WO2021010621A1 - 자가면역질환 및 염증성질환 펩타이드 치료제 - Google Patents
자가면역질환 및 염증성질환 펩타이드 치료제 Download PDFInfo
- Publication number
- WO2021010621A1 WO2021010621A1 PCT/KR2020/008536 KR2020008536W WO2021010621A1 WO 2021010621 A1 WO2021010621 A1 WO 2021010621A1 KR 2020008536 W KR2020008536 W KR 2020008536W WO 2021010621 A1 WO2021010621 A1 WO 2021010621A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- peptide
- mip2
- tlr
- tlr4
- tir
- Prior art date
Links
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K7/00—Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
- C07K7/04—Linear peptides containing only normal peptide links
- C07K7/08—Linear peptides containing only normal peptide links having 12 to 20 amino acids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/62—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
- A61K47/64—Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
- C07K14/4701—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
- C07K14/4713—Autoimmune diseases, e.g. Insulin-dependent diabetes mellitus, multiple sclerosis, rheumathoid arthritis, systemic lupus erythematosus; Autoantigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/01—Fusion polypeptide containing a localisation/targetting motif
- C07K2319/10—Fusion polypeptide containing a localisation/targetting motif containing a tag for extracellular membrane crossing, e.g. TAT or VP22
Definitions
- the present invention relates to a peptide that inhibits the Toll-like receptor (TLR) signaling pathway, and more particularly, a peptide that strongly binds to a TIR-containing molecule to inhibit the TLR, in particular, the TLR4 signaling pathway.
- TLR Toll-like receptor
- It relates to a composition for the prevention or treatment of an autoimmune disease or inflammatory disease comprising a fusion peptide in which a cell-penetrating peptide is bound to the peptide, a TLR antagonist including the peptide or fusion peptide, and the peptide or fusion peptide.
- TLR Toll-like receptor
- DAMPs danger-associated molecular patterns
- PAMPs pathogen-associated molecular patterns
- TIR-containing adapter molecules including TRIF (TICAM-1), MAL (TIRAP), MyD88 and TRAM (TICAM-2) and activation of NF- ⁇ B, interferon regulators (IRFs) and other transcription factors.
- TLR signaling through MyD88 can be MAL-dependent, or can bind directly to the TLR-TIR domain to induce a downstream signaling cascade.
- TLR3 regulates signal transmission by MyD88 and TRAM itself through TRIF.
- the TLR aggregates on a lipid raft located in the plasma membrane and initiates downstream signaling in a very sophisticated manner.
- the correct arrangement of the TLR-TIR and adapter molecules is critical to understanding how the TLR transmits signals.
- many empirical models Choen, CJ et al., Nat Med 13, 851-856 (2007)
- in silico models Guven-Maiorov, E. et al., Sci Rep 5) , 13128 (2015)
- MAL TIR domain adapter protein
- TIRAP Myeloid differentiation primary response 88
- the N-terminus containing the positively charged motif immobilizes the MAL on the plasma membrane through the PIP2 molecule (Patra, MC & Choi, S., Front Immunol 9, 75 (2018)).
- MAL-TIR may exist in the form of a back to back symmetric dimer and a CD loop promoted by ⁇ C and ⁇ D helix.
- cryo-EM cryo-electron microscopy
- MAL-TIR forms spontaneous and reversible filaments promoted by BB-loops in vitro.
- MAL forms intra-chain interactions by BB-loop in a head-to-tail manner, and the ⁇ B and ⁇ C helix and ⁇ D helix of one MAL-TIR and the CD loop of the other MAL-TIR.
- Inter-chain (interstrand) including-to form an interaction.
- the interchain-interaction is maintained in a triangular manner. That is, one subunit interacts with two other subunits in the protofilament.
- the present inventors assumed that the C-terminus of ⁇ C is as important as half of the N-terminus, and due to the stable helical structure, the peptide containing amino acids P155-T166 not only other MyD88-dependent TLR signaling but also MAL-TIR assembly It was assumed to be sufficient to destroy.
- the present inventors designed a peptide derived from the ⁇ C helix of MAL (defined as'MIP2'), and the peptide combined with a cell permeable peptide It was confirmed that it successfully inhibited TLR signaling, alleviated the symptoms of psoriasis in mice, alleviated the symptoms observed in the systemic lupus erythematosus mouse model, and confirmed that the arthritis mouse model exhibited a therapeutic effect, and the present invention was completed. .
- An object of the present invention is to provide a peptide that inhibits the TLR signaling pathway and a fusion peptide in which a cell-penetrating peptide is bound to the peptide.
- Another object of the present invention is to provide a TLR antagonist comprising the peptide or the fusion peptide.
- Another object of the present invention is to provide a composition for preventing or treating autoimmune diseases or inflammatory diseases comprising the peptide or the fusion peptide.
- Another object of the present invention is a method for preventing or treating autoimmune diseases or inflammatory diseases comprising administering the peptide or the fusion peptide, the peptide or the fusion for the prevention or treatment of autoimmune diseases or inflammatory diseases It is to provide the use of the peptide and the use of the peptide or the fusion peptide for the manufacture of a drug for the prevention or treatment of autoimmune diseases or inflammatory diseases.
- the present invention provides a peptide represented by the amino acid sequence of SEQ ID NO: 1.
- the present invention also provides a fusion peptide in which a cell-penetrating peptide is bound to the peptide.
- the present invention also provides a TLR antagonist comprising the peptide or the fusion peptide.
- the present invention also provides a composition for preventing or treating autoimmune diseases comprising the peptide or the fusion peptide.
- the present invention also provides a composition for preventing or treating inflammatory diseases comprising the peptide or the fusion peptide.
- the present invention also provides a method for preventing or treating autoimmune diseases or inflammatory diseases comprising administering the peptide or the fusion peptide.
- the present invention also provides the use of the peptide or the fusion peptide for the prevention or treatment of autoimmune diseases or inflammatory diseases.
- the present invention also provides the use of the peptide or the fusion peptide for the manufacture of a drug for preventing or treating autoimmune diseases or inflammatory diseases.
- Figure 1A shows the ⁇ C of MAL that promotes TIR-TIR interaction with MAL and other TIR-containing adapter proteins
- Figure 1B is to design MAL/MyD88 inhibitory peptide (MIP) and its derivatives using a computer. It shows the ⁇ C helix of the used MAL.
- 1C is a graph showing the results of measuring the TLR-inhibiting effect of MIP1 and MIP2 by measuring SEAP signals in HEK-BlueTM hTLR4 cells
- FIGS. 1D to 1E are IL- measured through ELISA in RAW264.7 cells. 6 and a graph showing the secretion level of TNF- ⁇ .
- 1F is a graph showing the broad range of TLR-inhibiting effects of MIP2 evaluated by measuring TNF- ⁇ levels in RAW264.7 cells.
- Figure 2 shows the toxicity of each peptide designated as MIP1, 2, 3 was evaluated in HEK-Blue TM hTLR4, RAW264.7, THP-1 cells,
- FIG. 2A shows that MIP1 and MIP2 peptides do not show toxicity in HEK-Blue TM hTLR4 cells
- Figs. 2B and 2C show that MIP2 does not show toxicity in RAW264.7 and THP-1 cells.
- Figure 2D shows that MIP2-1, MIP2-2 and MIP2-3 inhibit or do not inhibit the secretion of LPS-induced TNF- ⁇ cytokine and NO, respectively
- Figure 2E shows MIP2 without CPP, CPP alone and Hydrophobic CPP, PFVYLI, shows a response to LPS-stimulated RAW264.7 cells.
- 2F and 2G show the comparison of the weight and size of lymph nodes and spleens of systemic lupus erythematosus mice.
- FIG. 3 shows MIP2 inhibiting MyD88- and TRIF-dependent TLR4 signaling in mouse macrophages
- 3A to 3C show p-p65, p65, I ⁇ -B ⁇ , ATF3, p-IRF3, p-ERK, ERK, p-JNK, JNK, in RAW264.7 cells measured by Western blot analysis of total-protein extraction. It shows the protein expression levels of p-p38 and p38, and ⁇ -actin was used as a loading control.
- 3D shows phosphorylation of NF- ⁇ B (p-p65) measured by immunofluorescence staining and confocal microscopy, where Hoechst was used for nuclear staining (scale bar indicates 20 ⁇ m).
- FIG. 3E shows the expression levels of iNOS and COX2 completely inhibited by MIP2 as measured by Western blot, ⁇ -actin was used as a loading control.
- Figure 3F is a graph showing the level of NO secretion measured with a standard NO secretion kit
- Figures 3G and 3H are quantified by DAF-FM and DCF-DA staining, respectively, and the intracellular NO and substantially down-regulated by MIP2
- This is a graph showing the generation of ROS. Data shown are from at least 3 independent experiments (n ⁇ 3), with bars mean mean ⁇ SEM (*P ⁇ 0.05, **P ⁇ 0.01).
- Figure 4A shows the expression of multiple transcription factors including p-p65, I ⁇ -B ⁇ , p-ERK, ERK, p-JNK, JNK, p-p38 and p38 as measured by Western blot analysis, ⁇ -actin It was used as a loading control, and the phosphorylated state of the factors was substantially suppressed by MIP2.
- Figure 4B is a graph showing that when evaluated through ELISA, the secretion levels of TNF- ⁇ and IL-6 are inhibited concentration-dependently by MIP2.
- Figure 4C shows the inhibitory effect of MIP2 on NLPR3 measured in ATP-treated (10 mM) THP-1 cells after LPS-primed (100 ng/ml for 4h), Western blot NLRP3 and pro- The expression level of IL1 ⁇ was measured, and cytokine secretion of IL1 ⁇ was evaluated using ELISA.
- Figure 4D shows the expression levels of ATF3, p-IRF3, p-IRF7, pIRAK4 and pro-IL1 ⁇ measured by Western blot analysis in LPS-stimulated cells, ⁇ -actin was used as a loading control. Data shown are from at least 3 independent experiments (n ⁇ 3), with bars mean mean ⁇ SEM (*P ⁇ 0.05, **P ⁇ 0.01).
- FIG. 5A shows psoriasis induced by topical application of imiquimod (IMQ) in C57BL/6 male mice.
- IMQ imiquimod
- Three concentrations of MIP2 were administered intraperitoneally before IMQ application, and Methotrexate (MTX) evaluated the relative treatment of MIP2. It was used as a positive control for, and a photo of the back skin was taken on the 4th day of treatment, showing that MIP2 showed a therapeutic effect compared to the group treated with normal, untreated and MTX.
- Figure 5B is a graph showing the disease severity score based on the clinical Psoriasis Area and Severity Index (PASI), three purple lines represent the PASI scores of mice treated with MIP2 of 1, 10 and 20 nmol / g, respectively.
- PASI clinical Psoriasis Area and Severity Index
- FIG. 5C and 5D are graphs showing the effect of MIP2 on the spleen weight and body weight kinetics of mice during treatment regimen
- FIG. 5E shows the effect of MIP2 on the thickness of the epidermis (yellow arrow) and the dermis (green arrow).
- the skin thickness of each group was measured with a Leica DMi8 fluorescence microscope (scale bar 250 ⁇ m).
- Figure 5F shows the results of immunohistochemical analysis of the dorsal skin lesions of each group.
- the intensity of brown staining implies CD68 (macrophage marker) and CD4 and IL17 (TH17 cell marker) in the lesion, and the scale bar used is 75 ⁇ m for high magnification images.
- Data represent mean ⁇ SEM obtained from 5 skin tissue samples of each group (###P ⁇ 0.001 between NC and PBS, **P ⁇ 0.01, *P ⁇ 0.05 between PBS and MIP2 at 1, 10, or 20 nmol or MTX, according to two-tailed Student's t-test).
- FIG. 6A shows the results of experimental verification of the inhibitory effect of MIP2 in the SLE mouse model, and a significant reduction in hair loss was observed in SLE-prone MIP2-treated mice.
- 6B is a graph showing that the body weight did not change in MIP-treated mice
- FIGS. 6C to 6E are graphs showing that lymphadenitis and splennomegaly were significantly improved in MIP2-treated SLE-prone mice.
- Figure 6F is the albumin content of urine
- Figure 6G to Figure 6J is a result of evaluation of serum ANA, anti-dsDNA antibody, IL-6 and C3 complement by ELISA.
- 6K is a result of histological examination of the kidney through H&E and PAS staining (scale bar 100 ⁇ m), in which a glomerular mass having proliferative mesangial cells and diffused meniscus (black broken circles) in untreated mice In MIP2-treated mice, fibrocellular crescent (blue circle) was reformed and the glomerulus returned to normal.
- Figure 7 shows the therapeutic effect of MIP2 in the rheumatoid arthritis (RA) rat model
- Figure 7A shows the results of foot monitoring of normal, MIP2-treated (20 pmol/g of rat's weight) and untreated RA rats at 1-6 weeks (scale bar 10 mm).
- 7B is an ankle diameter and joint index score of rats measured at 1-6 weeks, and an accurate Wilcoxon Rank Sum test (the same numerically as the Mann-Whitney U test) was performed to compare the average value between the two groups (* P ⁇ 0.05 and **P ⁇ 0.01).
- FIG. 8A shows the liver histological examination through Oil red O stain (scale bar 50 ⁇ m). Significant macrovesicular fat changes were induced by the methionine-choline-deficient (MCD) diet, and MIP2 did not show a significant improvement in steatosis in a prophylactic and therapeutic manner (MIP2 Prev and MIP2, respectively. MIP2 Ther).
- Figure 8B shows the liver cholesterol and triglyceride levels of the 4 mice group, it was confirmed that the MCD diet-induced liver fat change was improved in both the MIP2 Prev and MIP2 Ther groups. Serum AST and ALT levels were not significantly improved in MIP2 (FIG. 8C ), and gene transcription factors related to inflammation (FIG. 8D) and fibrosis (FIG. 8E) were significantly improved with MIP2 administration. The levels of dysregulated Ppar ⁇ and Ppar transcription factor were recovered by administration of MIP2 (Fig. 8F).
- TLRs Toll-like receptors
- SLE systemic lupus erythematosus
- RA rheumatoid arthritis
- the stoichiometry and accurate alignment of the adapter TIR-TIR interactions are essential for TLR activation and the underlying signaling-cascade.
- the plasma membrane-fixed MAL has the potential for BB-loop-mediated self-oligomerization, and interacts with other TIR-domain-containing adapters through ⁇ C and ⁇ D helices. do.
- MAL can connect MyD88 and TLR9.
- Mutation studies have confirmed that the ⁇ C helix of MAL is important not only for MAL-MAL, but also for MAL-MyD88 and MAL-TLR4 interactions. Mutations of the C-terminal residues of the ⁇ C helix (Q163A and L165A) have been reported to inhibit NF- ⁇ B induction by 60% and 20%, respectively.
- the interchain-interface of MAL-protofilament has been proposed to be similar to the MAL-MyD88 and MAL-TLR4/2 TIR-TIR interfaces.
- peptides derived from the ⁇ D helix of TLR4 blocked recruitment of the adapter to TLR4.
- the present inventors believe that the peptide composed of the entire ⁇ C helix (including aa P155-T166) can maintain the helix structure and is sufficient to inhibit not only MAL-TIR assembly but also other MyD88-dependent TLR signaling regardless of the entire TIR domain. Expected.
- MIP2 MAL/MyD88 inhibitory peptide 2
- MAL- ⁇ C interface knowledge drug molecular structure (pharmacophore) model MAL- ⁇ C interface knowledge drug molecular structure (pharmacophore) model.
- MIP2 blocked MyD88 and TRIF-dependent LPS-induced TLR4 signaling in murine and human cell lines.
- MIP2 has shown therapeutic potential in psoriasis, SLE and RA disease models. Characteristic serological and histological biomarkers were significantly recovered, and disease symptoms were substantially improved.
- these in vitro and in vivo studies suggest that MIP2 has a broad specificity of TLR and is effective in controlling autoimmune diseases and/or inflammatory diseases caused by microorganisms, environmental or genetic factors.
- the present inventors investigated the interference of ligand-induced TLR signaling by conjugating a peptide containing the ⁇ C helix of MAL with a reported cell permeable peptide.
- CPP was an efficient carrier of the peptide
- PFV-conjugated peptide did not show a significant effect (Fig. 1C).
- the present inventors thought that it was due to the low cell permeability of the PFV-conjugated peptide, or because CPP had an additional TLR-inhibiting effect. According to previous studies independent of TLR and further investigation by the present inventors, there was no TLR-inhibiting effect when CPP was used alone.
- Lactam bridges and disulfide bonds have been widely used to limit structural freedom and to improve receptor selectivity and binding affinity of therapeutic peptides (Harrison, RS et al., Proc Natl Acad Sci USA 107, 11686-11691 (2010). )). Therefore, the present inventors applied disulfide structural restrictions to MIP2 (or MIP3). An unexpected cytotoxic effect was exhibited by MIP3, but MIP2 did not exhibit cytotoxicity at the same concentration (FIG. 2A). To confirm that the C-terminal residue, in particular L165, is important for the TLR-inhibiting effect of MIP2, a derivative was made by residue substitution. MIP2-1 (L165H and other substitutions) lost its potential to inhibit LPS-stimulated TLR4 signaling. However, since the effect was maintained in MIP2-2, the loss of the effect was not due to C157A (Fig. 2D, E).
- MIP2 not only showed a wide range of TLR-inhibiting effects, but also inhibited MyD88 and TRIF dependent pathways of TLR4 in LPS-stimulated RAW264.7 cells and THP1-derived macrophages (Figs. 3 and 4). This can be explained in part by the fact that MIP2 is composed of the ⁇ C helical structure of MAL, which is likely to bind to MAL, TLR4-TIR and MyD88-TIR (Fig. 1A). In addition, MAL and TRAM compete to combine with the overlapping interface of TLR4-TIR, in both cases MIP2 is involved.
- MIP2 Peptides partially overlapping with the C-terminus of MIP2 can interact with MyD88, excluding TLR4 and MyD88 and TRAM physical interactions. It was found that MIP2 possesses a wide range of TIR-domain binding capacity, and thus a wide range of TLR-inhibiting potential. NLRP3 induction and mature IL-1 ⁇ release are associated with TLR-stimulation. Both MyD88- and TRIF-dependent TLR pathways can sensitize and activate the NLRP3 inflammasome. The present inventors confirmed that MIP2 inhibits the expression of NLRP3 and pro-IL-1 ⁇ in ATP-activated THP-1 cells sensitized with LPS (Fig. 4C).
- MIP2 substantially alleviated symptoms of IMQ-induced psoriasis in C57BL/6 mice and limited the associated biomarkers (CD68, CD4 and IL17). MIP2 reduced the characteristics of SLE-related serum markers such as IL6, anti-dsDNA, ANA and urine albumin, and alleviated SLE-related lymphadenitis and splennomegaly. In addition, the treatment for 5-6 weeks completely cured the arthritis symptoms of rats and restored related parameters.
- the present invention relates to a peptide represented by the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 3.
- the peptide inhibits the signaling pathway of any one or more TLR (Toll-like receptor) selected from the group consisting of TLR4, TLR1/2, TLR2/6, TLR3, TLR7/8, TLR7 and TLR9. It can be characterized. Preferably, it may be characterized by inhibiting the TLR4 or TLR7 signaling pathway.
- TLR Toll-like receptor
- TLR ectodomain leads to rearrangement of the transmembrane and juxtamembrane regions, which promotes homotypic and heterotypic association of the TIR domain. As a result, it leads to recruitment of TIR-containing adapter molecules through cooperative interface sharing (Monie, TP, et al., Immunol Rev 227, 161-175 (2009)).
- the complexity and ambiguous stoichiometry of the TIR-TIR interaction is the reason for the lack of a generally accepted interface. It is generally accepted that the TIR-TIR interaction is an early occurrence event essential for TLR signaling.
- the peptide of the present invention has a wide specificity of TLR inhibition, and will be effective in controlling autoimmune complications caused by microorganisms or environmental factors. I can.
- the peptide may be characterized in that it is derived from the ⁇ C helix of MAL (TIR domain adapter protein; TIRAP).
- MAL TIR domain adapter protein
- the peptide may be characterized by binding to a TIR (Toll/interleukin-1 receptor) domain or a TIR-containing adapter (adaptor) molecule.
- TIR Toll/interleukin-1 receptor
- adaptor adapter
- the TIR domain is a domain of TLR, in particular, TLR4 signaling, and the TIR-containing adapter molecule is MAL (TIR domain adapter protein; TIRAP), MyD88 (Myeloid differentiation primary response 88), TRIF (TIR domain-containing adapter-inducing interferon).
- TIRAP TIR domain adapter protein
- MyD88 Myeloid differentiation primary response 88
- TRIF TIR domain-containing adapter-inducing interferon
- ⁇ TICAM-1
- TRAM TICAM-2
- peptide refers to a linear molecule formed by bonding of amino acid residues to each other by a peptide bond.
- the peptide may be prepared according to a chemical synthesis method known in the art, preferably a solid phase synthesis technique It may be manufactured according to, but is not limited thereto.
- TLR4 refers to a protein belonging to TLRs, a family of transmembrane proteins that functions as a monitor for pathogen infection, and refers to a protein encoded by the TLR4 gene, and CD 284 (cluster of differentiation 284). It is also named. Since TLR4 recognizes various pathogen-associated molecular patterns (PAMPs) including LPS of Gram-negative bacteria, it is very important for activation of the innate immune system.
- PAMPs pathogen-associated molecular patterns
- TLR4 signaling pathway refers to a signaling pathway through TLR4.
- TLR4 is signaled by several adapter proteins, and the signaling pathway works as TIRAP (also referred to as MAL) and MyD88, as well as TRAM and TRIF.
- TIRAP also referred to as MAL
- MyD88 MyD88-independent signaling
- the TLR4 mediated signaling pathway induces activation of MyD88-dependent and MyD88-independent signaling through a variety of sub-signaling molecules.
- Initiation of the MyD88-dependent pathway induces early stage activation of NF- ⁇ B and secretion of proinflammatory cytokines such as TNF- ⁇ and IL-6, and initiation of the MyD88-independent pathway leads to the activation of IRF3 and 7, and IFNs.
- TLR4 induces activation of MAPK including ERK, JNK and p38, and secretes inflammatory cytokines and IFN (Achek, A et al., Arch. Pharmacal Res . 39:1032-1049, 2016; Kawai, T et al., Nat. Immunol . 11: 373-384, 2010).
- Stimulation of TLR4 by related ligands in macrophages induces COX2 and iNOS, induces NO production, and produces mitochondria and intracellular ROS (Kim, JY et al., Eur. J. Pharmacol . 584:175- 184, 2008; Liu, K.-L. et al. J. Agric. Food Chem . 54:3472-3478, 2006; West, AP et al. Nature 472:476, 2011).
- the TLR4 signaling pathway may be characterized in that it is induced by LPS (lipopolysaccharide).
- the term “inhibition” refers to a phenomenon in which biological activity or activity decreases due to deficiency, incongruity, and many other causes, and partially or completely blocks, reduces, prevents, or activates the activity of TLR. It may be delaying, inactivating or down-regulating.
- inhibition of IL-6 and TNF- ⁇ secretion by blocking the TLR4 signaling pathway of the peptide Inhibition of NF- ⁇ B activity; Inhibition of NLRP3 and pro-IL-1 ⁇ expression; Inhibition of phosphorylation of IRF3, IRF7 and expression of ATF3; inhibition of iNOS and COX2 expression;
- the release of NO and the production of intracellular ROS may be inhibited, and the peptide may be characterized by inhibiting both MyD88-dependent and TRIF-dependent TLR4 signaling pathways.
- the present invention relates to a fusion peptide in which a cell penetrating peptide is bound to the peptide.
- the fusion peptide may be characterized by inhibiting the TLR4 or TLR7 signaling pathway, and the TLR4 signaling pathway may be characterized by being induced by LPS.
- IL-6 TNF- ⁇ secretion inhibition by blocking the TLR4 signaling pathway of the fusion peptide; Inhibition of NF- ⁇ B activity; Inhibition of NLRP3 and pro-IL-1 ⁇ expression; Inhibition of phosphorylation of IRF3, IRF7 and expression of ATF3; inhibition of iNOS and COX2 expression;
- the release of NO and the production of intracellular ROS may be inhibited, and the peptide may be characterized by inhibiting both MyD88-dependent and TRIF-dependent TLR4 signaling pathways.
- cell penetrating peptide is a kind of signal peptide, which is a combination of specific amino acid sequences used for the purpose of delivering polymer substances such as proteins, DNA, RNA, etc. into cells. It is a phosphorus peptide. Until now, it has been used for intracellular delivery of various small molecule compounds, proteins, peptides, RNA, DNA, and other high molecular materials.
- the fusion peptide of the present invention uses a cell-permeable peptide, and the cell-permeable peptide is not particularly limited as long as it has a characteristic of entering the cell by a mechanism of endocytosis, but preferably, the cell permeability listed in Table 1 below. It can be used by selecting from a peptide or a variant thereof. More preferably, the cell-penetrating peptide may be characterized in that it is represented by the amino acid sequence of SEQ ID NO: 6.
- Transportan includes those used in the form of the following variants: AGYLLGKINLKALAALAKKIL-NH2 (TP10, PepFect 3), AGYLLGKINLKALAALAKKIL-NH2 (TP10, PepFect 6), AGYLLGKLLOOLAAAALOOLL-NH2 (TP10, PepFect 6).
- PepFect 14 AGYLLGKTNLKALAALAKKIL-NH2 (NickFect 1), AGYLLGKTNLKALAALAKKIL-NH2 (NickFect 2) and AGYLLGKTNLKALAALAKKIL-NH2 (Nickfect 3).
- an experiment was performed by selecting the Penetratin sequence (RQIKIWFQNRRMKWKK; SEQ ID NO: 6) and PFVYLI (SEQ ID NO: 7) among the cell permeable peptides of Table 1, and other cell-permeable peptides in addition to the actually used cell-permeable peptide Even when fused with the peptide of the present invention, it will be apparent to those skilled in the art that an effect similar to that of the present invention appears.
- it may be characterized by fusion of a cell-penetrating peptide comprising the amino acid sequence represented by SEQ ID NO: 6.
- the fusion peptide may be characterized in that a cell-penetrating peptide is bound to the N-terminus or C-terminus of the peptide.
- a cell-penetrating peptide is bound to the N-terminus of the peptide, but is not limited thereto.
- the fusion peptide to which a cell-penetrating peptide is linked to the N-terminus of the peptide may preferably be characterized in that it consists of the amino acid sequence of SEQ ID NO: 8.
- variants of the amino acid sequence may also be included within the scope of the present invention, and specifically, the variant is 70% or more, preferably 80% or more, more preferably 90% or more, and even more preferably as SEQ ID NO: 8 May include all peptides having sequence homology of 95% or more, even more preferably 98% or more, and most preferably 99% or more.
- the term “homology” refers to a degree of similarity between a wild type amino acid sequence and a wild type nucleic acid sequence.
- the present invention relates to a TLR antagonist comprising the peptide or the fusion peptide.
- the TLR may be any one or more selected from the group consisting of TLR4, TLR1/2, TLR2/6, TLR3, TLR7/8, TLR7 and TLR9.
- the term “antagonist” refers to a molecule that partially or completely inhibits the influence of other molecules such as receptors or intracellular mediators by any mechanism.
- TLR antagonist refers to a substance capable of directly or indirectly or substantially interfering, reducing or inhibiting the biological activity of TLR, and preferably, a peptide reactive with TLR binds directly to the TLR or TIR domain. And, by blocking the TLR signaling pathway by neutralizing the activity of TLR, it is possible to reduce the secretion of inflammatory cytokines, NO and ROS by inducing a decrease in the activation of NF- ⁇ B and MAPKs.
- the peptide represented by SEQ ID NO: 1 of the present invention inhibits the TLR4 signaling pathway induced by LPS, thereby secreting IL-6, NO and ROS, and activating NF- ⁇ B and MAPKs. It is excellent in the effect of inhibiting the TLR4 signal transduction pathway and can be usefully used as a composition for preventing or treating autoimmune diseases and inflammatory diseases.
- TLR-inhibiting molecules targeting ligand-binding domains.
- PPIs protein-protein interfaces
- the insolubility of small molecules to the TIR-domain may be due in part to the lack of sufficient structural information, flat protein-protein interfaces, and poorly defined pockets.
- Peptide-drugs are easy to digest with enzymes and have low bioavailability (Newland, A. et al., British journal of haematology 135, 547-553 (2006)), but offer lower toxicity and improved target selectivity compared to small molecules.
- the present invention relates to a composition for preventing or treating autoimmune diseases comprising the peptide or the fusion peptide.
- autoimmune disease is caused by a process in which a problem occurs in inducing or maintaining self-tolerance, and an immune response to the self-antigen occurs, thereby attacking the own tissue.
- the self-tolerance refers to immunologic unresponsiveness that does not adversely react to antigenic substances constituting self.
- the autoimmune diseases of the present invention include psoriasis, rheumatoid arthritis, psoriatic arthritis, experimental autoimmune arthritis, asthma, Crohn's disease, multiple sclerosis, experimental autoimmune encephalomyelitis, myasthenia gravis, thyroiditis, experimental form of uveitis, Hashimoto's thyroiditis, primary myxedema, Thyroid poisoning, pernicious anemia, autoimmune atrophy gastritis, Addison's disease, early menopause, male infertility, childhood diabetes, Goodpasture syndrome, common pemphigus, pemphigus, sympathetic ophthalmitis, lens uveitis, autoimmune hemolytic anemia, idiopathic leukocytosis , Primary cholangiosclerosis, chronic active hepatitis, latent cirrhosis, ulcerative colitis, Sjogren's syndrome, scleroderma, Wegener's granulomatosis, polymyositis,
- composition for preventing or treating autoimmune diseases may contain a pharmaceutically effective amount of the peptide or fusion peptide alone, or may contain one or more pharmaceutically acceptable carriers, excipients, or diluents.
- the pharmaceutically effective amount refers to an amount sufficient to prevent, improve, and treat symptoms of autoimmune diseases.
- pharmaceutically acceptable means that it is physiologically acceptable and does not cause allergic reactions such as gastrointestinal disorders, dizziness or similar reactions when administered to humans.
- the carrier, excipient and diluent include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, gum acacia, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, Polyvinylpyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate, and mineral oils.
- fillers, anti-aggregating agents, lubricants, wetting agents, flavoring agents, emulsifying agents and preservatives may additionally be included.
- composition of the present invention may include one or more known active ingredients having an autoimmune disease treatment effect together with the peptide or fusion peptide.
- compositions of the present invention may be formulated using methods known in the art to provide rapid, sustained or delayed release of the active ingredient after administration to a mammal other than human.
- the formulation may be in the form of a powder, granule, tablet, emulsion, syrup, aerosol, soft or hard gelatin capsule, sterile injectable solution, or sterile powder.
- the pharmaceutical composition of the present invention may be administered through various routes including oral, transdermal, subcutaneous, intravenous, or intramuscular, and the dosage of the active ingredient may include various factors such as the route of administration, the patient's age, sex, weight, and the patient's severity. It may be appropriately selected according to the present invention, and the composition for preventing or treating autoimmune diseases according to the present invention may be administered in parallel with a known compound having an effect of preventing, improving or treating symptoms of autoimmune diseases.
- the present invention relates to a composition for preventing or treating inflammatory diseases comprising the peptide or the fusion peptide.
- the term "inflammatory disease” refers to TNF- ⁇ , IL-1, IL-6, and prostaglandins secreted by immune cells such as macrophages by excessively promoting the immune system due to harmful stimulation such as inflammation inducing factors or irradiation.
- prostaglandin refers to a disease caused by an inflammatory substance (inflammatory cytokine).
- Inflammatory diseases of the present invention include insulin-dependent diabetes, eczema, allergies, atopic dermatitis, acne, atopic rhinitis, pulmonary inflammation, allergic dermatitis, chronic sinusitis, contact dermatitis, seborrheic dermatitis, Gastritis, gout, gout arthritis, ulcers, chronic bronchitis, ulcerative colitis, ankylosing spondylitis, sepsis, vasculitis, bursitis, temporal arteritis, solid cancer, Alzheimer's disease, arteriosclerosis, obesity, viral infection and non-alcoholic fat It is characterized by being selected from the group consisting of hepatitis (nonalcoholic steatohepatitis), but is not limited thereto.
- composition for preventing or treating inflammatory diseases includes a pharmaceutical preparation containing the above-described peptide as an active ingredient, descriptions of the contents overlapping with the composition of the present invention will be omitted.
- the present invention relates to a method for preventing or treating autoimmune diseases comprising administering the peptide or the fusion peptide.
- the present invention relates to the use of the peptide or the fusion peptide for the prevention or treatment of autoimmune diseases.
- the present invention relates to the use of the peptide or the fusion peptide for the manufacture of a drug for preventing or treating autoimmune diseases.
- the present invention relates to a method for preventing or treating inflammatory diseases comprising administering the peptide or the fusion peptide.
- the present invention relates to the use of the peptide or the fusion peptide for preventing or treating inflammatory diseases.
- the present invention relates to the use of the peptide or the fusion peptide for the manufacture of a medicament for preventing or treating inflammatory diseases.
- the prevention or treatment method, use, and use of the present invention uses the above-described “the peptide or the fusion peptide or a composition comprising the same”, and therefore, overlapping descriptions are omitted.
- Example 1-1 Peptide synthesis, reagent and cell line optimization
- MIP1, MIP2, and MIP3 were measured by Shimadzu LCMS-2020, and were 2182.6Da, 3710.5Da and 3680.5Da, respectively.
- LPS Esscherichia coli 0111: B4
- ATP adenosine triphosphate
- PAM3CSK4 TLR1/2
- Poly I:C
- Imiquimod IMQ; R837, TLR7
- R848 TLR7/8
- CpG-ODN CpG-ODN
- FSL-1 TLR2/6) was purchased from InvivoGen (San Diego, CA, USA).
- fetal bovine serum FBS
- normocin InvivoGen, San Diego, CA, USA
- DMEM high sugar Dulbecco's modified Eagle's medium
- HEK-BlueTM hTLR4 cells InvivoGen, San Diego, CA, USA
- RAW264.7 cells were cultured.
- THP-1 cells were cultured in RPMI 1640 medium supplemented with 1% penicillin/streptomycin solution and 10% FBS, and 80 nM phorbol 12-myristate 13-acetate (PMA; Sigma-Aldrich Co., St. Louis, MO, USA).
- PMA phorbol 12-myristate 13-acetate
- Example 1-2 Cell viability analysis
- HEK-BlueTM hTLR4 cells were dispensed at a density of 5 ⁇ 10 4 /well, and RAW264.7 and THP-1 cells were dispensed at 2 ⁇ 105/well. All cells were cultured overnight in 96-well plates (BD Biosciences, San Jose, CA, USA).
- Cell viability was determined by colorimetric 1-(4,5-dimethylthiazol-2-yl)-3,5-diphenylformazan(MTT) assay (Sigma-Aldrich) and/or MTS(3-(4,5-dimethylthiazol-2-yl) Measured using -5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (Promega Madison, WI, USA), which was performed as previously described (Kwon, HK et al. ., Toxicological sciences: an official journal of the Society of Toxicology 148, 204-219 (2015)).
- HEK-BlueTM hTLR4 cells were aliquoted at a density of 2 ⁇ 10 5 /well and cultured overnight in 24-well plates (BD Biosciences). Cells were treated with different concentrations of MIPs in the presence and/or absence of LPS (100 ng/ml). A portion (200 ⁇ l) of the supernatant from the treated cells was transferred to a micro-centrifuge tube, and heated at 65° C. for 10 minutes using a heating block (FINEPCR Co., Seoul, Korea). Thereafter, the supernatant was transferred to new 96-well plates (BD Biosciences), and SEAP production was detected using a HEK-BlueTM detection kit (InvivoGen, San Diego, CA, USA). Absorbance was measured at 620 nm using a microplate reader spectrophotometer system (Molecular Devices Inc., Silicon Valley, CA, USA).
- THP-1-derived macrophages and RAW264.7 cells were aliquoted into 96-well plates (BD Biosciences) at a density of 2 ⁇ 10 5 /well and grown overnight. After 24 hours of treatment, IL-6 secretion was evaluated using Mouse IL-6 ELISA MAXTM Deluxe (BioLegend, San Diego, CA, USA), and the Mouse TNF alpha ELISA Ready-SET-Go!® kit (eBioscience, San) Diego, CA, USA) was used to evaluate TNF- ⁇ production. After 24 hours of MIP2 treatment in THP-1 cells, the secretion levels of IL-6 and TNF- ⁇ were evaluated using Human IL-6 and TNF alpha ELISA MAXTM Deluxe (BioLegend, San Diego, CA, USA) kits, respectively. . The plates were then analyzed at each wavelength using a microplate spectrophotometer system (Molecular Devices).
- Example 1-5 Protein quantification and western blot analysis
- Total-protein extraction was performed using M-PER mammalian protein extraction reagent (Thermo Fisher Scientific, Inc.). The concentration of protein was measured with a bicinchoninic acid (BCA) assay kit (Sigma-Aldrich). Western blot analysis including gel electrophoresis and development was performed using a Mini-PROTEAN Tetra Cell and mini trans-blot electrophoretic transfer cell system (Bio-Rad Laboratories, Hercules, CA, USA) (Kwon, HK, et al. , Sci Rep 5, 15623 (2015)).
- BCA bicinchoninic acid
- the membranes are p-p65, p-JNK, JNK, p-IRF3, p-IRF7, p-ERK, ERK, p-p38, p38, I ⁇ -B ⁇ , IL1 ⁇ , NLPR3 and p-IRAK4 (Cell Signaling Technology Inc., Danvers. , MA, USA); p-ERK, p-p38, JNK, ATF3, COX2 and ⁇ -actin (Santa Cruz Biotechnology Inc., Dallas, TX, USA); And immunoblotting by gently shaking overnight at a temperature of 4° C. with a specific primary antibody (1:500-1000) against iNOS (BD Biosciences).
- the membrane was thoroughly washed with PBST (phosphate buffered saline with Tween 20), and incubated with peroxidase-binding anti-mouse or anti-rabbit IgG antibody (1:1000) for 2 hours. Proteins were detected with SuperSignal West Pico ECL solution (Thermo Fisher Scientific, Inc.), and the detected proteins were visualized with ChemiDocTM Touch Imaging System (Bio-Rad Laboratories).
- PBST phosphate buffered saline with Tween 20
- Example 1-6 Intracellular NO, ROS evaluation and NO secretion analysis
- RAW264.7 cells were aliquoted into a 6cm culture dish (SPL Life Sciences, Pochun, Korea) at 1 ⁇ 10 6 and cultured overnight. After LPS stimulation and MIP2 treatment, intracellular NO (Nitric Oxide) and ROS (reactive oxygen species) were quantified using DAF-FM and DCF-DA dyes (Thermo Fisher Scientific, Inc.) (Kwon, HK, et al. ., Sci Rep 5, 15623 (2015)). The fluorescence intensity of the cells was analyzed using a FACSAria III instrument with Diva software (BD Biosciences).
- RAW264.7 cells were dispensed into 96-well plates (BD Biosciences) at a density of 2 ⁇ 10 5 /well, and cultured overnight. NO secretion was measured with the Nitric Oxide Detection Kit (iNtRON Biotechnology Inc., Seongnam, Korea) (Kwon, HK et al., Toxicological sciences: an official journal of the Society of Toxicology 148, 204-219 (2015)). The absorbance was measured at 550 nm using a microplate spectrophotometer system (Molecular Devices).
- RAW264.7 cells were dispensed on 24-well plates (BD Biosciences) at a density of 2 ⁇ 10 5 /well, and cells were treated with MIP2 (50 ⁇ M) for 1 hour before LPS stimulation (100 ng/ml). Cells were fixed with 3.7% formaldehyde solution (Sigma-Aldrich) and immersed in 0.2% Triton X-100 solution (AMRESCO, Solon, OH, USA) for 15 minutes. The cells were then washed with PBS and blocked using 2% BSA solution (Thermo Fisher Scientific, Inc.). Thereafter, these cells were incubated with an anti-p-p65 antibody (1:1000; Santa Cruz Biotechnology Inc.) for 2 hours, and washed thoroughly with PBS.
- the cells were incubated with Alexa Fluor 546 secondary antibody (Invitrogen, Carlsbad, CA, USA) for 1 hour and washed 3 times with PBS. Nuclei were stained using Hoechst 33258 solution (5 ⁇ M; Sigma-Aldrich). Fluorescence intensity was measured with a confocal microscope (LSM-700; Carl Zeiss Microscopy GmbH, Kunststoff, Germany), and images were analyzed using Zen 2009 software.
- Alexa Fluor 546 secondary antibody Invitrogen, Carlsbad, CA, USA
- Example 2-1 Psoriasis mouse model
- mice 6-7 weeks old C57BL/6 mice were purchased from Orient Bio Inc. (Seongnam, South Korea). Mice were stored under specific pathogen-free conditions and provided with a standard experimental diet (STD) of free intake. All animal experiments were approved by Ajou University's Animal Experiments and Use Committee (IACUC) (approval number 2017-0002). 62.5mg Aldara cream (with 5% IMQ; Aldara, 3M Pharmaceutical LLC.) was applied for 5 consecutive days, resulting in symptoms similar to psoriasis. IMQ was not applied to the normal group, only PBS was injected. MIP2 (1, 10 or 20 nmol/g) or PBS (control) was administered one day before the Aldara cream (5%) was applied to the mice.
- IACUC Ajou University's Animal Experiments and Use Committee
- mice were euthanized by respiratory anesthesia, and skin lesions and spleen samples were collected for histological examination.
- an objective scoring system was developed based on clinical PASI. Factors such as erythema, scaling, wrinkles, and thickening were independently scored from 0 to 4: 0, none; 1, slightly; 2, the middle; 3, considerable; 4, very remarkable. Erythema, scaling and thickness were scored under the close supervision of experienced researchers. The cumulative score (erythema+scaling+thickening, scale 0-12) indicates the severity of psoriasis symptoms.
- the skin sample obtained from the dorsal lesion of the mouse was fixed in a 4% paraformaldehyde solution, embedded in paraffin, and then a 7 ⁇ m thick section was made on a glass slide. Sections were stained with H&E to check the thickness of the epidermis and dermis. The skin thickness was measured using a Leica DMi8 fluorescence microscope using a Leica LAS X Hardware Configurator. IMQ-mediated inflammation in the skin was evaluated using a mouse specific HRC/DAB Detection IHC Kit (Abcam, cat. # AB64259) using primary antibodies that recognize CD68, CD4 and IL-17.
- Example 2-2 SLE mouse model
- mice Six female wild type (C57BL/6) and lupus-prone mice (MRL/lpr) with initial weights of 18-20 and 38-40 g, respectively, were purchased from Jackson Laboratory (Bar Harbor, ME, USA). All animal experiments were reviewed and approved by Ajou University Medical Center IACUC (approval number 2017-0022). Mice were acclimated for 1 week and then reared under pathogen-free conditions according to the approved guidelines of Ajou University School of Medicine. Mice in the vehicle group were injected with PBS intraperitoneally (ip), while others were injected with 10 nmol/g (in PBS) of MIP2 for 20 days (4 weeks, 5 times a week), and their weights were monitored daily. .
- mice were euthanized at the end of the experiment, and blood, urine, and tissue were collected. After 1 hour incubation, the blood sample was collected in a serum separation tube, centrifuged at 20°C for 10 minutes at 3000 rpm, and then stored at -80°C. The collected urine samples were immediately stored at -80°C, and the tissue samples were thoroughly washed with PBS and immersed in an RNA stabilizing solution (Qiagen Sciences, Maryland, MD, USA). The concentrations of anti-nuclear antibody, anti-dsDNA antibody, IL-6 and C3 complement were analyzed with an ELISA kit (MyBiosource, San Diego, CA).
- mice urine albumin levels were analyzed using the Mouse Albumin ELISA Kit (41-ALBMS-E01, Alpco Diagnostics, Salem, NH, USA). Kidney tissue was collected, then fixed in 4% paraformaldehyde (PFA) and refrigerated at 4° C. overnight. Then, the fixed tissue was inserted into paraffin, sectioned to a thickness of 2-4 ⁇ m, and stained with PAS (Periodic Acid-Schiff) or H&E reagent. Three independent measurements were performed per sample.
- PFA paraformaldehyde
- Example 2-3 CIA (collagen-induced arthritis; collagen-induced arthritis) model
- RA was induced by injecting a mixed solution of 250 ⁇ L (including 0.5mg collagen) (bovine type II collagen and complete Freund's adjuvant in a 1:1 ratio) to 4-week-old male Lewis rats.
- 250 ⁇ L including 0.5mg collagen
- Bovine type II collagen and complete Freund's adjuvant in a 1:1 ratio Three weeks after disease development, symptoms of edema and erythema were observed individually in the parietal bone, ankle joint, knee joint and foot of the rat.
- Rats were divided into 3 groups (each group contained 3 animals): normal (non-CIA, PBS only), untreated CIA and MIP2-treated CIA.
- CIA rats of the treatment group were individually injected with 100 ⁇ l MIP2 at a concentration of 20 pmol/g (5.94 ⁇ g/joint) to the knee joint.
- ankle diameter and AI (arthritis index) score the hind paws of each group individual rat were observed at weeks 1, 2, 3, 4, 5 and 6 of treatment regimen.
- Ankle diameter was measured using Vernier calipers, and AI was blind tested and then scored as follows: 0, no swelling or erythema; +1, slight edema and erythema; +2, moderate edema and shackles; +3, limited use of the joint for the metatarsal and swelling with enlarged symptoms; +4, excessive swelling due to severe symptoms associated with joint stiffness and total hind paws.
- Ankle diameter and AI score were independently measured three times, and the results were expressed as mean ⁇ standard deviation (SD).
- Example 2-4 NASH (Nonalcoholic steatohepatitis; nonalcoholic steatohepatitis) mouse model
- mice 24 9-week-old male wild-type (C57BL/6J) mice (initial weight 21-28 g) were purchased from Japan SLC, Inc. (Shizuoka, Japan). From 10 weeks of age, mice were separated into 4 groups of 6 mice. The normal chow diet control group, the MCD group of the MCD diet, the MCD diet and the MIP2 prevention group of MIP2 were treated from the beginning, and the MCD diet and the MIP2 treated group of MIP2 were treated from the 3rd week. Mice in the control group or MCD group were injected with PBS intraperitoneally (i.p.), others were administered MIP2 at 10 nmol/g (in PBS) 7 times a week for 6 weeks, and body weight was monitored daily.
- PBS intraperitoneally i.p.
- mice were euthanized, and blood and liver tissue were collected. After 1 hour incubation, blood samples were collected in a serum separation tube, centrifuged at 3000 rpm, 20° C. for 10 minutes, and stored at -80° C. Liver lysate was separated into RIPA buffer (Biosesang, R2002, KOREA) according to the manufacturer's instructions. Triglyceride of liver lysate was measured using a Triglyceride quantification colorimetric kit (Biovision K622-100, Inc.). Cholesterol of liver lysate was measured using Total Cholesterol and Cholesteryl Ester colorimetric kit (Biovision K603-100, Inc.).
- qRT-PCR analysis was performed using an ABI Prism 7500 qRT-PCR system (Applied Biosystems, Foster City, CA). Gene expression was detected with SYBR Premix Ex Taq (Takara RR420A, Inc.), and normalized to the reference gene GAPDH by the delta-delta Ct method (2- ⁇ Ct), and relative gene expression was measured.
- Example 3 Peptide design and in silico (computer programming in virtual experiment) analysis
- Dodecapeptide (MAL/MyD88 inhibitory peptide (MIP)) was designed from ⁇ C helix, and structural stability and cell permeability were investigated.
- MIP MAL/MyD88 inhibitory peptide
- PFVYLI a reported cell penetrating peptide (hereinafter referred to as'PFV' (Rhee, M. & Davis, P., J Biol Chem 281, 1233-1240 (2006)). )) and classified as MIP1, or conjugated with penetratin (CPP (Derossi, D., et al., J Biol Chem 269, 10444-10450 (1994)) and classified as MIP2.
- CPP conjugated with penetratin
- MIP2-1 L165H, C157A and M161A mutations
- MIP2-2 the C157A mutation
- MIP2-3 deficient in the MLQALT residue at the C-terminus was also evaluated, demonstrating that the effect was lost, demonstrating the importance of the C-terminus (Fig. 2D).
- MIP2 without CPP, CPP alone and hydrophobic CPP, PFVYLI did not show any inhibitory effect on LPS-stimulated RAW 264.7 cells (FIG. 2E ).
- MIP1 and MIP2 The modulatory effect of MIP1 and MIP2 on the MAL-mediated pathway was further evaluated. Although not MIP1, MIP2 showed concentration-dependent inhibition of LPS-stimulated secreted alkaline phosphatase (SEAP) signal in HEK-BlueTM hTLR4 cells (FIG. 1A ). This suggests that the hydrophobic PFV-carrier peptide exhibits low cell permeability when conjugated with MIP, or has an additional TLR4-inhibiting effect when CPP is bound to MIP. Furthermore, the present inventors observed that the PFV-conjugated peptide binds to an extracellular target.
- SEAP LPS-stimulated secreted alkaline phosphatase
- Example 4 Extensive TLR inhibition of MIP2 and confirmation of MyD88-dependent and TRIF-dependent TLR4 signaling inhibition in mouse macrophages
- TLR1/2 PAM3CSK4(TLR1/2), FSL-1(TLR2/6), Poly(I:C)(TLR3), Resiquimod(R848; TLR7/8), Imiquimod(IMQ) ;
- TLR7 TLR7
- TLR9 CpG-ODN
- TR6 significantly inhibited 3C (TLR1/2 ligand)-induced TNF- ⁇ and IL-1 ⁇ mRNA levels, but did not have a P2C (TLR2/6 ligand)-induced effect. Furthermore, this result suggests that MIP2 significantly inhibits endosome TLR. Western blot results, MIP2 substantially inhibited the phosphorylation of ERK and JNK (Fig. 3A). Unlike MIP2, TR6 has been reported to inhibit only ERK and not JNK in LPS-stimulated RAW264.7 cells.
- TLR7 and TLR9 require TIRAP. Nevertheless, in TIRAP-deficient cells and mice experiments, it responded normally to TLR3, TLR5, TLR7 and TLR9 ligands, and responses to TLR2 and TLR4 ligands have been demonstrated in many in vivo and in vitro studies to impair cytokine induction. (Yamamoto, M. et al., Nature 420, 324-329 (2002)). MIP2 strongly binds a wide range of TIR-containing molecules.
- TLR4 activates interferon only through the TRAM-TRIF pathway, whereas NF- ⁇ B can be activated through both MyD88 and TRIF (Yamamoto, M. et al., Science 301, 640-643 (2003)). It was found that MAL and TRAM later bind to promote TRIF or MyD88 interaction with TLR4. This is because MAL and TRAM share the TLR4 interface and compete for bonding. Therefore, the present inventors thought that MIP2 could inhibit both pathways of TLR4. MIP2 inhibited the phosphorylation of IRF3 and the expression of ATF3 in LPS-stimulated RAW264.7 cells (Fig. 3B).
- the TLR4-inhibiting effect of MIP2 was further confirmed through the expression and nuclear translocation levels of NF- ⁇ B. It was confirmed through Western blot data that MIP2 inhibits LPS-induced phosphorylation of NF- ⁇ B (p-p65) and stops I ⁇ -B ⁇ degradation (FIG. 3C). This result was further confirmed by confocal microscopy analysis, and MIP2 completely inhibited the phosphorylation of the p65 subunit of NF- ⁇ B and its transition to the nucleus (Fig. 3D).
- TLR stimulation is associated with the induction of nitric oxide (NO) and reactive oxygen species (ROS) and the expression of related enzymes such as nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX2) (West, AP et al., Nature). 472, 476-480 (2011)).
- MIP2 completely inhibits the expression of iNOS and COX2 (Fig. 3E), the extracellular and intracellular release of NO (Fig. 3F and 3G, respectively), and the production of intracellular ROS (Fig. 3H).
- MIP2 showed an inhibitory effect on MAPK in LPS-stimulated THP-1-derived macrophages.
- p38 expression was slightly inhibited, and this inhibitory effect was observed to be more pronounced in LPS-stimulated THP-1 cells (Fig. 4A).
- TLR upregulates the expression of NLRP3, caspase-1 and pro-IL-1 ⁇ (Bauernfeind, FG et al., J Immunol 183, 787-791 (2009)).
- a second signal such as ATP, Nigericin, Streptolysin O and Uric acid crystals
- LPS-primed cells have been reported to upregulate pro-IL-1 ⁇ but are not treated with IL-1 ⁇ and released.
- TLR uses both the MyD88- and TRIF- pathways. This was confirmed in the Western blot experiment of the present invention in which LPS-sensitized cells expressed NLRP3 upon ATP treatment.
- both NLRP3 and pro-IL-1 ⁇ expression were substantially inhibited in the presence of MIP2 (Fig. 4C).
- MIP2 inhibited not only the expression of IRF3 but also the expression of IRF7.
- expression of pro-IL-1 ⁇ and p-IRAK4 was suppressed in LPS-stimulated cells (Fig. 4D).
- MIP2 not only affects MyD88- and TRIF-dependent cytokine production, but also inhibits TLR-mediated NLRP3 inflammasome assembly.
- Psoriasis is a chronic inflammatory skin disease characterized by amplified proliferation and altered differentiation of epidermal cells as well as inflammatory cell infiltration into the skin.
- TLR is widely associated with the onset and progression of psoriasis (Hirai, T. et al., J Immunol 190, 4805-4811 (2013)).
- MIP2 possesses the potential of TLR-induced psoriasis treatment
- disease was induced by topical treatment of 62.5 mg of 5% IMQ-cream on the back skin in 6-7 week old C57BL/6 mice for 5 consecutive days (Fig. 5A).
- MIP2 was injected intraperitoneally (i.p.) for 6 days at a dose of 1, 10 or 20 nmol/g (mouse weight).
- MTX methotrexate
- PBS-injected mice was applied to mice.
- T cells or macrophages Skin hyperproliferative conditions and immune cell infiltration (T cells or macrophages) are the main features of psoriasis. Therefore, skin lesions of each animal group were collected and analyzed by immunohistochemical techniques using antibodies CD68 (macrophage marker), CD4 and IL17 (TH17 cell marker). MIP2 reduced overexpression of CD68, CD4 and IL-17 (brown staining) in psoriasis-mouse, which was similar to the MTX-treated group (Fig. 5F).
- MIP2 is expected to alleviate the induced psoriasis symptoms. This effect may be due to TLR7, a common target of imquimod (inducing psoriasis) and MIP2 (inhibiting the TLR7 pathway).
- TLR7 a common target of imquimod (inducing psoriasis)
- MIP2 inhibiting the TLR7 pathway.
- SLE systemic lupus erythematosus
- Example 7 Confirmation of the therapeutic effect of MIP2 in a SLE (systemic lupus erythematosus; systemic lupus erythematosus) mouse model
- MRL lymphoproliferation mice (so-called MRL/lpr, these mice spontaneously develop characteristic serological markers and peripheral pathology, typically lupus) at a dose of 10 nmol/g per mouse body weight for 20 days.
- MIP2 During the intraperitoneal injection of MIP2 (Fig. 6A). A significant reduction in hair loss was observed in SLE-prone MIP2-treated mice, while it continued to worsen in untreated mice (Fig. 6A). Damage to the heart, kidney, liver and spleen as well as a close relationship between weight gain and increased SLE markers (ANA, ROS, and spleen enlargement) have been reported (Toller-Kawahisa, JE et al., Free Radic Biol Med 86). , 362-373 (2015)). The present inventors confirmed that MIP2 prevents weight gain/reduction in mice fed a standard diet (FIG. 6B).
- Lymphadenopathy and splenomegaly are widely observed symptoms in SLE patients. Lymphadenitis and splennomegaly occur in about 50% and 10-40% of SLE patients, respectively (Bashal, F., Open Rheumatol J 7, 87-95 (2013)). It was confirmed that the size and weight of the lymph nodes significantly decreased in MIP2-treated SLE-prone mice (Figs. 6C and 2F). A similar pattern of reduction was recorded in the spleen size and weight of MIP2-treated mice ( Figures 6D, 6E and 2G).
- SLE related serological markers such as interleukin (IL), anti-dsDNA antibodies and antinuclear antibodies (ANA) and urine albumin levels (associated with glomerulonephritis) appear in SLE patient and animal models.
- relatively low complement components especially serum levels of C3 and C4, are common markers of SLE (Sandhu, V. & Quan, M., Open Rheumatol J 12, 171 (2018)).
- Albumin levels were significantly reduced in MIP2-treated MRL/lpr mice (Fig. 6F).
- a significant decrease in ANA and anti-dsDNA antibodies was observed compared to untreated mice (FIGS. 6G and 6H ).
- a significant decrease in serum-IL6 levels was recorded, and an elevation of serum-C3 was confirmed in MIP2-treated mice (FIGS. 6I and 6J).
- Example 8 Confirmation of the therapeutic effect of MIP2 for rheumatoid arthritis (RA)
- MIP2 human immunodeficiency protein
- rats were divided into three groups and each group was 1) normal/healthy rat; 2) untreated RA rats; 3) 3 rats of MIP2-treated RA rats were included. Rats of groups 2 and 3 were injected with MIP2 into the knee joint, and group 1 was injected with PBS. In this example, rats were evaluated phenotype for RA symptoms. No pathological examination has been performed, but the therapeutic effect of MIP2 can be reproduced in the future. The effect of MIP2 was evaluated by visually monitoring the rat's foot for 6 weeks at 1-week intervals, measuring the ankle diameter, and measuring the articular index (AI).
- AI articular index
- MIP2 has a wide range of therapeutic effects on immune-related diseases, suggesting that it may be a promising treatment targeting autoimmune diseases in the future.
- nonalcoholic steatohepatitis is associated with several organs such as liver, adipose tissue, gastrointestinal tract and brain, and the TLR pathway is one of the key pathways for the development of NASH.
- TLR7 or TLR4 knockout mice have been reported to be protected from non-alcoholic fatty liver disease (Rivera CA, et al., J Hepatol 2007, 47;571-579; Roh YS, et al. , The American Journal of Pathology . 188;2574-2588).
- MIP2 methionine-choline-deficient
- liver cholesterol and triglyceride levels significantly decreased when MIP2 was administered (FIG. 8B).
- serum alanine aminotransferase and aspartate aminotransferase in the MIP2 prophylactic group or treatment group (Fig. 8C)
- liver expression of inflammatory transcription factors TNF- ⁇ , Mcp1, IL-1 ⁇ and IL-6 by administration of MIP2 ( hepatic expression) decreased (Fig. 8D).
- hepatic expression of fibrotic transcription factors Col, Fn1 and Acta2 was reduced (Fig. 8E).
- Choline deficiency has reduced the Ppar ⁇ Ppar ⁇ increases the transcription factor, MIP2 had recovered both transcription factors in the induction MCD- NASH mouse model (Fig. 8F).
- MIP2 reduced liver fat content, inflammation and fibrosis-related gene expression, and restored the dysfunctional PPAR pathway in the NASH mouse model induced by the MCD diet.
- the effect of MIP2 on NASH showed more sufficient effect for prevention than treatment.
- the peptide according to the present invention exhibits an inhibitory effect on a wide range of TLR signaling pathways including TLR4, blocks MyD88- and TRIF-dependent TLR4 pathways, and in rheumatoid arthritis, psoriasis, systemic lupus erythematosus, and nonalcoholic steatohepatitis mouse models. Since it has a substantial disease alleviation effect, it can be usefully used as a therapeutic agent for immune-related diseases and inflammatory diseases requiring negative control of TLR.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Organic Chemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Immunology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Molecular Biology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Epidemiology (AREA)
- Rheumatology (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- Pain & Pain Management (AREA)
- Rehabilitation Therapy (AREA)
- Hematology (AREA)
- Diabetes (AREA)
- Toxicology (AREA)
- Zoology (AREA)
- Gastroenterology & Hepatology (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Peptides Or Proteins (AREA)
- Medicinal Preparation (AREA)
Abstract
Description
Claims (13)
- 서열번호 1 또는 서열번호 3의 아미노산 서열로 표시되는 펩타이드.
- 제1항에 있어서, 상기 펩타이드는 TLR4, TLR1/2, TLR2/6, TLR3, TLR7/8, TLR7 및 TLR9로 구성된 군에서 선택되는 어느 하나 이상의 TLR(Toll-like receptor)의 신호전달 경로를 억제하는 것을 특징으로 하는 펩타이드.
- 제1항에 있어서, 상기 펩타이드는 MAL(TIR domain adaptor protein; TIRAP)의 αC 나선으로부터 유래된 것을 특징으로 하는 펩타이드.
- 제1항에 있어서, 상기 펩타이드는 TIR(Toll/interleukin-1 receptor) 도메인 또는 TIR-함유 어댑터(adaptor) 분자에 결합하는 것을 특징으로 하는 펩타이드.
- 제1항의 펩타이드에 세포 투과성 펩타이드(cell penetrating peptide)가 결합된 융합 펩타이드.
- 제5항에 있어서, 상기 세포 투과성 펩타이드는 서열번호 6의 아미노산 서열로 표시되는 것을 특징으로 하는 융합 펩타이드.
- 제5항에 있어서, 상기 융합 펩타이드는 서열번호 8의 아미노산 서열로 표시되는 것을 특징으로 하는 융합 펩타이드.
- 제1항의 펩타이드 또는 제5항의 융합 펩타이드를 포함하는 TLR(Toll-like receptor) 길항제(antagonist).
- 제8항에 있어서, 상기 TLR은 TLR4, TLR1/2, TLR2/6, TLR3, TLR7/8, TLR7 및 TLR9로 구성된 군에서 선택되는 어느 하나 이상인 것을 특징으로 하는 TLR 길항제.
- 제1항의 펩타이드 또는 제5항의 융합 펩타이드를 포함하는 자가면역질환의 예방 또는 치료용 조성물.
- 제10항에 있어서, 상기 자가면역질환은 건선, 류마티스 관절염, 건선 관절염, 실험적 자가면역 관절염, 천식, 크론병, 다발성 경화증, 실험적 자가면역 뇌척수염, 중증 근무력증, 갑상선염, 실험적 형태의 포도막염, 하시모토 갑상선염, 원발성 점액수종, 갑상샘 중독증, 악성 빈혈, 자가면역 위축 위염, 애디슨 질환, 조기 폐경, 남성 불임증, 소아 당뇨병, 굿파스처 증후군, 보통 천포창, 유천포창, 교감성 안염, 수정체성 포도막염, 자가면역 용혈성 빈혈, 특발성 백혈구 감소, 원발성 담관 경화증, 만성 활동성 간염, 잠재성 간경변증, 궤양성 대장염, 쇼그렌 증후군, 경피증, 베게너 육아종증, 다발근육염, 피부근육염, 원판상루푸스 및 전신홍반루푸스로 이루어진 군에서 선택되는 것을 특징으로 하는 자가면역질환의 예방 또는 치료용 조성물.
- 제1항의 펩타이드 또는 제5항의 융합 펩타이드를 포함하는 염증성질환의 예방 또는 치료용 조성물.
- 제12항에 있어서, 상기 염증성질환은 인슐린-의존성 당뇨병, 습진, 알러지, 아토피성 피부염, 여드름, 아토피성 비염, 폐염증, 알레르기성 피부염, 만성 부비동염, 접촉성 피부염(contact dermatitis), 지루성 피부염(seborrheic dermatitis), 위염, 통풍, 통풍 관절염, 궤양, 만성 기관지염, 궤양성 대장염, 강직성 척추염(ankylosing spondylitis), 패혈증, 맥관염, 활액낭염, 측두 동맥염, 고형암, 알츠하이머병, 동맥경화증, 비만, 바이러스 감염 및 비알콜성 지방간염(nonalcoholic steatohepatitis)으로 이루어진 군에서 선택되는 것을 특징으로 하는 염증성질환의 예방 또는 치료용 조성물.
Priority Applications (6)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP20841421.9A EP4001295A4 (en) | 2019-07-16 | 2020-06-30 | PEPTIDE THERAPEUTIC AGENTS FOR AUTOIMMUNE AND INFLAMMATORY DISEASES |
CN202080064800.3A CN114502570A (zh) | 2019-07-16 | 2020-06-30 | 用于自身免疫疾病和炎性疾病的肽治疗剂 |
US17/627,018 US20220257782A1 (en) | 2019-07-16 | 2020-06-30 | Peptide therapeutics for autoimmune diseases and inflammatory diseases |
CA3147002A CA3147002C (en) | 2019-07-16 | 2020-06-30 | Peptide therapeutics for autoimmune diseases and inflammatory diseases |
AU2020313326A AU2020313326B2 (en) | 2019-07-16 | 2020-06-30 | Peptide therapeutics for autoimmune diseases and inflammatory diseases |
JP2022502549A JP2022540498A (ja) | 2019-07-16 | 2020-06-30 | 自己免疫疾患および炎症性疾患のためのペプチド治療薬 |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
KR1020190085927A KR102588548B1 (ko) | 2019-07-16 | 2019-07-16 | 자가면역질환 및 염증성질환 펩타이드 치료제 |
KR10-2019-0085927 | 2019-07-16 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2021010621A1 true WO2021010621A1 (ko) | 2021-01-21 |
Family
ID=74211078
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/KR2020/008536 WO2021010621A1 (ko) | 2019-07-16 | 2020-06-30 | 자가면역질환 및 염증성질환 펩타이드 치료제 |
Country Status (8)
Country | Link |
---|---|
US (1) | US20220257782A1 (ko) |
EP (1) | EP4001295A4 (ko) |
JP (1) | JP2022540498A (ko) |
KR (1) | KR102588548B1 (ko) |
CN (1) | CN114502570A (ko) |
AU (1) | AU2020313326B2 (ko) |
CA (1) | CA3147002C (ko) |
WO (1) | WO2021010621A1 (ko) |
Families Citing this family (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
KR102617426B1 (ko) | 2021-05-13 | 2023-12-27 | 차의과학대학교 산학협력단 | 수지상 세포의 TGF-β 수용체 활성화용 조성물 및 이를 이용한 자가면역질환의 예방 또는 치료 방법 |
CN114371135B (zh) * | 2021-10-25 | 2024-01-30 | 孙良丹 | 一种用于评价银屑病的评价系统及应用 |
KR102541402B1 (ko) * | 2022-02-18 | 2023-06-14 | 주식회사 레메디 | 아토피 피부염 예방 또는 치료 활성을 가지는 펩타이드 |
WO2024096677A1 (ko) * | 2022-11-03 | 2024-05-10 | 한국과학기술연구원 | Tlr 신호 억제 펩타이드 및 이를 포함하는 염증성 질환의 예방 또는 치료용 조성물 |
WO2024117293A1 (ko) * | 2022-11-29 | 2024-06-06 | 주식회사 젠센 | 자가면역질환 및 염증성 질환 펩타이드 치료제 |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20130203649A1 (en) * | 2012-02-07 | 2013-08-08 | University Of Maryland, Baltimore | Inhibitors of tlr signaling by targeting tir domain interfaces |
KR20160140031A (ko) * | 2015-05-29 | 2016-12-07 | 아주대학교산학협력단 | 신규한 tlr4 길항제 |
KR20180122940A (ko) * | 2017-05-04 | 2018-11-14 | 주식회사 젠센 | 톨-유사 수용체(tlr) 억제를 위한 펩타이드 및 이를 포함하는 약학적 조성물 |
Family Cites Families (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP3810731B2 (ja) * | 2002-11-29 | 2006-08-16 | 独立行政法人科学技術振興機構 | 哺乳動物のToll様受容体3に結合する新規アダプタータンパク質およびその遺伝子 |
WO2010151495A2 (en) * | 2009-06-26 | 2010-12-29 | University Of Florida Research Foundation Inc. | Materials and methods for treating and preventing viral infections |
AU2018399627B2 (en) * | 2018-01-02 | 2024-05-02 | Rush University Medical Center | Compositions and methods for treating neurological and other disorders |
-
2019
- 2019-07-16 KR KR1020190085927A patent/KR102588548B1/ko active IP Right Grant
-
2020
- 2020-06-30 AU AU2020313326A patent/AU2020313326B2/en active Active
- 2020-06-30 CA CA3147002A patent/CA3147002C/en active Active
- 2020-06-30 JP JP2022502549A patent/JP2022540498A/ja active Pending
- 2020-06-30 EP EP20841421.9A patent/EP4001295A4/en not_active Withdrawn
- 2020-06-30 CN CN202080064800.3A patent/CN114502570A/zh active Pending
- 2020-06-30 US US17/627,018 patent/US20220257782A1/en active Pending
- 2020-06-30 WO PCT/KR2020/008536 patent/WO2021010621A1/ko unknown
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20130203649A1 (en) * | 2012-02-07 | 2013-08-08 | University Of Maryland, Baltimore | Inhibitors of tlr signaling by targeting tir domain interfaces |
KR20160140031A (ko) * | 2015-05-29 | 2016-12-07 | 아주대학교산학협력단 | 신규한 tlr4 길항제 |
KR20180122940A (ko) * | 2017-05-04 | 2018-11-14 | 주식회사 젠센 | 톨-유사 수용체(tlr) 억제를 위한 펩타이드 및 이를 포함하는 약학적 조성물 |
Non-Patent Citations (39)
Title |
---|
ACHEK, A ET AL., ARCH. PHARMACOL RES., vol. 39, 2016, pages 1032 - 1049 |
AKIRA, S.UEMATSU, S.TAKEUCHI, O., CELL, vol. 124, 2006, pages 783 - 801 |
BASHAL, F., OPEN RHEUMATOL J, vol. 7, 2013, pages 87 - 95 |
BAUERNFEIND, F. G. ET AL., JLMMUNOL, vol. 183, 2009, pages 787 - 791 |
BOVIJN, C. ET AL., JBIOL CHEM, vol. 288, 2013, pages 12054 - 12066 |
CARCELLER, E. ET AL., SCI REP, vol. 6, 2016, pages 38825 |
CHEN, CJ ET AL., NAT MED, vol. 13, 2007, pages 851 - 856 |
COUTURE LEAH A., PIAO WENJI, RU LISA W., VOGEL STEFANIE N., TOSHCHAKOV VLADIMIR Y.: "Targeting Toll-like Receptor (TLR) Signaling by Toll/Interleukin-1 Receptor (TIR) Domain-containing Adapter Protein/MyD88 Adapter-like (TIRAP/Mal)-derived Decoy Peptides", JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY FOR BIOCHEMISTRY AND MOLECULAR BIOLOGY, vol. 287, no. 29, 1 July 2012 (2012-07-01), pages 24641 - 24648, XP055774156, ISSN: 0021-9258, DOI: 10.1074/jbc.M112.360925 * |
COUTURE, L. A. ET AL., J BIOL CHEM, vol. 287, 2012, pages 24641 - 24648 |
DEROSSI, D. ET AL., JBIOL CHEM, vol. 269, 1994, pages 10444 - 10450 |
FITZGERALD, K. A. ET AL., JEXPMED, vol. 198, 2003, pages 1043 - 1055 |
GAY, N. J. ET AL., NAT REV IMMUNOL, vol. 14, 2014, pages 546 - 558 |
GUVEN-MAIOROV, E. ET AL., SCI REP, vol. 5, 2015, pages 15623 |
HARRISON, R. S. ET AL., PROC NATL ACAD SCI USA, vol. 107, 2010, pages 11686 - 11691 |
HIRAI, T. ET AL., JLMMUNOL, vol. 190, 2013, pages 4805 - 4811 |
HONDA, K.TANIGUCHI, T., NAT REV IMMUNOL, vol. 6, 2006, pages 644 - 658 |
HOPKINS, A. L.GROOM, C. R., NAT REV DRUG DISCOV, vol. 1, 2002, pages 727 - 730 |
KAWAI,T ET AL., NAT. IMMUNOL., vol. 11, 2010, pages 373 - 384 |
KIM, J.Y ET AL., EUR. J. PHARMACOL., vol. 584, 2008, pages 175 - 184 |
KWON, H. K. ET AL., TOXICOLOGICAL SCIENCES: AN OFFICIAL JOURNAL OF THE SOCIETY OF TOXICOLOGY, vol. 148, 2015, pages 204 - 219 |
LIN, Z. ET AL., PLOS ONE, vol. 7, 2012, pages e34202 |
LIU, K.-L. ET AL., J. AGRIC. FOOD CHEM., vol. 54, 2006, pages 3472 - 3478 |
MONIE, T. P. ET AL., IMMUNOL REV, vol. 227, 2009, pages 161 - 175 |
NCBI Reference Sequence: AAL05628.1 TIR domain containing adaptor protein TIRAP[Mus musculus] (20 September 2001) definition; and fasta * |
NEWLAND, A. ET AL., BRITISH JOURNAL OF HAEMATOLOGY, vol. 135, 2006, pages 547 - 553 |
NIJLAND, R ET AL., MAR. DRUGS, vol. 12, 2014, pages 4260 - 4273 |
PARK, B.S ET AL., EXP. MOL. MED., vol. 45, 2013, pages e66 |
PIAO, W. ET AL., J IMMUNOL, vol. 190, 2013, pages 2263 - 2272 |
RHEE, M.DAVIS, P., J BIOL CHEM, vol. 281, 2006, pages 1233 - 1240 |
RIVERA CA ET AL., JHEPATOL, vol. 47, 2007, pages 571 - 579 |
ROH YS ET AL., THE AMERICAN JOURNAL OF PATHOLOGY, vol. 188, pages 2574 - 2588 |
SANDHU, V.QUAN, M., OPEN RHEUMATOL J, vol. 12, 2018, pages 171 |
SHAH MASAUD; KIM GI-YOUNG; ACHEK ASMA; CHO EUN-YOUNG; BAEK WOOK-YOUNG; CHOI YANG SEON; LEE WANG HEE; KIM DONG-JIN; LEE SANG HO; KI: "The αC helix of TIRAP holds therapeutic potential in TLR-mediated autoimmune diseases", BIOMATERIALS, ELSEVIER, AMSTERDAM, NL, vol. 245, 17 March 2020 (2020-03-17), AMSTERDAM, NL, XP086118035, ISSN: 0142-9612, DOI: 10.1016/j.biomaterials.2020.119974 * |
STEVENS, S. L. ET AL., JNEUROSCI, vol. 31, 2011, pages 8456 - 8463 |
TOLLER-KAWAHISA, J. E. ET AL., FREE RADIC BIOL MED, vol. 86, 2015, pages 362 - 373 |
WEST, A. P. ET AL., NATURE, vol. 472, 2011, pages 476 - 480 |
WU, Y. W.TANG, W.ZUO, J. P., ACTA PHARMACOL SIN, vol. 36, 2015, pages 1395 - 1407 |
YAMAMOTO, M. ET AL., NATURE, vol. 420, 2002, pages 324 - 329 |
YAMAMOTO, M. ET AL., SCIENCE, vol. 301, 2003, pages 640 - 643 |
Also Published As
Publication number | Publication date |
---|---|
CA3147002C (en) | 2023-09-26 |
KR102588548B1 (ko) | 2023-10-16 |
KR20210009197A (ko) | 2021-01-26 |
AU2020313326A1 (en) | 2022-02-03 |
US20220257782A1 (en) | 2022-08-18 |
AU2020313326B2 (en) | 2023-05-11 |
JP2022540498A (ja) | 2022-09-15 |
EP4001295A1 (en) | 2022-05-25 |
EP4001295A4 (en) | 2023-05-31 |
CN114502570A (zh) | 2022-05-13 |
CA3147002A1 (en) | 2021-01-21 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2021010621A1 (ko) | 자가면역질환 및 염증성질환 펩타이드 치료제 | |
DK2650008T3 (en) | Use of Cell Permeable Peptide Inhibitors by the JNK Signal Transduction Pathway for the Treatment of Various Cancer Diseases | |
WO2015156649A1 (ko) | 섬유증 억제 활성을 가지는 펩티드 및 이를 포함하는 조성물 | |
WO2013169077A1 (ko) | 악액질 예방 또는 치료용 조성물 | |
WO2021167324A1 (en) | Improved cell-permeable nuclear import inhibitor synthetic peptide for inhibition of cytokine storm or inflammatory disease and use thereof | |
KR20180122940A (ko) | 톨-유사 수용체(tlr) 억제를 위한 펩타이드 및 이를 포함하는 약학적 조성물 | |
WO2022220632A1 (ko) | 세포 투과 펩티드, 항암 펩티드 및 이를 포함하는 암 예방 또는 치료용 약학적 조성물 | |
ES2407454T3 (es) | Métodos de tratamiento de trastornos neuronales utilizando péptidos MNTF y análogos de los mismos | |
AU2006244080C1 (en) | Use of peptides derived from the growth factor AMP-18 for the treatment of mucositis | |
WO2021177518A1 (ko) | 혈중 콜레스테롤 저하용, 심혈관 대사질환의 예방 또는 치료용 및 항염용 약학적 조성물 | |
WO2020116810A1 (ko) | Fas 신호전달 억제용 펩티드를 포함하는 비만, 지방간 또는 지방간염의 예방 또는 치료용 약학적 조성물 | |
WO2018203613A1 (ko) | 톨-유사 수용체(tlr) 억제를 위한 펩타이드 및 이를 포함하는 약학적 조성물 | |
WO2020141704A1 (ko) | Tlr4 신호전달 경로를 억제하는 펩타이드 및 그 용도 | |
US8017576B2 (en) | Methods and compositions to treat mucositis | |
WO2022015106A1 (ko) | 종양괴사인자-자극된 유전자 6을 발현하는 중간엽 줄기세포를 포함하는 골관절염의 예방 또는 치료용 조성물 | |
WO2020046002A1 (ko) | 다중의 질환 바이오마커의 기능 및 발현을 억제하는 펩타이드의 신규한 용도 | |
WO2020091513A1 (ko) | Tlr 억제 펩타이드를 포함하는 루푸스 예방 또는 치료용 조성물 | |
WO2024112143A1 (ko) | Stat1-tmd의 면역 관련 질환 용도 | |
WO2024215162A1 (ko) | 단일 융합 단백질 및 이를 포함하는 약학적 조성물 | |
WO2012036410A2 (ko) | 변형된 인간 종양 괴사 인자 수용체-1 폴리펩티드 또는 그의 절편 및 그의 제조방법 | |
WO2022250470A1 (ko) | Asm 단백질에 특이적으로 결합하는 항체를 유효성분으로 포함하는 뇌질환 치료용 약학적 조성물 | |
WO2018048046A2 (ko) | mTOR 저해제를 함유하는 황반변성 치료용 의약조성물 | |
WO2021242046A1 (ko) | 신규한 항염증성 펩타이드 및 이의 용도 | |
WO2024167312A1 (ko) | 조절 t 세포에 특이적으로 존재하는 dkk-1 단백질 및 그 용도 | |
WO2024117293A1 (ko) | 자가면역질환 및 염증성 질환 펩타이드 치료제 |
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: 20841421 Country of ref document: EP Kind code of ref document: A1 |
|
ENP | Entry into the national phase |
Ref document number: 3147002 Country of ref document: CA |
|
ENP | Entry into the national phase |
Ref document number: 2022502549 Country of ref document: JP Kind code of ref document: A |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
ENP | Entry into the national phase |
Ref document number: 2020313326 Country of ref document: AU Date of ref document: 20200630 Kind code of ref document: A |
|
ENP | Entry into the national phase |
Ref document number: 2020841421 Country of ref document: EP Effective date: 20220216 |