet or the dry form of age-related macular degeneration; (iv) reti nopathy, i n particular selected from diabetic retinopathy, (arterial hypertension induced) hypertensive retinopathy, exudative retinopathy, radiation induced retinopathy, sun-i nduced solar retinopathy, trauma-i nduced reti nopathy, e.g.
  • the JNK inhibitor is preferably appl ied as eye drops, which may be applied to both eyes or to one eye only, wherei n the pharmaceutical composition comprising the JNK inhibitor accordi ng to the i nvention is typical ly a sol ution, preferably an ophthalamic solution, e.g.
  • a pharmaceutical composition comprises i n particular 0.001 % - 1 0 % of the JNK inhibitor as described herei n, preferably 0.01 % - 5 % of the J NK i nhibitor as described herei n, more preferably 0.05 % - 2 % of the J NK inhibitor as described herein, even more preferably 0.1 % - 1 % of the JNK inhibitor as described herei n, i n particular for treating and/or preventi ng dry eye syndrome; and/or the JNK inhibitor is preferably applied systemical ly, in particular intravenously, whereby the dose - - ranges preferably from 0.001 mg/kg to 1 0 mg/kg, more preferably from 0.01 mg/kg to 5 mg/kg, even more preferably from 0.1 mg/kg to 2 mg/kg, whereby such administration is for example particularly useful to treat and/or prevent uveitis, whereby the treatment schedule may comprises
  • diseases of the skin in particular papulosquamous disorders, in particular selected from psoriasis in general, for example psoriasis vulgaris, nummular psoriasis, plaque psoriasis, general ized pustular psoriasis, impetigo herpetiformis, Von Zumbusch's disease, acrodermatitis continua, guttate psoriasis, arthropathis psoriasis, distal interphalangeal psoriatic arthropathy, psoriatic arthritis muti lans, psoriatic spondylitis, psoriatic juveni le arthropathy, psoriatic arthropathy in general, and/or flexural psoriasis; parapsoriasis i n general, for example large-plaque parapsoriasis, smal l-plaque parapsoriasis, retiform para
  • the disorder/disease to be prevented and/or treated is psoriasis, for example psoriasis vulgaris, nummular psoriasis, plaque psoriasis, generalized pustular psoriasis, impetigo herpetiformis, Von Zumbusch's disease, acrodermatitis continua, guttate psoriasis, arthropathis psoriasis, distal interphalangeal psoriatic arthropathy, psoriatic arthritis mutilans, psoriatic spondyl itis, psoriatic juvenile arthropathy, psoriatic arthropathy i n general, and/or flexural psoriasis; wherein for the treatment and/or prevention of the skin diseases the JNK inhibitor is preferably applied in doses (per kg body weight) i n the range of 1 pg/kg to 1 00 mg/kg, more preferably 1
  • arthritis and diseases/disorders of the joi nt in particular selected from arthritis in general, osteoarthritis (degenerative joint disease), septic arthritis, rheumatoid arthritis, psoriatic arthritis, and related autoimmune diseases and arthritis; wherein for the treatment and/or prevention of the skin diseases the JNK inhibitor is preferably appl ied in doses (per kg body weight) i n the range of 1 pg/kg to 1 00 mg/kg, more preferably 1 0 pg/kg to 50 mg/kg, even more preferably 50 pg/kg to 1 0 mg/kg, particularly preferably 1 00 pg/kg to 5 mg/kg, if appl icable repeatedly, for example dai ly or weekly for several, e.g.
  • cancer and tumor diseases in particular selected from (i) liver cancer and liver carcinoma in general, in particular liver metastases, liver cell carcinoma, hepatocellular carcinoma, hepatoma, intrahepatic bile duct carcinoma, cholangiocarcinoma, hepatoblastoma, angiosarcoma (of liver), and other specified or unspecified sarcomas and carcinomas of the liver; (ii) prostate cancer and/or prostate carcinoma; and/or (iii) colon cancer and colon carcinoma in general, in particular cecum carcinoma, appendix carcinoma, ascending colon carcinoma, hepatic flexure carcinoma, transverse colon carcinoma, splenic flexure carcinoma, descending colon carcinoma, sigmoid colon carcinoma, carcinoma of overlapping sites of colon and/or malignant carcinoid tumors of the colon, wherein for the treatment and/or prevention of the cancer and tumor diseases the JNK inhibitor is preferably applied in doses (per kg body weight) in the range of 1 pg/kg to 1 00 mg/
  • diseases and/or disorders of the urinary system in particular ureteritis; urinary tract infection (bladder infection, acute cystitis); cystitis in general, in particular interstitial cystitis, Hunner's ulcer, trigonitis and/or hemorrhagic cystitis; urethritis, in particular nongonococcal urethritis or gonococcal urethritis; painful bladder syndrome; IC/PBS; urethral syndrome; and/or retroperitoneal fibrosis; preferably IC/PBS; wherein for the treatment and/or prevention of the diseases and/or disorders of the urinary system, preferably for the treatment and/or prevention of IC/PBS, the JNK inhibitor is preferably applied (i) systemically, more preferably intravenously, e.g.
  • the JNK inhibitor is also preferably applied (ii) intravesical ly, more preferably by intravesical infusion, preferably at a concentration of 10 pg/ml - 1000 mg/ml, more prefarbly 50 pg/ml - 500 mg/ml, even more preferably 100 pg/ml - 100 mg/ml, and particularly preferably 0.5 mg/ml - 50 mg/ml, preferably in single doses of 0.1 - 1 000 mg, more preferably 0.5 - 500 mg, even more preferably 1 - 100 mg, and particularly preferably 2 - 10 mg, preferably administered in one single dose, however, if applicable also preferably administered repeatedly, for example dai ly, every 2 or 3 days or weekly, for several, e.g. 2, 3, 4, 5, 6, 7, 8, 9, or 10, weeks.
  • neural, neuronal or neurodegenerative disorders in particular neurodegenerative disease, preferably Alzheimer's disease, for example Alzheimer's disease with early onset, Alzheimer's disease with late onset, Alzheimer's dementia senile and presenile forms, and/or Mild Cognitive Impairment, in particular Mild Cognitive Impairment due to Alzheimer's Disease, wherein for the treatment and/or prevention of the neural, neuronal or neurodegenerative disorders the JNK inhibitor is preferably applied in doses (per kg body weight) in the range of 1 pg/kg to 100 mg/kg, more preferably 10 pg/kg to 50 mg/kg, even more preferably 100 pg/kg to 1 0 mg kg, and particularly preferably 500 pg/kg to 1 mg/kg, whereby the JNK inhibitor is preferably adminsistered, if applicable, once or repeatedly, preferably weekly (once per week) for several, e.g.
  • the JNK inhibitor is preferably adminsistered, if applicable, once or repeatedly, preferably weekly (once per week) for
  • intra-CSF intra-cerebrospinal fluid
  • the JNK inhibitors of the present invention may be administered as stand-alone therapy, however, the JNK inhibitors of the present invention may also be administered in combination with other medications, e.g. with a PK inhibitor, e.g.
  • amyloid lowering agents include ⁇ -secretase (BACE1 ) inhibitors, ⁇ -secretase inhibitors (GSI) and modulators (GSM) and examples of such inhibitors, which are currently in clinical trials may be retrieved from Vassar R. (2014) BACE1 inhibitor drugs in clinical trials for Alzheimer's disease. Alzheimers Res Ther.;6(9):89 or from Jia Q, Deng Y, Qing H (2014) Potential therapeutic strategies for Alzheimer's disease targeting or beyond ⁇ -amyloid: insights from clinical trials. Biomed Res Int. 2014;2014:8371 57. - -
  • the J NK i nhibitors of the present invention will modulate the JNK activity in the subject.
  • the term "modulate” includes in particular the suppression of phosphorylation of c-jun, ATF2 or NFAT4 in any of the diseases disclosed herein, for example, by using at least one JNK i nhibitor comprising or consisti ng of an inhibitory (poly)peptide sequence according to any of sequences of SEQ ID NOs: 2 to 27, potentially comprising an additional transporter sequence, whereby - if a transporter sequence is attached - any of the sequences according to any one of SEQ ID NOs: 1 71 -1 90, or a variant thereof as defi ned herein, are preferred, and the sequence according to SEQ ID NO: 1 72, or a variant thereof as defi ned herein, is particularly preferred, as a competitive inhibitor of the natural c-
  • modulate also includes suppression of hetero- and homomeric complexes of transcription factors made up of, without being limited thereto, c-jun, ATF2, or NFAT4 and their related partners, such as for example the AP-1 complex that is made up of c-jun, AFT2 and c-fos.
  • Treatment of a subject with the pharmaceutical composition as disclosed above may be typical ly accomplished by administering ⁇ in vivo) an ("therapeutically effective") amount of said pharmaceutical composition to a subject, wherein the subject may be e.g. a human subject or an animal, whereby a human is particularly preferred.
  • the animal is preferably a non-human mammal, e.g., a non-human primate, mouse, rat, dog, cat, cow, horse or pig.
  • a non-human primate mouse, rat, dog, cat, cow, horse or pig.
  • the JNK inhibitor of the present invention for example, a JNK inhibitor comprising or consisting of an inhibitory (poly)peptide sequence according to any of sequences of SEQ ID NOs: 2 to 27, potential ly comprisi ng an additional transporter sequence, whereby - if a transporter sequence is attached - any of the sequences accordi ng to any one of SEQ I D NOs: 1 71 -1 90, or a variant thereof as defined herein, are preferred, and the sequence according to SEQ ID NO: 1 72, or a variant thereof as defi ned herei n, is particularly preferred, may be uti lized for the treatment of a tissue or organ prior to its transplantation.
  • a solution for the isolation, transport, perfusion, implantation or the like of an organ and/or tissue to be transplanted comprises the J NK i nhibitor accordi ng to the present i nvention, preferably in a concentration i n the range of 1 to 1 000 ⁇ , more preferably i n the range of 1 0 to 500 ⁇ , even more preferably in the range of 50 to 1 50 ⁇ .
  • the transplant is a kidney, heart, lung, pancreas, in particular pancreatic islets (also cal led islets of Langerhans), liver, blood cel l, bone marrow, cornea, accidental severed limb, i n particular fingers, hand, foot, face, nose, bone, cardiac valve, blood vessel or intestine transplant, preferably a kidney, heart, pancreas, i n particular - - pancreatic islets (also called islets of Langerhans), or skin transplant.
  • the JNK inhibitor according to the invention may be contained in the solution for the isolation of pancreatic islets. Such a solution may be for example injected into the pancreatic duct prior to isolation.
  • a solution containing the JNK inhibitor according to the invention is applied in isolation, transport, perfusion, transplantation or the like of an organ and/or tissue, in particular if the time of ischemia exceeds 1 5 min, more preferably, if the time of ischemia exceeds 20 min, even more preferably if the time of ischemia is at least 30 min.
  • ischemia times may apply to warm and/or cold ischemia time, however, it is particularly preferred if they apply exclusively to warm ischemia time (WIT), whereby WIT refers to the length of time that elapses between a donor's death, in particular from the time of cross-clamping or of asystole in non-heart-beating donors, until cold perfusion is commenced and to ischemia during implantation, from removal of the organ from ice until reperfusion.
  • WIT warm ischemia time
  • the present invention is directed to specific uses (or methods of use) of the above disclosed JNK inhibitors or pharmaceutical compositions containing the same in a method for treatment of the human or animal body by therapy, in particular of the human body.
  • JNK signalling is involved in a multitude of diverse disease states and disorder and inhibition of said signalling has proposed and successfully tested for many of these.
  • the inventors of the present invention found that the JNK inhibitors disclosed herein are effective JNK inhibitors for the treatment of the diseases as disclosed in the following.
  • Treatment of a human or animal body by therapy refers to any kind of therapeutic treatment of a respective subject. It includes for example prevention of onset of the disease or symptoms (prophylaxis), i.e. typically prior to manifestation of the disease in the patient.
  • the term also includes the "mere" treatment of symptoms of a given disease, i.e. the treatment will ameliorate pathogenesis by reducing disease-associated symptoms, without necessarily curing the underlying cause of the disease and symptoms. Certainly, curing the underlying cause of the disease is also encompassed by the term.
  • the term also encompasses a treatment which delays or even stops progression of the respective disease.
  • the JNK inhibitors according to the present invention may be administered for example prophylactically prior to potential onset of a foreseeable disorder, e.g. prior to a planned surgical intervention or planned exposure to stressful stimuli.
  • a surgical intervention could for example bear the risk of inflammation of the respective wound or neighbouring tissue.
  • Exposure to stressful stimuli like radiation could lead to apoptosis of affected tissue and cells.
  • the JNK inhibitors according to the present invention may, for example, be administered at least once up to about 4 weeks in advance.
  • the JNK inhibitors may for example be administered at least 24 hours, at least 48 hours, at least 1 week, at least 2 weeks or 4 weeks in advance.
  • the diseases and disorders to be treated and/or prevented with the JNK inhibitors as disclosed herein may be acute or chronic.
  • JNK inhibitors of the present invention may be used in general for the treatment and/or prevention of diseases of various organs, such as diseases of the eye, diseases of the bone, neural diseases, neuronal diseases, neurodegenerative diseases, diseases of the skin, immune and/or autoimmune diseases, diseases of the eye, diseases of the mouth, diseases of the kidney, diseases of the urinary system, inflammatory diseases, metabolic diseases, cardiovascular diseases, proliferative diseases (in particular cancers and tumors), diseases of the ear, diseases of the intestine, diseases of the respiratory system (e.g. lung diseases), infectious diseases, and various other diseases, the present invention specifically refers to the following diseases:
  • skin diseases and diseases of the subcutaneous tissue are to be mentioned, in particular inflammatory skin diseases, more specifically skin diseases selected from the group consisting of eczema, Psoriasis, dermatitis, acne, mouth ulcers, erythema, Lichen plan, sarcoidosis, vascularitis and adult linear IgA disease,.
  • Dermatitis encompasses e.g. atopic dermatitis or contact dermatitis.
  • the skin diseases and diseases of the subcutaneous tissue to be treated and/or prevented with the JNK inhibitor as described herein can be selected from papulosquamous disorders in general, in particular psoriasis in general, for example psoriasis vulgaris, nummular psoriasis, plaque psoriasis, generalized pustular psoriasis, impetigo herpetiformis, Von Zumbusch's disease, acrodermatitis continua, guttate psoriasis, arthropathis psoriasis, distal interphalangeal psoriatic arthropathy, psoriatic arthritis mutilans, psoriatic spondylitis, psoriatic juvenile arthropathy, psoriatic arthropathy in general, and/or flexural psoriasis, parapsoriasis in general, for example large-plaque parapsoriasis, small- plaque parapsori
  • Examples for preferred skin diseases which can be treated with the JNK inhibitors of the present invention are psoriasis and lupus erythematosus.
  • skin diseases and diseases of the subcutaneous tissue which can preferably be treated and/or prevented with the JNK inhibitors as disclosed herein are papulosquamous disorders. These include psoriasis, parapsoriasis, pityriasis rosea, lichen planus and other papulosquamous disorders for example pityriasis rubra pilaris, lichen nitidus, lichen striatus, lichen ruber moniliformis, and infantile popular acrodermatitis.
  • the disease to be treated and/or prevented by the JNK inhibitor according to the invention is selected from the group of psoriasis and parapsoriasis, whereby psoriasis is particularly preferred.
  • psoriasis include psoriasis vulgaris, nummular psoriasis, plaque psoriasis, generalized pustular psoriasis, impetigo herpetiformis, Von Zumbusch's disease, acrodermatitis continua, guttate psoriasis, arthropathis psoriasis, distal interphalangeal psoriatic arthropathy, psoriatic arthritis m utilans, psoriatic spondylitis, psoriatic juvenile arthropathy, psoriatic arthropathy in general, and/or flexural psoriasis.
  • parapsoriasis examples include large-plaque parapsoriasis, small- plaque parapsoriasis, retiform parapsoriasis, pityriasis lichenoides and lymphomatoid papulosis.
  • Anti-inflammatory treatment upon tissue or organ transplantation is treatable by the inventive molecules in particular upon heart, kidney, and skin (tissue), lung, pancreas, liver, blood cells (e.g. any kind of blood cell, such as platelets, white blood cells, red blood cells), bone marrow, cornea, accidental severed limbs (fingers, hand, foot, face, nose etc.), bones of whatever type, cardiac valve, blood vessels, segments of the intestine or the intestine as such.
  • blood cells e.g. any kind of blood cell, such as platelets, white blood cells, red blood cells
  • bone marrow e.g. any kind of blood cell, such as platelets, white blood cells, red blood cells
  • cornea e.g. any kind of blood cell, such as platelets, white blood cells, red blood cells
  • accidental severed limbs fis, hand, foot, face, nose etc.
  • bones of whatever type e.g. a graft vs. host or host vs graft reaction occurs upon organ/
  • inventive molecules may also be employed whenever transplantation surgery is carried, in particular in case of skin (or, pancreas, liver, lung, heart, kidney) graft vs. host or host vs. skin (or, pancreas, liver, lung, heart, kidney) graft reaction.
  • neurodegenerative diseases in particular those associated with chronic inflammation, tauopathies and amyloidoses and prion diseases are addressed by the inventive molecules.
  • Other such neurodegenerative disease refer to the various forms of dementia, e.g. frontotemporal dementia and dementia with lewy bodies, schizophrenia and bipolar disorder, spinocerebellar ataxia, spinocerebellar atrophy, multiple system atrophy, motor neuron disease, corticobasal degeneration, progressive supranuclear palsy or hereditary spastic paraparesis.
  • Another field of indication is pain (e.g. neuropathic, incident, breakthrough, psychogenic, phantom, chronic or acute forms of pain).
  • Another field of use is the treatment of bladder diseases, in particular for treating loss of bladder fu nction (e.g. urinary incontinence, overactive bladder, interstitial cystitis or bladder cancer) or stomatitis.
  • the i nventive molecules are used for the treatment of fibrotic diseases or fibrosis as wel l, in particular lung, heart, liver, bone marrow, mediastinum, retroperitoneum, skin, intestine, joint, and shoulder fibrosis.
  • Whi le inflammatory diseases of the mouth and the jaw/mandible are treatable in general by the inventive molecules, gingivitis, osteonecrosis (e.g. of the jaw bone), peri-implantitis, pulpitis, and periodontitis are particularly suitable for the use of these inventive molecules for therapeutic purposes.
  • diseases and/or disorders of the mouth or the jaw bone to be treated and/or prevented with the J NK inhibitor as described herein can be selected from pulpitis in general, in particular acute pulpitis, chronic pulpitis, hyperplastic pulpitis, ulcerative pulpitis, irreversible pulpitis and/or reversible pulpitis; peri implantitis; periodontitis in general, i n particular chronic periodontitis, complex periodontitis, simplex periodontitis, aggressive periodontitis, and/or apical periodontitis, e.g.
  • gingivitis in general, in particular acute gingivitis, chronic gingivitis, plaque-induced gingivitis, and/or non- pl aque-i nduced gi ngivitis; pericoronitis, in particular acute and chronic pericoronitis; sialadenitis (sialoadenitis); parotitis, in particular infectious parotitis and autoimmune parotitis; stomatitis in general, in particular aphthous stomatitis (e.g., minor or major), Bednar's aphthae, periadenitis mucosa necrotica recurrens, recurrent aphthous ulcer, stomatitis herpetiformis, gangrenous stomatitis, denture stomatitis, ulcerative stomatitis, vesicular sto
  • polypes are effectively treatable by using the inventive molecules.
  • the disease is selected from the group consisti ng of glomerulonephritis in general, in particular membrano-proliferative glomerulonephritis, mesangio- proliferative glomerulonephritis, rapidly progressive glomerulonephritis, acute kidney injury (" ⁇ , also cal led "acute renal fai lure” or "acute kidney fai lure") in general, i n particular prerenal AKI, intrinsic AKI, postrenal AKI, AKI with tubular necrosis for example acute tubular necrosis, renal tubular necrosis, AKI with cortical necrosis for example acute cortical necrosis and renal cortical necrosis, AKI with medul lary necrosis, for example medul lary (papil lary) necrosis, acute medullary - -
  • papil lary necrosis and chronic medul lary (papi l lary) necrosis, or other AKI
  • nephrophathies in general, i n particular membranous nephropathy or diabetic nephropathy, nephritis i n general, in particular lupus nephritis, pyelonephritis, interstitial nephritis, tubulointerstitial nephritis, chronic nephritis or acute nephritis, and minimal change disease and focal segmental glomerulosclerosis.
  • diseases and/or disorders of the kidney can be selected from glomerulonephritis in general, for example nonproliferative glomerulonephritis, i n particular minimal change disease, focal segmental glomerulosclerosis, focal segmental glomerular hyal inosis and/or sclerosis, focal gl omerulonephritis, membranous glomerulonephritis, and/or thin basement membrane disease, and proliferative glomerulonephritis, in particular membrano-prol iferative glomerulonephritis, mesangio-prol iferative glomerulonephritis, endocapi l lary proliferative glomerulonephritis, mesangiocapi l lary prol iferative glomerulonephritis, dense deposit disease (membranoproliferative glomerular glomerulonephritis, i n particular minimal change disease, focal segmental glomerul
  • Glomerulonephritis refers to several renal diseases, whereby many of the diseases are characterised by inflammation either of the glomeruli or small blood vessels in the kidneys, but not all diseases necessarily have an inflammatory component.
  • Acute kidney injury (“AKI”, also called “acute renal failure” or “acute kidney failure”) is an abrupt loss of kidney function, which is often investigated in a renal ischemia/ reperfusion injury model.
  • Nephropathies i.e. damage to or disease of a kidney, includes also nephrosis, which is non-inflammatory nephropathy, and nephritis, which is inflammatory kidney disease.
  • a larger number of diseases or disorders may be linked to inflammatory processes, but do not necessarily have to be associated with such inflammatory processes.
  • the following diseases or disorders are specifically disclosed in this regard as being treatable by the use of the inventive molecules: Addison's disease, Agammaglobulinemia, Alopecia areata, Amytrophic lateral sclerosis, Antiphospholipid syndrome, Atopic allergy, Autoimmune aplastic anemia, Autoimmune cardiomyopathy, Autoimmune enteropathy, Autoimmune hemolytic anemia, Autoimmune inner ear, disease, Autoimmune lymphoproliferative syndrome, Autoimmune polyendocrine syndrome, Autoimmune progesterone dermatitis, Idiopathic thrombocytopenic purpura, Autoimmune urticaria, Balo concentric sclerosis, Bullous pemphigoid, Castleman's disease, Cicatricial pemphigoid, Cold agglutinin disease, Comp
  • any kind of inflammatory eye disease may be treated by the use of the inventive molecules, the following eye-related diseases are specifically dislosed: inflammation after corneal surgery, non- infective keratitis, chorioretinal inflammation, and sympathetic ophthalmia.
  • the JNK inhibitors as described herein can be used to treat and/or prevent inflammatory and noninflammatory diseases of the eye, in particular selected from uveitis, in particular anterior, - - intermediate and/or posterior uveitis, sympathetic uveitis and/or panuveitis; scleritis in general, in particular anterior scleritis, brawny scleritis, posterior scleritis, and scleritis with corneal involvement; episcleritis in general, in particular episcleritis periodica fugax and nodular episcleritis; retinitis; conjunctivitis in general, in particular acute conjunctivitis, mucopurulent conjunctivitis, atopic conjunctivitis, toxic conjunctivitis, pseudomembraneous conjunctivitis, serous conjunctivitis, chronic conjunctivitis, giant pupillary conjunctivitis, follicular conjunc
  • the "dry” form of advanced AMD results from atrophy of the retinal pigment epithelial layer below the retina, which causes vision loss through loss of photoreceptors (rods and cones) in the central part of the eye.
  • Neovascular the "wet” form of advanced AMD, causes vision loss due to abnormal bl ood vessel growth (choroidal neovascularization) in the choriocapillaris, through Bruch's membrane, ultimately leading to blood and protein leakage below the macula. Bleeding, leaking, and scarring from these blood vessels eventually cause irreversible damage to the photoreceptors and rapid vision loss, if left untreated.
  • the inventive molecules are suitable for treating both forms of AMD.
  • the JNK inhibitors of the present invention can be used to treat and/or prevent Dry eye syndrome (DES).
  • Dry eye syndrome also called keratitis sicca, xerophthalmia, keratoconjunctivitis sicca (KCS) or cornea sicca
  • DES Dry eye syndrome
  • KCS keratoconjunctivitis sicca
  • cornea sicca is an eye disease caused by eye dryness, which, in tu rn, is caused by either decreased tear production or increased tear film evaporation.
  • Typical symptoms of dry eye syndrome are dryness, burning and a sandy-gritty eye irritation. Dry eye syndrome is often associated with ocular surface inflammation. If dry eye syndrome is left untreated or becomes severe, it can produce complications that can cause eye damage, resulting in impaired vision or even in the loss of vision.
  • Untreated dry eye syndrome can in particular lead to pathological cases in the eye epithelium, squamous metaplasia, loss of goblet cells, thickening of the corneal su rface, corneal erosion, punctate keratopathy, epithelial defects, corneal ulceration, corneal neovascularization, corneal scarring, corneal thinning, and even corneal perforation.
  • the JNK inhibitors according to the present invention may be utilized in treatment and/or prevention of dry eye syndrome, e.g. due to aging, diabetes, contact lenses or other causes and/or after eye surgery or trauma, in particular after Lasik (laser-assisted in situ keratomileusis), commonly referred to simply as laser eye surgery.
  • the standard treatment of dry eye may involve the administration of artificial tears, cyclosporine (in particular cyclosporine A; e.g. Restasis®); autologous serum eye drops; lubricating tear ointments and/or the administration of (cortico-)steroids, for example in the form of drops or eye ointments.
  • cyclosporine in particular cyclosporine A; e.g. Restasis®
  • autologous serum eye drops lubricating tear ointments and/or the administration of (cortico-)steroids, for example in the form of drops or eye ointments.
  • the present invention also relates to the use of the JNK inhibitor as described herein in a method of treatment of dry eye syndrome, wherein the method comprises the combined administration of the JNK inhibitor as defined herein together with a standard treatment for dry eye, in particular with any one of the above mentioned treatments.
  • Particularly preferred is the combination with cyclosporine A and most preferably with artificial tears.
  • Combined administration comprises the parallel administration and/or subsequent administration (either first the JNK inhibitor described herein and then the (cortico)steroids or vice versa).
  • subsequent and parallel - - administration may also be combined, e.g. the treatment is started with JNK i nhibitors described herein and at a later point in time in the course of the treatment (cortico)steroids are given in parallel, or vice versa.
  • the JNK inhibitors of the present invention can be used to treat and/or prevent inflammatory diseases of the sclera, the cornea, the iris, the ci liary body, the retina and/or the choroid of the eye.
  • the JNK inhibitors of the present invention can be used to treat and/or prevent uveitis, i.e. an inflammation of the uvea.
  • uveitis i.e. an inflammation of the uvea.
  • the uvea consists of the middle, pigmented vascular structures of the eye and includes the iris, the ci liary body, and the choroid.
  • uveitis is classified as anterior uveitis, intermediate uveitis, posterior uveitis, and/or panuveitis, whereby the latter is the inflammation of al l the layers of the uvea.
  • uveitis i n cludes sympathetic ophthalmia (sympathetic uveitis), which is a bilateral diffuse granulomatous uveitis of both eyes fol lowing trauma to one eye.
  • Anterior uveitis which is particularly preferred to be treated with the JNK i nhibitors of the present invention, i ncludes iridocyclitis and ulceris. Iritis is the inflammation of the anterior chamber and iris. Iridocyclitis presents the same symptoms as ulceris, but also includes inflammation in the vitreous cavity.
  • iridocyclitis to be prevented and/or treated with the JNK inhibitors of the present invention i nclude - but are not limited to - acute iri docyclitis, subacute iridocycl itis and chronic iridocyclitis, primary iridocycl itis, recurrent iridocyclitis and secondary iridocyclitis, lens-induced iridocycl itis, Fuchs' heterochromic cyclitis, and Vogt-Koyanagi syndrome.
  • Intermediate uveitis, also known as pars planitis in particular includes vitritis, which is inflammation of cells in the vitreous cavity, sometimes with "snowbanking" or deposition of inflammatory material on the pars plana.
  • Posterior uveitis includes i n particular chorioretinitis, which is the inflammation of the retina and choroid, and chorioditis (choroid only).
  • the JNK inhibitors as disclosed herein can be used to treat and/or prevent chorioretinal inflammation in general, for example focal and/or disseminated chorioretinal i nflammation, chorioretinitis, chorioditis, retinochoroiditis, posterior cyclitis, Harada's disease, chorioreti nal inflammation i n infectious and parasitic diseases and/or retinitis, i.e. an inflammation of the reti na.
  • Inflammatory diseases damaging the reti na of the eye in general are i ncluded, in addition to retinitis in particular reti nal vasculitis, for example Eales disease and retinal perivasculitis.
  • an inflammation of the sclera for example anterior scleritis, brawny scleritis, posterior scleritis, scleritis with corneal involvement and scleromalacia perforans; episcleritis, in particular episcleritis periodica fugax and nodular episcleritis; and keratitis, which is an i nflammation of the cornea, i n particular corneal ulcer, superficial keratitis, macular keratitis, filamentary keratitis, photokeratitis, punctate keratitis, keratoconjunctivitis, for example exposure - - keratoconjunctivitis, keratoconjunctivitis sicca (dry eyes), neurotrophic keratoconjunctivitis, ophthalmia nodosa, phlyctenular keratoconjunctivitis, vernal keratoconjunctivitis and
  • the JNK inhibitors as disclosed herein are particularly useful to treat and/or prevent post- surgery (or "post-procedural") or post-trauma inflammation of the eye.
  • Post-surgery refers in particular to a surgery performed on and/or in the eye, for example cataract surgery, laser eye surgery, glaucoma surgery, refractive surgery, corneal surgery, vitreo-retinal surgery, eye muscle surgery, oculoplastic surgery, and/or surgery involving the lacrimal apparatus.
  • the surgery referred to in “post-surgery” is a complex eye surgery and/or an uncomplicated eye surgery.
  • JNK i nhibitors as disclosed herein to treat and/or prevent post- surgery or post-trauma intraocular i nflammation, which may be for example (but not limited to) inflammation of postprocedural bleb.
  • retinopathy Another particularly preferred eye disease to be treated and/or prevented with the J NK inhibitors according to the i nvention is retinopathy.
  • reti nopathy include diabetic reti nopathy, hypertensive retinopathy (e.g., arterial hypertension i nduced), exudative retinopathy, radiation induced retinopathy, sun-induced solar retinopathy, trauma-induced reti nopathy, e.g. Purtscher's retinopathy, retinopathy of prematurity (ROP) and/or hyperviscosity-related retinopathy, non-diabetic proliferative retinopathy, and/or prol iferative vitreo-retinopathy.
  • ROP retinopathy of prematurity
  • the JNK inhibitors as disclosed herein are particularly preferred for the treatment and/or prevention of diabetic reti nopathy and reti nopathy of prematurity, respectively.
  • Reti nopathy of prematurity ROP
  • RLF retrolental fibroplasia
  • ROP can be mi ld and may resolve spontaneously, but it may lead to bl i ndness i n serious cases. As such, al l preterm babies are at risk for ROP, and very low birth weight is an additional risk factor. Both oxygen toxicity and relative hypoxia can contribute to the development of ROP.
  • the inventive molecules are suitable for treating ROP.
  • the i nventive molecules are particularly suitable to treat all forms of reti nopathy, i n particular diabetes mel l itus i nduced reti nopathy, arterial hypertension induced hypertensive reti nopathy, radiation i nduced retinopathy (due to exposure to ionizing radiation), sun-induced solar - - retinopathy (exposure to sunl ight), trauma-induced reti nopathy (e.g. Purtscher's reti nopathy) and hyperviscosity-related retinopathy as seen in disorders which cause paraproteinemia).
  • reti nopathy i n particular diabetes mel l itus i nduced reti nopathy
  • arterial hypertension induced hypertensive reti nopathy due to exposure to ionizing radiation
  • sun-induced solar - - retinopathy exposure to sunl ight
  • trauma-induced reti nopathy e.g. Purtscher's reti nopathy
  • hyperviscosity-related retinopathy
  • the J NK inhibitors as disclosed herein are particularly useful to treat and/or prevent arthritis and related disease and/or disorders of joint.
  • Arthritis is a form of joi nt disorder that involves inflammation of one or more joints.
  • the most common form, osteoarthritis (degenerative joint disease), is a result of trauma to the joint, i nfection of the joint, or age.
  • Other arthritis forms are rheumatoid arthritis, psoriatic arthritis, and related autoimmune diseases.
  • Septic arthritis is caused by joi nt i nfection.
  • joint pai n is primary, and is considered the mai n feature.
  • the arthritis is considered to be secondary to the main disease; these include psoriasis (Psoriatic arthritis), reactive arthritis, Ehlers- Danlos Syndrome, haemochromatosis, hepatitis, Lyme disease, Sjogren's disease, Hashimoto's Thyroiditis, Inflammatory bowel disease (including Crohn's disease and ulcerative colitis), Henoch- Schonlein purpura, Hyperimmunoglobulinemia D with recurrent fever, Sarcoidosis, Whipple's disease, TNF receptor associated periodic syndrome, Wegener's granulomatosis (and many other vasculitis syndromes), Familial Mediterranean fever and sSystemic lupus erythematosus.
  • An undifferentiated arthritis is an arthritis that does not fit into well-known cl inical disease categories, possibly being an early stage of a definite rheumatic disease.
  • diseases and/or disorders arthritis relating to athritis which may be treated and/or prevented with the JNK inhibitors as disclosed herein, can be selected from pyogenic arthritis, in pa rticular staphylococcal arthritis and polyarthritis, pneumococcal arthritis and polyarthritis, other streptococcal arthritis and polyarthritis, and arthritis and polyarthritis due to other bacteria; direct i nfections of joint in infectious and parasitic diseases i n general; postinfective and reactive arthropathies, in particular arthropathy following intesti nal bypass, postdysenteric arthropathy, postimmunozation arthropathy, Reiter's disease, and other reactive arthropathies; inflammatory po lyarthropathies, in particular rheumatoid arthritis with rheumatoid factor, for example Felty's syndrome, rheumatoid lung disease with rheumatoid arthritis, r
  • a further class of inflammatory-associated diseases to be treated by the use of the inventive molecules is the following: acute disseminated encephalomyelitis, antisynthetase syndrome, autoimmune hepatitis, autoimmune peripheral neuropathy, pancreatitis, in particular autoimmune pancreatitis, Bickerstaff's encephalitis, Blau syndrome, Coeliac disease, Chagas disease, chronic inflammatory demyelinating polyneuropathy, osteomyelitis, in particular chronic recurrent multifocal osteomyelitis, Churg-Strauss syndrome, Cogan syndrome, giant-cell arteritis, CREST syndrome, vasculitis, in particular cutaneous small-vessel vasculitis or urticarial vasculitis, dermatitis, in particular dermatitis herpetiformis, dermatomyositis, systemic scleroderma, Dressler's syndrome, drug-induced lupus erythematosus, discoid lupus erythe
  • neuromyelitis optica thyroiditis, in particular Ord's thyroiditis, rheumatism, in particular palindromic rheumatism, Parsonage-Turner syndrome, perivenous encephalomyelitis, polyarteritis nodosa, polymyalgia rheumatica, polymyositis, cirrhosis, in particular primary biliary cirrhosis, cholangitis, in particular primary sclerosing cholangitis, progressive inflammatory neuropathy, Rasmussen's encephalitis, chondritis, - - in particular polychondritis, e.g.
  • the inventive molecules are used for the treatment of the fol lowi ng diseases or disorders: persistent or acute i nflammatory diseases of the ski n, i n particular psoriasis, dry eye disease (Dry Eye Syndrome), uveitis, persistent or acute inflammatory diseases damaging the reti na of the eye, retinopathy, i n particular diabetic retinopathy or retinopathies caused by other diseases, age-related macular degeneration (AMD), i n particular th e wet or the dry form of age-related macular degeneration, retinopathy of prematurity (ROP), persistent or acute i nflammatory diseases of the mouth, i n particular peri-implantitis, pulpitis, periodontitis, anti-inflammatory treatment upon tissue or organ transplantation, i n particular upon heart, kidney, and skin (tissue) transplantation, graft rejection upon heart, kidney or ski n (tissue) transplantation, inflammatory brain diseases
  • the disorder/disease to be prevented and/or treated is a neurodegenerative disease, i n particular tauopathies, preferably Alzheimer's disease, for example Al zheimer's disease with early onset, Alzheimer's disease with late onset, Alzheimer's dementia senile and preseni le forms.
  • a neurodegenerative disease i n particular tauopathies, preferably Alzheimer's disease, for example Al zheimer's disease with early onset, Alzheimer's disease with late onset, Alzheimer's dementia senile and preseni le forms.
  • AD Alzheimer's disease
  • B-amyloid
  • NFTs neurofibri llary tangles
  • AD amyloid precursor protein
  • BACE1 beta-site APP cleaving enzyme 1
  • presenilin 1 the beta-site APP cleaving enzyme 1
  • ⁇ accumulations can lead to synaptic dysfunction, altered kinase activities resulting in NFTs formation, neuronal loss and dementia (Hardy and Higgins, 1 992, Science 256: - -
  • AD pathogenesis is thus bel ieved to be triggered by the accumulation of ⁇ , whereby ⁇ self-aggregates into oligomers, which can be of various sizes, and forms diffuse and neuritic plaques in the parenchyma and blood vessels.
  • ⁇ ol igomers and plaques are potent synaptotoxins, block proteasome function, inhibit mitochondrial activity, alter i ntracellular Ca 2+ levels and stimulate inflammatory processes. Loss of the normal physiological functions of ⁇ is also thought to contribute to neuronal dysfunction.
  • interacts with the signall i ng pathways that regulate the phosphorylation of the microtubule-associated protein tau.
  • Hyperphosphorylation of tau disrupts its normal function in regulati ng axonal transport and leads to the accumulation of neurofibri llary tangles (NFTs) and toxic species of soluble tau. Furthermore, degradation of hyperphosphorylated tau by the proteasome is inhibited by the actions of ⁇ . These two proteins and thei r associated signal ling pathways therefore represent important therapeutic targets for AD.
  • JNKs C-Jun N-terminal kinases
  • JNK1 , JNK2, and JNK3 are serine-threonine protein kinases, coded by three genes JNK1 , JNK2, and JNK3, expressed as ten different isoforms by mRNA alternative splicing, each isoforms being expressed as a short form (46 kDa) and a long form (54 kDa) (Davis, 2000, Cell 1 03: 239-52).
  • Whi le JNK1 and JNK2 are ubiquitous, JNK3 is mainly expressed in the brain (Kyriakis and Avruch, 2O01 , Physiol Rev 81 : 807-69).
  • JNKs are activated by phosphorylation (pJNK) through MAPKinase activation by extracellular stimuli, such as ultraviolet stress, cytokines and ⁇ peptides and they have multiple functions including gene expression regulation, cel l proliferation and apoptosis (Dhanasekaran and Reddy, 2008, Oncogene 27: 6245-51 ).
  • the JNK i nhibitors according to the present invention reduce tau hyperphosphorylation and, thus, neuronal loss. Therefore, the J NK i nhibitors according to the present invention can be useful for treating and/or preventing tauopathies.
  • Tauopathies are a class of neurodegenerative diseases associated with the pathological aggregation of tau protein i n the human brai n.
  • the best-known tauopathy is Alzheimer's disease (AD), wherein tau protein is deposited withi n neurons in the form of neurofibri llary tangles (NFTs), which are formed by hyperphosphorylation of tau protei n.
  • the degree of NFT involvement i n AD is defined by Braak stages.
  • Braak stages I and II are used when NFT involvement is confi ned mainly to the transentorhi nal region of the brain, stages II I and IV when there is also involvement of limbic regions such as the hippocampus, and V and VI when there is extensive neocortical i nvolvement. This should not be confused with the degree of senile plaque involvement, which progresses differently.
  • the JNK i nhibitors can be used accordi ng to the present invention for treati ng and/or preventing tauopathies, in particular Alzheimer's disease with NFT involvement, for example AD with Braak stage I, AD with Braak stage II, AD with Braak stage II I, AD with Braak stage IV and/or AD with Braak stage V. - -
  • NFTs neurofibrillary tangles
  • Further tauopathies i.e. conditions in which neurofibrillary tangles (NFTs) are commonly observed, and which can thus be treated and/or prevented by the JNK inhibitors according to the present invention, include progressive supranuclear palsy although with straight filament rather than PHF (paired helical filaments) tau; dementia pugilistica (chronic traumatic encephalopathy); frontotemporal dementia and parkinsonism linked to chromosome 1 7, however without detectable ⁇ -amyloid plaques; Lytico-Bodig disease (Parkinson-dementia complex of Guam); tangle- predominant dementia, with NFTs similar to AD, but without plaques; ganglioglioma and gangliocytoma; meningioangiomatosis; subacute sclerosing panencephalitis; and/or lead encephalopathy, tuberous sclerosis, Hallervorden-Spatz disease, and lipofus
  • tauopathies which can be treated and/or prevented by the JNK inhibitors according to the present invention, include Pick's disease; corticobasal degeneration; Argyrophilic grain disease (AGD); frontotemporal dementia and frontotemporal lobar degeneration.
  • ALD Argyrophilic grain disease
  • tau proteins are deposited in the form of inclusion bodies within swollen or "ballooned" neurons.
  • Argyrophilic grain disease (AGD), another type of dementia, which is sometimes considered as a type of Alzheimer disease and which may co-exist with other tauopathies such as progressive supranuclear palsy, corticobasal degeneration, and also Pick's disease, is marked by the presence of abundant argyrophilic grains and coi led bodies on microscopic examination of brain tissue.
  • the non-Alzheimer's tauopathies are sometimes grouped together as "Pick's complex".
  • the disorder/disease to be prevented and/or treated by the JNK inhibitor according to the present invention is Mild Cognitive Impairment (MCI), in particular MCI due to Alzheimer's Disease.
  • Mild Cognitive Impairment (MCI) is different from Alzheimer's Disease, i.e. Mi ld Cognitive Impairment (MCI) is typically not Alzheimer's Di sease, but is a disease on its own classified by ICD-1 0 in F06.7.
  • ICD-1 0 F06.7
  • MCI is described as a disorder characterized by impairment of memory, learning difficulties, and reduced ability to concentrate on a task for more than brief periods.
  • MCI Mild cognitive impairment
  • MCI The diagnosis of MCI is described for example by Albert MS, DeKosky ST, Dickson D, Dubois B, Feldman H H, Fox NC, Gamst A, Holtzman DM, Jagust WJ, Petersen RC, Snyder PJ, Carrillo MC, Thies B, Phelps CH (201 1 )
  • the diagnosis of mi ld cognitive impairment due to Alzheimer's disease recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guideli nes for Alzheimer's disease; Alzheimers Dement.;7(3):270-9.
  • MCI may be at the onset of whatever type of dementia or represents an ephemeric form of cognitive impairment which may disappear over time without resulting in a clinical manifestation of dementia.
  • a person with MCI is at an increased risk of developing Alzheimer's or another dementia, in particular at an increased risk of developing Alzheimer's Disease, without however necessari ly developing dementia, in particular Alzheimer's Disease.
  • No medications are currently approved by the U.S. Food and Drug Admi nistration (FDA) to treat Mild Cognitive Impairment. Drugs approved to treat symptoms of Alzheimer's Disease have not shown any lasting benefit in delaying or preventing progression of MCI to dementia.
  • the JNK inhibitors of the present invention may also be used for the treatment of diseases and/or disorders of the uri nary system i n particular selected from ureteritis; uri nary tract infection (bladder infection, acute cystitis); cystitis in general, i n particular i nterstitial cystitis, Hunner's ulcer, trigonitis and/or hemorrhagic cystitis; urethritis, in particular nongonococcal urethritis or gonococcal urethritis; pai nful bladder syndrome; IC/PBS; urethral syndrome; and/or retroperitoneal fibrosis, preferably cystitis in general, i n particular interstitial cystitis.
  • ureteritis uri nary tract infection (bladder infection, acute cystitis); cystitis in general, i n particular i nterstitial cystitis, Hunner's ulcer, trigonitis and/or hemorrhagi
  • IC i nterstitial cystitis
  • BPS blade pain syndrome
  • PBS pressureful bladder syndrome
  • IC/PBS includes all cases of urinary pain that can't be attributed to other causes, such as i nfection or urinary stones.
  • i nterstitial cystitis is typically used alone when describi ng cases that meet al l of the IC criteria, for example as established by the National I nstitute of Diabetes and Digestive and Kidney Diseases (NIDDK).
  • NIDDK National I nstitute of Diabetes and Digestive and Kidney Diseases
  • the JNK i nhibitors of the present invention may also be used for the treatment of metabolic disorders, for example for the treatment of diabetes (type 1 or type 2, in particular type 1 ), Fabry disease, Gaucher disease, hypothermia, hyperthermia hypoxia, l ipid histiocytosis, lipidoses, metachromatic leukodystrophy, mucopolysaccharidosis, Niemann-Pick disease, obesity, and Wolman's disease.
  • metabolic disorders may be of hereditary form or may be acquired disorders of - - carbohydrate metabol ism, e.g., glycogen storage disease, disorders of ami no acid metabolism, e.g., phenylketonuria, maple syrup urine disease, glutaric acidemia type 1 , Urea Cycle Disorder or Urea Cycle Defects, e.g., Carbamoyl phosphate synthetase I deficiency, disorders of organic acid metabolism (organic acidurias), e.g., alcaptonuria, disorders of fatty acid oxidation and m itochondrial metabolism, e.g., Medium-chain acyl-coenzyme A dehydrogenase deficiency (often shortened to MCADD.), disorders of porphyrin metabolism, e.g.
  • acute intermittent porphyria, di sorders of purine or pyrimidine metabol ism e.g., Lesch-Nyhan syndrome, Disorders of steroid metabolism, e.g., l ipoid congenital adrenal hyperplasia, or congenital adrenal hyperplasia, disorders of mitochondrial function, e.g., Kearns-Sayre syndrome, disorders of peroxisomal function, e.g., Zel lweger syndrome, or Lysosomal storage disorders, e.g., Gaucher's disease or Niemann Pick disease.
  • Lesch-Nyhan syndrome Disorders of steroid metabolism, e.g., l ipoid congenital adrenal hyperplasia, or congenital adrenal hyperplasia
  • disorders of mitochondrial function e.g., Kearns-Sayre syndrome
  • disorders of peroxisomal function e.g., Zel lweger syndrome
  • Lysosomal storage disorders
  • the J NK i nhibitors of the present invention may also be used for the treatment of neoplasms in particular cancer (malignant neoplasms) and/or tumor diseases, in particular selected from sol id tumors in general; hematologic tumors i n general, in particular leukemia, for example acute lymphocytic leukemia (L1 , L2, L3), acute lymphoid leukaemia (ALL), acute myelogenous leukemia (AML), chronic lymphocytic leukaemia (CLL), chronic myeloid leukaemia (CML), promyelocytic leukemia (M3), monocytic leukemia, myeloblasts leukemia (M1 ), myeloblasts leukemia (M2), megakaryoblastic leukemia (M7) and myelomonocytic leukemia (M4); myeloma, for example multiple myeloma; lymphomas, for example non-Hodgkin
  • bronchial carcinoma is certainly not only a proliferative disease but would also belong in the group of diseases of the respiratory system including lung diseases.
  • classification of individual diseases is not considered to be limiting or concluding but is considered to of exemplary nature only. It does not preclude that individual disease states recited in one class are factually also su itable examples for the application of the JNK inhibitors of the present invention as treatment in another class of disease states.
  • a person skilled in the art will readily be capable of assigning the different disease states and disorders to matching classifications.
  • the present invention contemplates the use of a JNK inhibitor as defined herein for the treatment and/or prevention of various diseases states and disorders.
  • the present invention does not contemplate to use the JNK inhibitors as defined herein for immunizing non- human animals, e.g. for the production of monoclonal antibodies. Such methods are herein not considered to be methods for treatment of the animal body by therapy.
  • the present invention provides a JNK inhibitor as defined herein for the (in vitro) treatment of a tissue or organ transplant prior to its transplantation.
  • the term "prior to its transplantation” comprises the time of isolation and the time of perfusion/transport.
  • the treatment of a tissue or organ transplant "prior to its transplantation” refers for example to treatment during the isolation and/or during perfusion and/or during transport.
  • a solution used for isolation of of a tissue or organ transplant as well as a solution used for perfusion, transport and/or otherwise treatment of a tissue or organ transplant can preferably contain the JNK inhibitor according to the invention.
  • CIT cold ischemia time
  • WIT warm ischemia time
  • CIT is the length of time that elapses between an organ being removed from the donor, in particular the time of perfusion/treatment of an organ by cold solutions, to its transplantation into the recipient.
  • WIT is in general a term used to describe ischemia of cells and tissues under normothermic conditions.
  • WIT refers to the length of time that elapses between a donor's death, in particular from the time of cross-clamping or of asystole in non-heart- beating donors, until cold perfusion is commenced.
  • WIT may also refer to ischemia during implantation, from removal of the organ from ice until reperfusion.
  • a transplant originating from a brain-dead donor is typically not subjected to WIT, but has 8- 12 hrs of CIT (time needed for transportation from the procurement hospital to the isolation lab), whereas a transplant from a non-heart beating donor is typically exposed to a longer WIT and also 8-12 hrs of CIT.
  • CIT is usually limited (typically 1 - 2 hrs, for example in islet autotransplantation in patients with chronic pancreatitis).
  • Ischemia is an inevitable event accompanying transplantation, for example kidney transplantation.
  • Ischemic changes start with brain death, which is associated with severe hemodynamic disturbances: increasing intracranial pressure results in bradycardia and decreased cardiac output; the Cushing reflex causes tachycardia and increased blood pressure; and after a short period of stabilization, systemic vascular resistance declines with hypotension leading to cardiac arrest.
  • Free radical-mediated injury releases proinflammatory cytokines and activates innate immunity. It has been suggested that all of these changes-the early innate response and the ischemic tissue damage play roles in the development of adaptive responses, which in turn may lead to transplant rejection.
  • ischemic tissue damage Hypothermic storage of the organ and/or tissue of various durations before transplantation add to ischemic tissue damage.
  • the final - - stage of ischemic injury occurs during reperfusion.
  • Reperfusion injury the effector phase of ischemic injury, develops hours or days after the initial insult. Repair and regeneration processes occur together with cellular apoptosis, autophagy, and necrosis; the fate of the organ depends on whether cell death or regeneration prevails.
  • the whole process has been described as the ischemia- reperfusion (l-R) injury. It has a profound influence on not only the early but also the late function of a transplanted organ or tissue. Prevention of l-R injury can thus already be started before organ recovery by donor pretreatment.
  • transplants may be (pre-)treated by the JNK inhibitors according to the present invention in order to improve their viability and functionality until transplanted to the host.
  • the transplant is in particular a kidney, heart, lung, pancreas, in particular pancreatic islets (also called islets of Langerhans), liver, blood cell, bone marrow, cornea, accidental severed limb, in particular fingers, hand, foot, face, nose, bone, cardiac valve, blood vessel or intestine transplant, preferably a kidney, heart, pancreas, in particular pancreatic islets (also called isl ets of Langerhans), or skin transplant.
  • the present invention provides a JNK inhibitor as defined herein for the treatment of a tissue or organ transplant, or an animal or human who received a tissue or organ transplant during or after transplantation.
  • the term "after transplantation” refers in particular to reperfusion of the organ or tissue, for example a kidney, whereby reperfusion begins for example by unclamping the respective blood flow.
  • the treatment with a JNK inhibitor according to the present invention after transplantation refers in particular to the time interval of up to four hours after reperfusion, preferably up to two hours after reperfusion, more preferably up to one hour after reperfusion and/or at the day(s) subsequent to transplantation.
  • the JNK inhibitor according to the present invention may be administered for example to an animal or human who received a tissue or organ transplant as pharmaceutical composition as described herein, for example systemically, in particular intravenously, in a dose in the range of 0.01 - 10 mg/kg, preferably in the range of 0.1 - 5 mg/kg, more preferably in the range of 0.5 - 2 mg/kg at a single dose or repeated doses.
  • the transplant is in particular a kidney, heart, lung, pancreas, in particular pancreatic islets (also called islets of Langerhans), liver, blood cell, bone marrow, cornea, accidental severed limb, in particular fingers, hand, foot, face, nose, bone, cardiac valve, blood vessel or intestine transplant, preferably a kidney, heart, pancreas, in particular pancreatic islets (also called islets of Langerhans), or skin transplant.
  • pancreatic islets also called islets of Langerhans
  • synthesis of the J NK inhibitor with SEQ ID NO: 1 72 is set out below.
  • a person ski l led in the art wi ll know that said synthesis may also be used for and easi ly adapted to the synthesis of any other JNK i nhibitor accordi ng to the present invention.
  • the JNK inhibitor with SEQ ID NO: 1 72 was manufactured by solid-phase peptide synthesis using the Fmoc (9-fluorenylmethyloxycarbonyl) strategy.
  • the l i nker between the peptide and the resin was the Rink amide l inker (p-[Fmoc-2,3-dimethoxybenzyl]-phenoxyacetic acid).
  • the peptide was synthesized by successive Fmoc deprotection and Fmoc-amino acid coupling cycles.
  • the completed peptide was cleaved by trifluoroacetic acid (TFA) directly to yield the - - crude C-terminal amide, which was then purified by preparative reverse phase HPLC.
  • the purified fractions were pooled in a homogeneous batch that is treated by ion exchange chromatography to obtain its acetate salt.
  • the peptide was then freeze-dried. 1 . 1 Solid Phase Synthesis of the Peptide
  • the p-methylbenzhydrylamide resin (MBHA-resin) was first washed with dichloromethane/ dimethylformamide/diisoproplyethylamine under nitrogen. The washed resin was then coupled to the Rink amide linker (p-[Fmox-2,4-dimethoxybenzyl]-phenoxyacetic acid) in PyBOB(benzotriazole-1 -yl-oxy-tris-pyrrolidino-phosphonium hexafluorophosphate)/ diisopropyl- ethylamine/1 -hydroxybenzotriazole to yield Fmoc-Rink amide-MBHA resin.
  • Rink amide linker p-[Fmox-2,4-dimethoxybenzyl]-phenoxyacetic acid
  • the Fmoc-Rink amide-MBHA resin was deprotected by washing it in 35% (v/v) piperidine/dimethylformamide, followed by dimethylformamide. The deprotection reaction took approximately 1 6 minutes.
  • Fmoc-protected amino acids e.g., 2 eq of amino acid and HOBt (1 - hydroxybenzotriazole) in dimethylformamide/dichloromethane (50/50) were added to the resin followed by addition of 2 eq of the coupling agent diisopropylcarbodiimide (DIC). The coupling reaction took from one hour to overnight depending upon the respective amino acid being added.
  • the peptide was cleaved from the resin in a mixture of trifluoroacetic acid/1 ,2-ethanedthiol/ thioanisole/water/phenol (88/2.2/4.4/4.4/7 v/v), also called TFA/K reagent, for 4 hours at room temperature.
  • the reaction volume was 1 mL/1 OOmg of peptide resi n.
  • the mixture temperature was regulated to stay below 30°C. 1 .5 Extraction of the peptide from the resin:
  • the crude peptide was then purified by reverse-phase HPLC to a purity of >95%.
  • the purified fractions were concentrated on a rotavaporator and freeze-dried.
  • the concentrated freeze-dried pools of purified peptide with the sequence of SEQ ID NO: 1 72 was dissolved in water and purified by ion exchange chromatography on Dowex acetate, 50-1 00 mesh resin.
  • Fmoc-Rink amide linker 1 45069-56-3 p-[Fmoc-2,4-dimethoxybenzyl]- 539.6 phenoxyacetic acid
  • JNK inhibitors of the present invention may be prepared in similar manner.
  • Example 2 Inhibitory efficacy of selected INK inhibitors according to the present invention
  • the method allows to measure in vitro, in a non radioactive standardized assay, the ability of a candidate compound to decrease the phosphorylation of the c-Jun specific substrate by JNK. Moreover, it will be illustrated how to determine the inhibitory effect (IC50) and the Ki of a chosen compound for JNK. The method is suitable to verify whether a candidate compound does or does not inhibit JNK activity. And a - - person skilled in the art will certainly understand how to adapt the below methods for his specific purposes and needs.
  • His-JNK1 (ref 14-327, Upstate, 10 pg in 100 ⁇ : concentration: 2.2 ⁇ ) 5nM final
  • AprotA beads (ref 676061 7M, PerkinElmer, lot 540-460-A, concentration 5mg/ml) 20 pg/ml final
  • Optiplate 384well white plate (ref 6007299, PerkinElmer, lot 654280/2008) 96well plate for peptide dilution (ref 82.1 581 , Sarstedt)
  • the bioluminescent energy transfer was read on the Fusion Alpha Plate reader (Perkin Elmer).
  • An electronic EDP3 pipette 20-300 (Ref 1 7007243; Rainin) was used to fill in the plate with the Enzme-Antibody mix, the Subtrate-ATP mix and the Beads.
  • a PIPETMAN® Ultra multichannel 8X20 (Ref 21040; Gilson) was used to fill in the plate with the inhibitory compounds. Buffer and solutions - -
  • Stop Buffer 20mM Tris-base pH 7.4, 200mM NaCl, 80mM EDTA-K (pH de 8 with KOH instead of NaOH), 0.3% BSA
  • the mixes were added with the pipette in different corner of the well. After the filling in of the plate with each mix, the plate was tapped (Keep one side fix and let the opposite side tap the table) to let the mix go down the walls of the wells.
  • the bioluminescent energy transfer was read on the Fusion Alpha Plate reader (Perkin Elmer).
  • Al I compounds should at least be tested in triplicate in 3 independent experiments for each isoform of JNK. Possibly concentrations of the compounds to be tested were 0, 0.03 nM, 0.1 nM, 0.3 nM, 1 nM, 3 nM, 10 nM, 30 nM, 1 00 nM, 300 nM, 1 ⁇ , 3 ⁇ , 1 0 ⁇ , 30 ⁇ , and 1 00 ⁇ . Controls were samples either without JNK or without substrate (c-Jun).
  • Antibody [final] 10nM (anti Phospho cjun (S63))
  • Ki IC50 / (1 + ([Substrate] / Km of the substrate)) may be used to calculate the Ki values.
  • the cell line used for this experiment was HL-60 (Ref CCL-240, ATCC, Lot 1 1 6523) b) Culture medium and plates
  • PBS 1 0X (Ref 7001 1 , Invitrogen, Lot 8277): diluted to 1 X with sterile H 2 O - -
  • Trvpsine-0.05% EDTA (Ref L-1 1 660, PAA, Lot L66007-1 1 94)
  • Poly-D-lysine coating solution (Sigma P901 1 Lot 095K5104): final diluted in PBS 1 x
  • Acidic wash buffer 0.2M Glycin, 0.15M NaCi, pH 3.0
  • Ripa lysis buffer 10mM NaH 2 P0 4 pH 7.2, 1 50mM NaCi, 1 % Triton X-100, 1 mM EDTA pH 8.0, 200 ⁇ Na 3 V0 2 , 0.1 % SDS, 1 X protease inhibitor cocktail (Ref 1 1 873580001 , Roche, Lot 13732700) d) Microscopy and fluorescence plate reader
  • FITC marked peptide internalization was studied on suspension cells.
  • Cells were plated into poly-DL-lysine coated dishes at a concentration of 1 x 10 6 cells/ml. Plates were then incubated for 24 h at 37 °C, 5 % C0 2 and 100% relative humidity prior to the addition of a known concentration of peptide. After peptide addition, the cells were incubated 30 mi n, 1 , 6 or 24 h at 37 °C, 5 % C0 2 and 100 % relative humidity.
  • the acid wash was carried out in determining cellular uptake of Fab/cell-permeating peptide conjugates, followed by two PBS washes. Cells were broken by the addition of the RIPA lysis buffer. The relative amount of internalized peptide was then determined by fluorescence after background subtraction and protein content normalization.
  • the 6 well culture plates are coated with 3 ml of Poly-D-Lys (Sigma P901 1 ; 25 pg/ml in PBS), the 24 well plates with 600 ⁇ and the 96 well plates with 125 ⁇ and incubated for 4 h at 37°C, C0 2 5 % and 1 00 % relative humidity.
  • Poly-D-Lys Sigma P901 1 ; 25 pg/ml in PBS
  • the cells were plated into the dishes in 2.4 ml medium (RPMI) at plating densities of 1 ⁇ 00 ⁇ 00 cells/ml for suspension cells. After inoculation, the plates were incubated at 37°C, 5 % CO2 and 100 % relative humidity for 24 hours prior to the addition of the peptide.
  • Adherent cells should be at a density of 90-95% the day of treatment and were plated in DMEM :
  • the cells were treated with the desired concentration of FITC labeled peptide (stock solution at a concentration of 1 0 mM in H 2 0).
  • the cells were incubated 0 to 24 hours (e.g. 30 min, 1 , 6 or 24 hours) at 37 °C, CO2 5 % and 100 % relative humidity.
  • Suspension cells (or cells, which don attach well to the dish):
  • the lysed cells were then centrifuged 30 min at 1 0000 g at 4 °C to remove the cellular debris. Remove the supernatant and store it at -80 °C in a coated "Eppendorf tube" (coated with 1 ml of poly D-Lys for 4 hours and washed twice with 1 ml PBS).
  • each protein extract was determined by a standard BCA assay (Kit N°23225, Pierce), following the instructions of the manufacturer.
  • the relative fluorescence of each sample is determined after reading 10 ⁇ of each sample in a fluorescence plate reader (Fusion Alpha, Perkin Elmer), background subtraction and normalization by protein concentration.
  • the time dependent internalization (uptake) of FITC-labeled TAT derived transporter constructs into cells of the HL-60 cell line was carried out as described above using sequences transporter peptides of SEQ ID NOs: 52-96, 43, and 45-47. These sequences are listed below in Table 4.
  • TAT derived sequences as shown in Table 4 are preferred, which exhibit an Y in position 2, particularly when the sequence exhibits 9 aa and has the consensus sequence rXXXrXXXr (SEQ ID NO: 31 ).
  • Sandwich ELISA allows measuring the amount of antigen between two layers of antibodies (i.e. capture and detection antibody).
  • the antigen to be measured must contain at least two antigenic sites capable of binding to antibody, since at least two antibodies act in the sandwich.
  • Either monoclonal or polyclonal antibodies can be used as the capture and detection antibodies in Sandwich ELISA systems.
  • Monoclonal antibodies recognize a single epitope that allows fine detection and quantification of small differences in antigen.
  • a polyclonal is often used as the capture antibody to pull down as much of the antigen as possible.
  • Sandwich ELISA is that the sample does not have to be purified before analysis, and the assay can be very sensitive (up to 2 to 5 times more sensitive than direct or indirect).
  • the method may be used to determine the effect of the JNK inhibitors of the present invention in vitro/ cell culture.
  • compound efficacy is indicated by the decrease of the cytokine levels (the variation of optical density (absorbance at 450 nm)) as compared to non-treated samples and is monitored by ELISA. Results are express in ng/ml.
  • TopSeal-A 96well microplate seals (Ref 600585, PerkinElmer).
  • Wash buffer ELISA PBS 1 X + 0.01 % Tween20. Prepare 1 litre PBS 1 X (PBS10X: ref 7001 1 , GIBCO) and add l OOul of Tween20 (ref P1 379, Sigma) slowly while mixing with magnetic agitator)
  • IFN-y Human IFN- ⁇ ELISA set, BD OptEIATM (ref 555142, DB).
  • IL- ⁇ ⁇ Human IL-1 ⁇ ELISA set II, BD OptEIATM (ref 557953, BD)
  • IL-1 0 Human IL-1 0 ELISA set II, BD OptEIATM (ref 5551 57, DB).
  • IL-12 Human IL-12 (p70) ELISA set, BD OptEIATM (ref 5551 83, DB).
  • IL-1 5 Human IL-1 5 ELISA Set, BD OptEIATM (ref 559268, DB).
  • IL-2 Human IL-2 ELISA set, BD OptEIATM (ref 5551 90, DB).
  • IL-4 Human IL-4 ELISA set, BD OptEIATM (ref 5551 94, DB).
  • IL-5 Human IL-5 ELISA set, BD OptEIATM (ref 555202, DB).
  • IL-6 Human IL-6 ELISA setl, BD OptEIATM (ref 555220, DB).
  • IL-8 Human IL-8 ELISA set, BD OptEIATM (ref 555244, DB).
  • MCP-1 Human MCP-1 ELISA set
  • BD OptEIATM (ref 5551 79, BD)
  • TNF-a Kit human TNF ELISA set
  • BD OptEIATM (ref 555212, DB).
  • the samples are culture medium supernatant from cultured human cells (typically whole blood, WBC, PBMC, Purified subtype of WBC, cancerous cell lines). Remove any particulate material by centrifugation (400g 5min 4°C) and assay immediately or store samples at -20°C. Avoid repeated freeze-thaw cycles. - -
  • step 1 1 dilute the samples in assay diluent directly into the plate (add first assay diluent, then the samples and pipette up and down):
  • step 3 Do one wash as in step 3 with 1 50 ⁇ of wash buffer. The plates are now ready for sample addition.
  • detector MIX detection antibody +Secondary Streptavidin-HRP antibody in assay diluent
  • the data are presented in pg/ml of cytokine release or in %, compared to the induced condition without inhibitor treatment.
  • Example 5 THP1 differentiation - stimulation for cytokine release
  • cytokine production from human PMA differentiated THP1 cells challenged by LPS for 6h was induced in order to test the ability of JNK inhibitors of the present invention, in particular of a JNK inhibitor with SEQ ID NO: 1 72, to reduce stimulation-induced cytokine release.
  • THP1 cells were stimulated ex-vivo by different ligands for the readout of cytokine release.
  • JNK inhibitor efficacy is indicated by the decrease of the cytokine levels as compared to non-treated samples and is monitored by ELISA.
  • the toxicity of the compound are evaluated by the reduction of a tretazolium salt (MTS) to formazan, giving a purple colour.
  • MTS tretazolium salt
  • Penicilline (1 OOunit/ml) / Streptomycine (100 g/ml) (Ref P4333, Sigma)
  • the RPMI medium is then filtrated with a 0.22 M fi lter (Ref SCGPU05RE, Millipore).
  • PBS 1 0X (Ref 7001 1 , Invitrogen): diluted to 1 X with sterile H 2 O
  • LPS ultrapure Lipopolysaccharide, Ref tlrl-eklps, Invivogen, concentration 5mg/ml
  • Stock solution of LPS 3 g/ml in PBS at 4°C.
  • Use directly to prepare a 4X concentrated solution of 40ng/ml in RPMl medium (min 1 800 ⁇ /plate; for 5 plates: 125 ⁇ of LPS 3 g/ml + 9250 ⁇ RPMl).
  • TNF- Kit human TNF ELISA set, BD OptEIA (ref 555212, DB).
  • Control compound SP600125 (ref ALX-270-339-M025, Alexis, concentration: 20mM DMSO)
  • TopSeal-A 96well microplate seals (Ref 600585, PerkinElmer). b. Method - -
  • the plates had been coated with 200 ⁇ of poly D-Lysine (1 x) and incubated 2 hours at 37°C, C0 2 5% and 100% relative humidity. Cell plating
  • the cells were counted. The desired number of cells was taken and resuspended in the amount of media necessary to get a dilution of 1 ⁇ 00 ⁇ 00 cells/ml. 100nM of PMA was added to induce the differentiation of the THP1 from suspension monocytes to adherent macrophages. The cells were plated into the wells in 100 I medium at plating densities of 100'OOOcel Is/well. After inoculation, the plates were incubated at 37°C, 5% C02 and 1 00% relative humidity 3 days to let them differentiate, prior to the addition of experimental drugs.
  • Experimental drug were prepared at the concentration of 1 0 mM in H 2 0 or DMSO and stored at - 80°C. Prior to each daily use, one aliquot of JNK inhibitor was defrost and diluted to reach a 4X concentrated solution (120 M) in RPMI medium and then to the desired concentration in RPMI. The SP600125 was diluted to reach a 4X concentrated solution (40 M) in RPMI medium and then to the desired concentration in RPMI containing 0.8% DMSO.
  • the plates were treated with 50 ⁇ of medium or a solution of 4X the final desired drug concentration (0, 10OnM, 1 , 3, 10 or 30 M final for JNK compound or at 0, 10, 1 0OnM, 1 , 3 or 10 M final for the SP600125 positive control). Following drug addition, the plates were incubated for an additional 1 h at 37°C, 5% C0 2 and 100% relative humidity.
  • cytotoxic effect of the compounds was evaluated by MTS absorbance (e.g. see example 4) and cells were observed using an inverted microscope (Axiovert 40 CFL; Zeiss; 10X).
  • Analyses of the data are performed as indicated in the ELISA (see example 4). Briefly, for ELISA: Average the triplicate readings for each standard control and each sample. Subtract the average zero standard optical density (O.D). Create a standard curve plotting the log of the cytokine concentration versus the log of the O.D and the best fit line can be determined by regression analysis. If samples have been diluted, the concentration read from the standard curve must be multiplied by the dilution factor. A standard curve should be generated for each set of samples assayed. The outliers data were avoid using Grugg's test. Then the data which weret in the interval of two times the SD, were discard. The independent experiments are taken into account if the positive control showed data as previously observed.
  • O.D optical density
  • the independent experiments are pooled (N > 3).
  • the average of the absorbance of the medium alone was considerate as the background and subtracted to each absorbance value.
  • the average of triplicate of the non treated cells of each compound was considerate as the 1 00% viability.
  • the average of triplicate of each compound was normalized by its 100%. The outliers data were avoid using Grugg's test. Then the data which did't in the interval of two times the SD, were discard.
  • the independent experiments are pooled (N > 3).
  • Example 6 INK inhibitor of SEP ID NO: 1 72 and TNFa release in Primary Rat or human whole blood cells
  • Whole blood is collected from anesthetized rat or human healthy volunteers using a venipuncture connected to a pre-labeled vacuum tube containing sodium citrate. Tubes are gently mixed by inversion 7-8 times; and are then kept at RT unti l stimulation. JNK inhibitor of SEQ ID NO: 1 72_is prepared 6 times concentrated in PBS, and 30 ⁇ /well of mix is added into 96-well plate. Whole blood is diluted by 1 :2 in PBS and 120 ⁇ of diluted blood is added in each well where either PBS alone or JNK inhibitor of SEQ ID NO: 1 72 has been previously added. Whole blood is incubated at - -
  • Activators are the prepared, 30pl/well of LPS, 6 times concentrated. After 60min incubation, LPS is added to the blood, blood is mixed by p ipetting up and down, and then kept for 4h under agitation (85rpm), at 37°C. After the 4h incubation, the plates are centrifuged at about 770g, 4°c for 1 5 min in a pre-cooled centrifuge. Supernatants are finally collected and kept at -20°c until cytokine measurement. Cytokine (IL-6, IL- 2, IFNy and TNFa) were then measured using standard Elisa kits (e.g.
  • Results are expressed as pg/ml of supernatant of the measured cytokine.
  • PMA+ionomycin instead of LPS as activator/stimulant.
  • Example 7 Half-life of specific INK inhibitors disclosed herein
  • the JNK inhibitors with the sequence of SEQ ID NOs: 196, 197, and 1 72 (0.1 mM final concentration) were digested in human serum (10 and 50% in PBS 1 x). The experiment was performed as described by Tugyi et al. (Proc Natl Acad Sci U S A, 2005, 413-41 8). . The remaining intact peptide was quantified by UPLC-MS. Stability was assessed for SEQ ID NOs: 196, 1 97, and 1 72 identically but in two separate assays.
  • JNK inhibitor with SEQ ID NO: 1 96 was totally degraded into amino acids residues within 6 hours, the JNK inhibitor with SEQ ID NO: 1 72 was completely degraded only after 14 days.
  • the JNK inhibitor with SEQ ID NO: 1 97 was still stable after 30 days.
  • Example 8 Dose-dependent inhibition by INK inhibitor with sequence of SEQ ID NO: 1 72 of
  • lymph nodes were harvested and kept in complete RPMI medium. LN were smashed with complete RPMI on 70pm filter using a 5ml piston. A few drops of media were added to keep strainer wet. Cells were centrifuged for 7 min at 450g and 4°c. Pellet was resuspended in 5 ml fresh medium. Cells were passed again through cell strainer. An aliquot of cells was counted, while cells were centrifuged again 1 0min at 1400 rpm and 4°c. Cells were resupended in MACS buffer (80 ⁇ of MACS buffer per 1 0 7 cells).
  • Example 9 INK inhibitor and TNFa/IL-2 release in human whole blood:
  • CD3/CD8 stimulation CD3 antibody was coated at 2 ⁇ g/mL in PBS overnight at 4°C. The day of experiment, wells were washed three times with PBS and left in PBS until use at 37°C. CD28 antibody was added 1 h after SEQ ID NO: 1 72 at final concentration of 2pg/mL; supernatants were collected after 3 days of stimulation.
  • Example 10 Anti-Inflammatory potency in a rat model of endotoxins induced uveitis (EIU)
  • the anti-inflammatory potency of the JNK inhibitor of SEQ ID NO: 1 72 was tested in albino rats following intravenous administration (EIU/LPS model).
  • the aim of this study was to determine the effects of single intravenous injections of SEQ ID NO: 1 72 (0.01 5, 0.1 8, and 1 .80 mg/kg) on the inflammatory response in an endotoxins-induced uveitis albino rat model and to compare these affects to those obtained with prior art JNK inhibitor of SEQ ID NO: 1 97 (2 mg/kg).
  • As a further control served phosphate sodic dexamethasone ("reference") and vehicle (0.9 % NaCl).
  • EI U was induced by footpad injection of lipopolysaccharide (LPS, 1 mg/kg). 24 hours after LPS injection, inflammatory response was evaluated by clinical scoring. The intensity of clinical ocular inflammation was scored on a scale from 0 to 4 for each eye:
  • the lower doses (0.1 8 and 0.01 5 mg/kg) reduced by 33% (mean score: 2.4 ⁇ 0.3, median: 2) and 36% (mean score: 2.3 + 0.3, median: 2) the inflammation, respectively.
  • the reduction was significant with p ⁇ 0.001 .
  • Example 1 1 Dose-responsive effects after intravenous administration of INK inhibitor after 14 days in a rat model of chronic established type II collagen arthritis Rat collagen arthritis is an experimental model of polyarthritis that has been widely used for preclinical testing of numerous anti-arthritic agents that are either under preclinical or clinical investigation or are currently used as therapeutics in this disease. The hallmarks of this model are rel iable onset and progression of robust, easily measurable polyarticular inflammation, marked cartilage destruction in association with pannus formation, and mild to moderate bone resorption and periosteal bone proliferation.
  • Mild mild loss of toluidine blue staining with focal mild (superficial) chondrocyte loss and/or collagen disruption
  • Moderate moderate loss of toluidine blue staining with multifocal moderate (depth to middle zone) chondrocyte loss and/or collagen disruption, smaller tarsals affected to 1/2 to 3/4 depth with rare areas of full thickness loss
  • Marked marked loss of toluidine blue staining with multifocal marked (depth to deep zone) chondrocyte loss and/or collagen disruption, 1 or 2 small tarsals surfaces have full thickness loss of cartilage
  • Severe severe diffuse loss of toluidine blue staining with multifocal severe (depth to tide mark) chondrocyte loss and/or collagen disruption affecting more than 2 cartilage surfaces
  • Mild mild loss of toluidine blue staining with focal mild (superficial) chondrocyte loss and/or collagen disruption, may have few small areas of 50% depth of cartilage affected
  • Moderate moderate loss of toluidine blue staining with multifocal to diffuse moderate (depth to middle zone) chondrocyte loss and/or collagen disruption, may have 1 -2 small areas of full thickness loss affecting less than 1 /4 of the total width of a surface and not more than 25% of the total width of all surfaces
  • Marked marked loss of toluidine blue staining with multifocal to diffuse marked (depth to deep zone) chondrocyte loss and/or collagen disruption or 1 surface with near total loss and partial loss on others, total overall loss less than 50% of width of all surfaces combined
  • Severe severe diffuse loss of toluidine blue staining with multifocal severe (depth to tide mark) chondrocyte loss and/or collagen disruption on both femurs and/or tibias, total overall loss greater than 50% of width of all surfaces combined
  • Moderate obvious resorption of medullary trabecular and cortical bone without full thickness defects in cortex, loss of some medullary trabeculae, lesion apparent on low magnification, osteoclasts more numerous, 1 /4 to 1 /3 of tibia or tarsals affected at marginal zones
  • D isease severity in the disease control group increased from days 1 to 5 with day 4-5 having the greatest daily increase. Then the incremental increases were smaller to the peak at day 7. From that point forward, acute swelling generally decreased and calliper measures were decreased.
  • the treatment groups followed this general pattern as well.
  • Body weight loss was observed in all disease groups whereas the normal control group had a weight increase. Body weight loss was significantly (25%, p ⁇ 0.05 by ANOVA) inhibited for rats treated with 5 mg/kg SEQ ID NO: 1 72 as compared to vehicle treated disease controls. When compared to disease controls using a Student's t-test, inhibition of body weight loss was also significant for rats treated with 1 mg/kg SEQ ID NO: 1 72 (21 %, p ⁇ 0.05) or Dex (21 %, p ⁇ 0.05). Results of treatment with SEQ ID NO: 1 72 were dose responsive for this parameter.
  • Dai ly ankle diameter measurements were significantly (p ⁇ 0.05 by 2-way RM ANOVA) reduced toward normal for rats treated with 5 mg/kg SEQ ID NO: 1 72 (p ⁇ 0.05 days 4-12) or Dex (p ⁇ 0.05 d3-14) as compared to disease controls.
  • Ankle diameter AUC was significantly (p ⁇ 0.05 by ANOVA) reduced toward normal for rats treated with 5 mg/kg SEQ ID NO: 1 72 (43% reduction), 1 mg/kg SEQ ID NO: 1 72 (27%), or Dex (97%) as compared to disease controls. Results of treatment with SEQ ID NO: 1 72 were dose responsive for this parameter. - -
  • Relative liver weights were not significantly (by ANOVA) affected for rats in any treatment group as compared to disease controls.
  • Spleen weights relative to body weight were significantly (p ⁇ 0.05 by ANOVA) reduced for rats treated with Dex as compared to disease controls. Relative spleen weights for Dex treated rats were also significantly reduced as compared to normal controls. Relative spleen weights were not significantly affected for rats treated with SEQ ID NO: 1 72.
  • Thymus weights relative to body weight were significantly (p ⁇ 0.05 by ANOVA) reduced for rats treated with Dex as compared to disease controls. Relative thymus weights for Dex treated rats were also significantly reduced as compared to normal controls. Relative thymus weights were not significantly affected for rats treated with SEQ ID NO: 1 72.
  • Al l ankle histopathology parameters were significantly (by Mann-Whitney U test) reduced toward normal for rats treated with 5 mg/kg SEQ ID NO: 1 72 (25% reduction of summed scores) as compared to disease controls.
  • Al l knee histopathology parameters were significantly (by Mann-Whitney U test) reduced toward normal for rats treated with 5 mg/kg SEQ ID NO: 1 72 (73% reduction of summed scores) as compared to disease controls.
  • Results of treatment with SEQ ID NO: 1 72 were dose responsive.
  • Example 12 Effect of the all-D-retro-inverso IN -inhibitor (poly-)peptide of SEQ ID NO: 197 and the INK inhibitor (poly-)peptide of SEQ ID NO: 1 72 at three doses in a Scopolamine- Induced Model of Dry Eye in Mice Study concept
  • the objective of this study was to assess the effects of two different compounds, the all-D-retro- inverso JNK-inhibitor (poly-)peptide of SEQ ID NO: 1 97 and the JNK inhibitor (poly-)peptide of SEQ ID NO: 1 72, at three dose levels in a mouse model of scopolamine-induced dry eye.
  • the peptides of SEQ ID NO: 1 97 and SEQ ID NO: 1 72 were tested for efficacy in this murine model of dry eye.
  • the peptides were both tested at a low, medium and a high dose.
  • the concentrations measured in the formulation samples for low, medium and high dose levels were 0.06% (w/v), 0.25% (w/v) and 0.6% (w/v), respectively, and for SEQ ID NO: 1 72 the concentrations measured in the formulation samples for the low, medium and high dose levels, were 0.05% (w/v), 0.2% (w/v) and 0.6% (w/v), respectively.
  • the study consisted of a total of 9 groups of female C57BL/6 mice, comprising 8 groups of 12 mice each and an additional group of 4 mice.
  • Bilateral short-term dry eye was induced by a combination of scopolamine hydrobromide (Sigma-Aldrich Corp., St. Louis, MO) injection (subcutaneous (SC), four times daily, 0.5 mg/dose, Days 0-21 ) and by exposing mice to the drying environment of constant air draft.
  • scopolamine hydrobromide Sigma-Aldrich Corp., St. Louis, MO
  • SC subcutaneous
  • mice of Groups 1 -8 were treated three times daily (TID) for 21 days with bilateral topical ocular (oculus uterque; OU) administration (5 ⁇ /eye/dose) of vehicle (0.9% sterile saline; negative control article); the peptide of SEQ ID NO: 1 97 (0.06%, 0.25% and 0.6%), the peptide of SEQ ID NO: 1 72 (0.05%, 0.2% and 0.6%); or cyclosporine (0.05%; positive control, an immunosuppressant drug used to reduce the activity of the immune system).
  • Mice of Group 9 were maintained as un-induced, (no dry eye) untreated controls.
  • Tissues from the right eyes were fixed and then evaluated microscopically.
  • Tissues from the left eyes were flash-frozen in liquid nitrogen and stored frozen at -80 °C for possible subsequent analyses. - -
  • the (poly-)peptide of SEQ ID NO: 1 97 was obtained from Polypeptide Laboratories (France) as a 1 .5-mL clear plastic microfuge vial containing 300.65 mg of dry powder.
  • the (poly-)peptide of SEQ ID NO: 1 72 was obtained from Polypeptide Laboratories (France) as a 1 .5-mL clear plastic microfuge vial containing 302.7 mg of dry powder.
  • the (poly-)peptides of SEQ ID NO: 1 72 and of SEQ ID NO: 1 97 were formulated in sterile saline (vehicle). Dosing solutions at each concentration were sterilized using 0.2-pm filters, aliquoted to multiple pre-labeled vials, and frozen at -20 °C. The concentrations measured in the formulation samples for the peptide of SEQ ID NO: 1 97 were 0.058%, 0.25% and 0.624%, rounded to 0.06%, 0.25% and 0.6%. The concentrations measured in the formulation samples for the peptide of SEQ ID NO: 1 72 were 0.053%, 0.21 7% and 0.562%, rounded to 0.05, 0.2% and 0.6%.
  • each animal Prior to entry into the study, each animal underwent a SLE and i ndirect ophthalmic exami nation usi ng topical ly-appl ied fluorescei n. Ocular fi ndi ngs were recorded using the Draize scale ocular scoring. SLE and Draize scoring were repeated three times a week duri ng the in-life period.
  • the TBUT test was conducted three times weekly by measuring the time elapsed in seconds between a complete bli nk after application of fluorescei n to the cornea and the appearance of the first random dry spot i n the tear fi lm.
  • 0.1 % liquid sodium fluorescei n was dropped into the conjunctival sac, the eyelids were manual ly closed three times and then held open reveali ng a continuous fluorescein-contai ni ng tear film covering the cornea, and the time (in seconds) required for the film to break (appearance of a dry spot or streak) was recorded.
  • corneal epithelial damage was graded using a sl it-lamp with a cobalt blue fi lter after another drop of 0.1 % fluorescein was reapplied to the cornea; the cornea then was scored per the Draize ocular scale.
  • Tear production was measured three times a week in both eyes using PRTT test strips (Zone-Quick; Menicon, Nagoya, Japan). Prior to the first treatment of the day, a thread was applied to the lateral canthus of the conjunctival fornix of each eye for 30 seconds under slit-lamp biomicroscopy. Tear m igration up the tread (i .e., the length of the wetted cotton thread) was measured using a mi l l imeter scale. 5.
  • both eyes from each animal including the globes, lacrimal glands, eyelids, and conjunctivae, were excised.
  • the right eye and associated tissues were fixed by overnight submersion in modified Davidson's solution followed by transfer to 1 0% neutral buffered formalin (N BF).
  • the fixed tissues of the right eye were dehydrated, embedded in paraffin, sectioned at 3 to 5- ⁇ thicknesses, and slide-mounted tissues were stained with hematoxyl i n and eosin (H & E). Stained sl i des were evaluated via light microscopy.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne l'utilisation de nouvelles molécules inhibitrices de JNK et leur utilisation dans une méthode de traitement thérapeutique du corps d'un humain ou d'un animal.
PCT/EP2015/001293 2013-06-26 2015-06-26 Nouvelle utilisation de molécules inhibitrices de jnk pour le traitement de diverses maladies WO2015197193A2 (fr)

Priority Applications (5)

Application Number Priority Date Filing Date Title
JP2016575150A JP6843625B2 (ja) 2013-06-26 2015-06-26 様々な疾患の処置のためのjnk阻害剤分子の新規使用
EP15734295.7A EP3160989A2 (fr) 2014-06-26 2015-06-26 Nouvelle utilisation de molécules inhibitrices de jnk pour le traitement de diverses maladies
US15/321,904 US20170128516A1 (en) 2013-06-26 2015-06-26 Use for JNK Inhibitor Molecules for Treatment of Various Diseases
US16/430,697 US11331364B2 (en) 2014-06-26 2019-06-04 Use for JNK inhibitor molecules for treatment of various diseases
US17/719,068 US20220280591A1 (en) 2013-06-26 2022-04-12 Use For JNK Inhibitor Molecules For Treatment Of Various Diseases

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
PCT/EP2014/001737 WO2014206564A1 (fr) 2013-06-26 2014-06-26 Nouvelle utilisation pour des molécules inhibitrices de la jnk, pour le traitement de diverses maladies
EPPCT/EP2014/001737 2014-06-26
PCT/EP2014/002723 WO2015197097A1 (fr) 2014-06-26 2014-10-08 Nouvelle utilisation pour des molécules inhibitrices de la jnk, pour le traitement de diverses maladies
EPPCT/EP2014/002723 2014-10-08

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/321,904 A-371-Of-International US20170128516A1 (en) 2013-06-26 2015-06-26 Use for JNK Inhibitor Molecules for Treatment of Various Diseases
US16/430,697 Continuation US11331364B2 (en) 2013-06-26 2019-06-04 Use for JNK inhibitor molecules for treatment of various diseases

Publications (2)

Publication Number Publication Date
WO2015197193A2 true WO2015197193A2 (fr) 2015-12-30
WO2015197193A3 WO2015197193A3 (fr) 2016-02-25

Family

ID=54938878

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2015/001293 WO2015197193A2 (fr) 2013-06-26 2015-06-26 Nouvelle utilisation de molécules inhibitrices de jnk pour le traitement de diverses maladies

Country Status (2)

Country Link
EP (1) EP3160989A2 (fr)
WO (1) WO2015197193A2 (fr)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016207413A1 (fr) * 2015-06-26 2016-12-29 Xigen Inflammation Ltd. Nouvelle utilisation d'inhibiteurs peptidiques à perméabilité cellulaire de la voie de transduction du signal jnk pour le traitement de troubles cognitifs légers
US9624267B2 (en) 2010-06-21 2017-04-18 Xigen Inflammation Ltd. JNK inhibitor molecules
US10023615B2 (en) 2008-12-22 2018-07-17 Xigen Inflammation Ltd. Efficient transport into white blood cells
CN109303782A (zh) * 2018-10-24 2019-02-05 厦门大学 Jnk-in-8在制备干性年龄相关性黄斑变性的神经保护剂中的应用
CN109862903A (zh) * 2016-04-28 2019-06-07 新加坡国立大学 治疗性sall4肽
US10596223B2 (en) 2011-12-21 2020-03-24 Xigen Inflammation Ltd. JNK inhibitor molecules for treatment of various diseases
US10624948B2 (en) 2013-06-26 2020-04-21 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of various diseases
CN112203674A (zh) * 2018-03-29 2021-01-08 S.I.S舒洛夫创新科学有限公司 用于抑制炎性细胞因子的药物组合物
US10967038B2 (en) 2010-10-14 2021-04-06 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of chronic or non-chronic inflammatory eye diseases
RU2764770C1 (ru) * 2017-12-28 2022-01-21 Хайсенсбайо Новый пептид
US11331364B2 (en) 2014-06-26 2022-05-17 Xigen Inflammation Ltd. Use for JNK inhibitor molecules for treatment of various diseases
US11779628B2 (en) 2013-06-26 2023-10-10 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of various diseases

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007031098A1 (fr) * 2005-09-12 2007-03-22 Xigen S.A. Inhibiteurs peptidiques permeables aux cellules de la voie de transduction de signal jnk
WO2009143865A1 (fr) * 2008-05-30 2009-12-03 Xigen S.A. Utilisation d'inhibiteurs peptidiques des voies de traduction du signal jnk perméables aux cellules pour le traitement de diverses maladies
WO2011160653A1 (fr) * 2010-06-21 2011-12-29 Xigen S.A. Nouvelles molécules inhibant jnk
WO2013091670A1 (fr) * 2011-12-21 2013-06-27 Xigen S.A. Nouvelles molécules inhibitrices de jnk pour le traitement de diverses maladies

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10023615B2 (en) 2008-12-22 2018-07-17 Xigen Inflammation Ltd. Efficient transport into white blood cells
US9624267B2 (en) 2010-06-21 2017-04-18 Xigen Inflammation Ltd. JNK inhibitor molecules
US10967038B2 (en) 2010-10-14 2021-04-06 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of chronic or non-chronic inflammatory eye diseases
US10596223B2 (en) 2011-12-21 2020-03-24 Xigen Inflammation Ltd. JNK inhibitor molecules for treatment of various diseases
US10624948B2 (en) 2013-06-26 2020-04-21 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of various diseases
US11779628B2 (en) 2013-06-26 2023-10-10 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of various diseases
US11331364B2 (en) 2014-06-26 2022-05-17 Xigen Inflammation Ltd. Use for JNK inhibitor molecules for treatment of various diseases
WO2016207413A1 (fr) * 2015-06-26 2016-12-29 Xigen Inflammation Ltd. Nouvelle utilisation d'inhibiteurs peptidiques à perméabilité cellulaire de la voie de transduction du signal jnk pour le traitement de troubles cognitifs légers
CN109862903A (zh) * 2016-04-28 2019-06-07 新加坡国立大学 治疗性sall4肽
RU2764770C1 (ru) * 2017-12-28 2022-01-21 Хайсенсбайо Новый пептид
CN112203674A (zh) * 2018-03-29 2021-01-08 S.I.S舒洛夫创新科学有限公司 用于抑制炎性细胞因子的药物组合物
CN109303782A (zh) * 2018-10-24 2019-02-05 厦门大学 Jnk-in-8在制备干性年龄相关性黄斑变性的神经保护剂中的应用

Also Published As

Publication number Publication date
WO2015197193A3 (fr) 2016-02-25
EP3160989A2 (fr) 2017-05-03

Similar Documents

Publication Publication Date Title
US20220280591A1 (en) Use For JNK Inhibitor Molecules For Treatment Of Various Diseases
EP3160989A2 (fr) Nouvelle utilisation de molécules inhibitrices de jnk pour le traitement de diverses maladies
US20200206301A1 (en) JNK Inhibitor Molecules For Treatment Of Various Diseases
US10624948B2 (en) Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of various diseases
US11779628B2 (en) Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of various diseases
JP7165693B2 (ja) 様々な疾患の処置のためのjnkシグナル伝達経路の細胞透過性ペプチド阻害剤の新規使用
US11331364B2 (en) Use for JNK inhibitor molecules for treatment of various diseases
WO2015197098A1 (fr) Nouvelle utilisation d'inhibiteurs peptidiques perméables aux cellules de la voie de transduction du signal jnk pour le traitement de diverses maladies
WO2016055160A2 (fr) Nouvelle utilisation d'inhibiteurs peptidiques perméables aux cellules de la voie de transduction du signal jnk pour le traitement de diverses maladies

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: 15734295

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2016575150

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 15321904

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2015734295

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015734295

Country of ref document: EP